Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Virol J ; 21(1): 158, 2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39004752

RESUMO

BACKGROUND: West Nile virus (WNV) is a rapidly spreading mosquito-borne virus accounted for neuroinvasive diseases. An insight into WNV-host factors interaction is necessary for development of therapeutic approaches against WNV infection. CD11b has key biological functions and been identified as a therapeutic target for several human diseases. The purpose of this study was to determine whether CD11b was implicated in WNV infection. METHODS: SH-SY5Y cells with and without MEK1/2 inhibitor U0126 or AKT inhibitor MK-2206 treatment were infected with WNV. CD11b mRNA levels were assessed by real-time PCR. WNV replication and expression of stress (ATF6 and CHOP), pro-inflammatory (TNF-α), and antiviral (IFN-α, IFN-ß, and IFN-γ) factors were evaluated in WNV-infected SH-SY5Y cells with CD11b siRNA transfection. Cell viability was determined by MTS assay. RESULTS: CD11b mRNA expression was remarkably up-regulated by WNV in a time-dependent manner. U0126 but not MK-2206 treatment reduced the CD11b induction by WNV. CD11b knockdown significantly decreased WNV replication and protected the infected cells. CD11b knockdown markedly increased TNF-α, IFN-α, IFN-ß, and IFN-γ mRNA expression induced by WNV. ATF6 mRNA expression was reduced upon CD11b knockdown following WNV infection. CONCLUSION: These results demonstrate that CD11b is involved in maintaining WNV replication and modulating inflammatory as well as antiviral immune response, highlighting the potential of CD11b as a target for therapeutics for WNV infection.


Assuntos
Antígeno CD11b , Replicação Viral , Vírus do Nilo Ocidental , Humanos , Replicação Viral/efeitos dos fármacos , Vírus do Nilo Ocidental/fisiologia , Vírus do Nilo Ocidental/imunologia , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Linhagem Celular Tumoral , Febre do Nilo Ocidental/imunologia , Febre do Nilo Ocidental/virologia , Neuroblastoma/imunologia , Neuroblastoma/virologia , Interações Hospedeiro-Patógeno/imunologia , Sobrevivência Celular/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/genética
2.
F1000Res ; 12: 719, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38903860

RESUMO

Background: Paediatric neuroblastoma and brain tumours account for a third of all childhood cancer-related mortality. High-risk neuroblastoma is highly aggressive and survival is poor despite intensive multi-modal therapies with significant toxicity. Novel therapies are desperately needed. The Zika virus (ZIKV) can access the nervous system and there is growing interest in employing ZIKV as a potential therapy against paediatric nervous system tumours, including neuroblastoma. Methods: Here, we perform extensive data mining, integration and re-analysis of ZIKV infection datasets to highlight molecular mechanisms that may govern the oncolytic response in neuroblastoma cells. We collate infection data of multiple neuroblastoma cell lines by different ZIKV strains from a body of published literature to inform the susceptibility of neuroblastoma to the ZIKV oncolytic response. Integrating published transcriptomics, interaction proteomics, dependency factor and compound datasets we propose the involvement of multiple host systems during ZIKV infection. Results: Through data mining of published literature, we observed most paediatric neuroblastoma cell lines to be highly susceptible to ZIKV infection and propose the PRVABC59 ZIKV strain to be the most promising candidate for neuroblastoma oncolytic virotherapy. ZIKV induces TNF signalling, lipid metabolism, the Unfolded Protein Response (UPR), and downregulates cell cycle and DNA replication processes. ZIKV infection is dependent on sterol regulatory element binding protein (SREBP)-regulated lipid metabolism and three protein complexes; V-ATPase, ER Membrane Protein Complex (EMC) and mammalian translocon. We propose ZIKV non-structural protein 4B (NS4B) as a likely mediator of ZIKVs interaction with IRE1-mediated UPR, lipid metabolism and mammalian translocon. Conclusions: Our work provides a significant understanding of ZIKV infection in neuroblastoma cells, which will facilitate the progression of ZIKV-based oncolytic virotherapy through pre-clinical research and clinical trials.


Assuntos
Neuroblastoma , Terapia Viral Oncolítica , Proteômica , Zika virus , Humanos , Neuroblastoma/terapia , Neuroblastoma/metabolismo , Neuroblastoma/virologia , Terapia Viral Oncolítica/métodos , Zika virus/fisiologia , Proteômica/métodos , Linhagem Celular Tumoral , Infecção por Zika virus/terapia , Infecção por Zika virus/virologia , Infecção por Zika virus/metabolismo , Transcriptoma
3.
Viruses ; 13(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34834918

RESUMO

INTRODUCTION: ZIKV is a highly neurotropic virus that can cause the death of infected neuroprogenitor cells through mitochondrial damage and intrinsic apoptotic signaling. In this context, the role of reactive oxygen species (ROS) in neuronal cell death caused by ZIKV still remains elusive. OBJECTIVE: We aimed at evaluating the role of these cellular components in the death of human undifferentiated neuroblastoma cell line infected with ZIKV. RESULTS: ZIKV infection resulted in the extensive death of SH-SY5Y cells with the upregulation of several genes involved in survival and apoptotic responses as well as the colocalization of mitochondrial staining with ZIKV Envelope (E) protein. Notably, levels of intracellular reactive oxygen species (ROS) were not altered during ZIKV infection in undifferentiated SH-SY5Y cells, and consistent with these results, the treatment of infected cells with the widely studied ROS scavenger N-acetylcysteine (NAC) did not prevent cell death in these cells. CONCLUSION: Altogether, our results suggest that excessive ROS production is not the main trigger of SH-SY5Y cells death in ZIKV infection.


Assuntos
Apoptose , Neuroblastoma/fisiopatologia , Espécies Reativas de Oxigênio/metabolismo , Infecção por Zika virus/fisiopatologia , Zika virus/fisiologia , Linhagem Celular Tumoral , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/virologia , Estresse Oxidativo , Zika virus/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia
4.
Viruses ; 13(11)2021 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-34834998

RESUMO

SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) is primarily responsible for coronavirus disease (COVID-19) and it is characterized by respiratory illness with fever and dyspnea. Severe vascular problems and several other manifestations, including neurological ones, have also been frequently reported, particularly in the great majority of "long hauler" patients. SARS-CoV-2 infects and replicates in lung epithelial cells, while dysfunction of endothelial and neuronal brain cells has been observed in the absence of productive infection. It has been shown that the Spike protein can interact with specific cellular receptors, supporting both viral entry and cellular dysfunction. It is thus clear that understanding how and when these receptors are regulated, as well as how much they are expressed would help in unveiling the multifaceted aspects of this disease. Here, we show that SH-SY5Y neuroblastoma cells express three important cellular surface molecules that interact with the Spike protein, namely ACE2, TMPRSS2, and NRP1. Their levels increase when cells are treated with retinoic acid (RA), a commonly used agent known to promote differentiation. This increase matched the higher levels of receptors observed on HUVEC (primary human umbilical vein endothelial cells). We also show by confocal imaging that replication-defective pseudoviruses carrying the SARS-CoV-2 Spike protein can infect differentiated and undifferentiated SH-SY5Y, and HUVEC cells, although with different efficiencies. Neuronal cells and endothelial cells are potential targets for SARS-CoV-2 infection and the interaction of the Spike viral protein with these cells may cause their dysregulation. Characterizing RNA and protein expression tempo, mode, and levels of different SARS-CoV-2 receptors on both cell subpopulations may have clinical relevance for the diagnosis and treatment of COVID-19-infected subjects, including long hauler patients with neurological manifestations.


Assuntos
COVID-19/metabolismo , Células Endoteliais/metabolismo , Neuroblastoma/metabolismo , Receptores Virais/metabolismo , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/virologia , Linhagem Celular Tumoral , Células Endoteliais/virologia , Interações entre Hospedeiro e Microrganismos , Células Endoteliais da Veia Umbilical Humana , Humanos , Neuroblastoma/virologia , Neuropilina-1/metabolismo , Serina Endopeptidases/metabolismo , Internalização do Vírus
5.
Sci Rep ; 11(1): 10109, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980968

RESUMO

Misfolding of the cellular prion protein, PrPC, into the amyloidogenic isoform, PrPSc, which forms infectious protein aggregates, the so-called prions, is a key pathogenic event in prion diseases. No pathogens other than prions have been identified to induce misfolding of PrPC into PrPSc and propagate infectious prions in infected cells. Here, we found that infection with a neurotropic influenza A virus strain (IAV/WSN) caused misfolding of PrPC into PrPSc and generated infectious prions in mouse neuroblastoma cells through a hit-and-run mechanism. The structural and biochemical characteristics of IAV/WSN-induced PrPSc were different from those of RML and 22L laboratory prions-evoked PrPSc, and the pathogenicity of IAV/WSN-induced prions were also different from that of RML and 22L prions, suggesting IAV/WSN-specific formation of PrPSc and infectious prions. Our current results may open a new avenue for the role of viral infection in misfolding of PrPC into PrPSc and formation of infectious prions.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Humana/metabolismo , Influenza Humana/virologia , Neuroblastoma/metabolismo , Neuroblastoma/virologia , Proteínas Priônicas/química , Linhagem Celular Tumoral , Humanos , Influenza Humana/genética , Neuroblastoma/genética , Proteínas Priônicas/metabolismo , Conformação Proteica , Dobramento de Proteína
6.
J Virol ; 95(13): e0000321, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-33853962

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is an important oncogenic virus previously shown to be neurotropic, but studies on neuronal cell infection and pathogenesis are still very limited. Here, we characterized the effects of KSHV infection on neuronal SH-SY5Y cells by the recombinant virus rKSHV.219, which expresses both green fluorescent protein (GFP) and red fluorescent protein (RFP) to reflect the latent and lytic phases of infection. We demonstrated that infected cells have a higher growth rate and that KSHV infection can be sustained. Interestingly, the infected cells can transition spontaneously back and forth between lytic and latent phases of infection, producing progeny viruses but without any adverse effects on cell growth. In addition, transcriptome analysis of viral and cellular genes in latent and lytic cells showed that unlike other infected cell lines, the latently infected cells expressed both latent and most, but not all, of the lytic genes required for infectious virion production. The viral genes uniquely expressed by the lytic cells were mainly involved in the early steps of virus binding. Some of the cellular genes that were deregulated in both latently and lytically infected cells are involved in cell adhesion, cell signal pathways, and tumorigenesis. The downregulated cellular CCDN1, PAX5, and NFASC and upregulated CTGF, BMP4, YAP1, LEF1, and HLA-DRB1 genes were found to be associated with cell adhesion molecules (CAMs), hippo signaling, and cancer. These deregulated genes may be involved in creating an environment that is unique in neuronal cells to sustain cell growth upon KSHV infection and not observed in other infected cell types. IMPORTANCE Our study has provided evidence that neuronal SH-SY5Y cells displayed unique cellular responses upon KSHV infection. Unlike other infected cells, this neuronal cell line displayed a higher growth rate upon infection and can spontaneously transition back and forth between latent and lytic phases of infection. Unlike other latently infected cells, a number of lytic genes were also expressed in the latent phase of infection in addition to the established latent viral genes. They may play a role in deregulating a number of host genes that are involved in cell signaling and tumorigenesis in order to sustain the infection and growth advantages for the cells. Our study has provided novel insights into KSHV infection of neuronal cells and a potential new model for further studies to explore the underlying mechanism in viral and host interactions for neuronal cells and the association of KSHV with neuronal diseases.


Assuntos
Regulação Viral da Expressão Gênica/genética , Herpesvirus Humano 8/metabolismo , Neurônios/metabolismo , Ativação Viral/fisiologia , Latência Viral/fisiologia , Animais , Adesão Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Chlorocebus aethiops , Células HEK293 , Infecções por Herpesviridae/patologia , Humanos , Infecção Latente/virologia , Neuroblastoma/metabolismo , Neuroblastoma/virologia , Neurônios/virologia , Células Vero , Replicação Viral/fisiologia
7.
Brain Res ; 1758: 147344, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33556379

RESUMO

Modelling cell infection in-a-dish can represent a useful tool to understand the susceptibility of different cell types towards severe acute respiratory coronavirus-2 (SARS-CoV-2) and to decipher its neurotropism. In this perspective, retinoic acid (RA)-differentiated neuroblastoma cell lines, SH-SY5Y and SK-N-BE(2) and glioblastoma cell lines, U-87 MG and U-373 MG, were infected with a SARS-CoV-2 strain, at various multiplicity-of-infection (MOI). We first demonstrated that the common entry genes - needed for invading epithelial cells - were expressed. RA-differentiation induced an upregulation of ace2 and tmprss2 gene expression while inducing downregulation of ctsb and ctsl. Using in situ hybridization and confocal analysis, SARS-CoV-2 gene S RNA was detected intracellularly at MOI 5.0, and localized in both soma and neuritic-like or glial-like processes. The infection was confirmed by quantification of viral gene E RNA and showed a dose-dependency, with few infected cells at MOI 0.1. After 24 h of infection, no cytopathic effect was observed in SH-SY5Y abilities to maintain neuritic processes or in U-373 MG for the uptake of glutamate. Unlike the permissive Vero E6 cells, no significant apoptosis death was detected following SARS-CoV-2 infection of neuroblastoma or glioblastoma cells. This study demonstrates the susceptibility of neuronal- and glial-like cell lines towards SARS-CoV-2 infection at high MOIs. Once inside the cells, the virus does not seem to rapidly replicate nor exert major cytopathic effect. Overall, our results strengthen the idea that SARS-CoV-2 has a tropism for nervous cells that express commonly described entry genes.


Assuntos
COVID-19/virologia , Glioblastoma/virologia , Neuroblastoma/virologia , SARS-CoV-2/patogenicidade , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/patologia , Linhagem Celular Tumoral , Citoplasma/metabolismo , Glioblastoma/patologia , Humanos , Modelos Biológicos , Neuroblastoma/patologia , SARS-CoV-2/metabolismo , Serina Endopeptidases/metabolismo
8.
Int J Mol Sci ; 22(2)2021 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-33430186

RESUMO

The ability to establish, maintain, and reactivate from latency in sensory neurons within trigeminal ganglia (TG) is crucial for bovine herpesvirus 1 (BoHV-1) transmission. In contrast to lytic infection, the only viral gene abundantly expressed during latency is the latency-related (LR) gene. The synthetic corticosteroid dexamethasone consistently induces reactivation from latency, in part because the glucocorticoid receptor (GR) transactivates viral promoters that drive expression of key viral transcriptional regulator proteins (bICP0 and bICP4). Within hours after dexamethasone treatment of latently infected calves, LR gene products and ß-catenin are not readily detected in TG neurons. Hence, we hypothesized that LR gene products and/or ß-catenin restrict GR-mediated transcriptional activation. A plasmid expressing LR RNA sequences that span open reading frame 2 (ORF2-Stop) inhibited GR-mediated transactivation of the BoHV-1 immediate early transcription unit 1 (IEtu1) and mouse mammary tumor virus (MMTV) promoter activity in mouse neuroblastoma cells (Neuro-2A). ORF2-Stop also reduced productive infection and GR steady-state protein levels in transfected Neuro-2A cells. Additional studies revealed that the constitutively active ß-catenin mutant reduced the transactivation of the IEtu1 promoter by GR and dexamethasone. Collectively, these studies suggest ORF2 RNA sequences and Wnt/ß-catenin signaling pathway actively promote maintenance of latency, in part, by impairing GR-mediated gene expression.


Assuntos
Infecções por Herpesviridae/genética , RNA não Traduzido/genética , Proteínas Virais/genética , beta Catenina/genética , Animais , Bovinos , Dexametasona/farmacologia , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/patogenicidade , Humanos , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/patogenicidade , Camundongos , Neuroblastoma/genética , Neuroblastoma/virologia , Regiões Promotoras Genéticas/genética , RNA não Traduzido/farmacologia , Receptores de Glucocorticoides/genética , Células Receptoras Sensoriais/metabolismo , Células Receptoras Sensoriais/virologia , Fatores de Transcrição/genética , Gânglio Trigeminal/metabolismo , Gânglio Trigeminal/virologia , Latência Viral/genética , Via de Sinalização Wnt/efeitos dos fármacos
9.
Virus Res ; 292: 198245, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33253716

RESUMO

Enterovirus A71 (EV-A71) emerged as a leading cause of virus derived infant encephalitis in most Asian countries. Some recent studies point out the critical role of microRNA (miRNA) in the regulation of pyroptosis. However, the role of miRNAs in the regulation of EV-A71 infection-induced pyroptosis was not previously explored. In this study, we utilized microRNA array and real-time PCR to verify that miR-195 significantly down-regulate in EV-A71-infected SH-SY5Y human neuroblastoma cells. An inverse correlation of NLRX1 with miR-195 expression in EV-A71-infected SH-SY5Y cells was found. Target prediction of miR-195 showed that NLRX1 could directly interact with miR-195. Results from luciferase reporter assays, qRT-PCR and western blotting demonstrated the negative regulation between miR-195 and NLRX1. Silencing NLRX1 expression with small interfering RNAs (siRNAs-NLRX1) and over-expression of miR-195 also attenuate the EV-A71 associated pyroptosis. Our findings provided evidence showed that miR-195 can regulate EV-A71 infection-induced pyroptosis, by directly targeting NLRX1.


Assuntos
Enterovirus Humano A/fisiologia , Infecções por Enterovirus/genética , Infecções por Enterovirus/virologia , MicroRNAs/metabolismo , Proteínas Mitocondriais/metabolismo , Neuroblastoma/genética , Piroptose , Linhagem Celular Tumoral , Regulação para Baixo , Enterovirus Humano A/genética , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/fisiopatologia , Regulação da Expressão Gênica , Humanos , MicroRNAs/genética , Proteínas Mitocondriais/genética , Neuroblastoma/metabolismo , Neuroblastoma/virologia
10.
Vet Microbiol ; 247: 108762, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32768214

RESUMO

Bovine herpesvirus 1 (BoHV-1) is an important cattle pathogen, that may cause rhinotracheitis, abortions and shipping fever. Virus establishes latency in sensory neurons, but periodically could reactivate. Recent studies identified mouse neuroblastoma (Neuro-2A) cells as a novel cell culture model to study factors that regulate BoHV-1 productive infection in neuronal cells. Herein, following BoHV-1 infection in Neuro-2A, a reduced cell viability occurred. Membrane damage and death morphological alterations, features of apoptosis and necrosis, were distinguished in infected cells. In addition, biochemical signs of apoptosis (caspase 3 activation and PARP cleavage) were observed. These results were accompanied by incomplete autophagy due to enhanced amounts of autophagic markers (LC3-II, ATG5 and Beclin 1), in the presence of increased levels of p62. Interestingly, protein expression of viral infected cell protein 0 (bICP0) was detected in Neuro-2A cells, although BoHV-1 inefficiently replicates in these cells, because just low levels of viral yield were found. Taken together, our results suggest that BoHV-1 may exert its potential neurotoxicity through a combined mechanism of necrosis and apoptosis. Moreover, incomplete autophagy occurred during BoHV-1 replication in Neuro-2A cells, which were favourable for viral persistence.


Assuntos
Sobrevivência Celular , Herpesvirus Bovino 1/patogenicidade , Interações entre Hospedeiro e Microrganismos , Neurônios/virologia , Animais , Apoptose , Autofagia , Bovinos , Linhagem Celular Tumoral , Membrana Celular/patologia , Camundongos , Necrose , Neuroblastoma/virologia , Neurônios/fisiologia , Latência Viral
11.
FASEB J ; 34(7): 9650-9663, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32469133

RESUMO

Previous studies have indicated that the amino acid at position 333 in the glycoprotein (G) is closely related to rabies virus (RABV) pathogenicity. However, whether there are other amino acid residues in G that relate to pathogenicity remain unclear. The aim of this study is to find new amino acid residues in G that could strongly reduce RABV pathogenicity. The present study found that the pathogenicity of a virulent strain was strongly attenuated when the amino acid glycine (Gly) replaced the aspartic acid (Asp) at position 255 in G (D255G) as intracranial (i.c.) infection with this D255G mutant virus did not cause death in adult mice. The indexes of neurotropism of the D255G mutant strain and the parent GD-SH-01 are 0.72 and 10.0, respectively, which indicate that the D255G mutation decreased the neurotropism of RABV. In addition, the D255G mutation significantly decreased RABV replication in the mouse brain. Furthermore, the D255G mutation enhanced the immune response in mice, which contributed to the clearance of RABV after infection. The Asp255 â†’ Gly255 mutation was genetically stable in vitro and in vivo. In this study, we describe a new referenced amino acid site in G that relates to the pathogenicity of RABV.


Assuntos
Aminoácidos/genética , Glicoproteínas/genética , Mutação , Neuroblastoma/patologia , Raiva/patologia , Proteínas Virais/genética , Virulência/genética , Substituição de Aminoácidos , Animais , Modelos Animais de Doenças , Camundongos , Neuroblastoma/virologia , Raiva/virologia , Vírus da Raiva/crescimento & desenvolvimento , Vírus da Raiva/isolamento & purificação
12.
Viruses ; 12(5)2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32380717

RESUMO

The alternative splicing of pre-mRNAs expands a single genetic blueprint to encode multiple, functionally diverse protein isoforms. Viruses have previously been shown to interact with, depend on, and alter host splicing machinery. The consequences, however, incited by viral infection on the global alternative slicing (AS) landscape are under-appreciated. Here, we investigated the transcriptional and alternative splicing profile of neuronal cells infected with a contemporary Puerto Rican Zika virus (ZIKVPR) isolate, an isolate of the prototypical Ugandan ZIKV (ZIKVMR), and dengue virus 2 (DENV2). Our analyses revealed that ZIKVPR induced significantly more differential changes in expressed genes compared to ZIKVMR or DENV2, despite all three viruses showing equivalent infectivity and viral RNA levels. Consistent with the transcriptional profile, ZIKVPR induced a higher number of alternative splicing events compared to ZIKVMR or DENV2, and gene ontology analyses highlighted alternative splicing changes in genes associated with mRNA splicing. In summary, we show that ZIKV affects cellular RNA homeostasis not only at the transcriptional levels but also through the alternative splicing of cellular transcripts. These findings could provide new molecular insights into the neuropathologies associated with this virus.


Assuntos
Processamento Alternativo , Neuroblastoma/virologia , Infecção por Zika virus/genética , Zika virus/fisiologia , Ásia , Linhagem Celular Tumoral , Humanos , Transcrição Gênica , Zika virus/genética , Infecção por Zika virus/metabolismo , Infecção por Zika virus/virologia
13.
BMC Vet Res ; 16(1): 95, 2020 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-32199460

RESUMO

BACKGROUND: Wild boar-derived hepatitis E (HEV) genotype 3 virus has been successfully isolated in cell lines of human origin only. Considering the zoonotic potential and possible extrahepatic localisation of genotype 3 strain, it is important to investigate the viability of cell lines of different animal and tissue origins. Therefore, the objective of the present study was to determine the permissiveness of non-human primate (MARC-145 and Vero) and swine (PK-15) cell lines of kidney origin, and a mouse neuroblastoma (Neuro-2a) cell line for isolation of wild boar-derived HEV genotype 3. RESULTS: This study showed that MARC-145, PK-15, Neuro-2a and Vero cell lines were permissive to wild boar-derived HEV genotype 3 subtype 3i harbouring viral genome equivalents of 1.12 × 107 copies/ml, 2.38 × 105 copies/ml, 2.97 × 107 copies/ml and 4.01 × 107 copies/ml after five serial passages respectively. In all permissive cell lines, HEV was continuously recovered from growth medium between five and at least 28 days post-infection. Peak loads of HEV genome equivalents were observed on days 7, 12, 19 and 30 in MARC-145 (2.88 × 107 copies/ml), Vero (4.23 × 106 copies/ml), Neuro-2a (3.15 × 106 copies/ml) and PK-15 (2.24 × 107 copies/ml) cell lines respectively. In addition, successful virus isolation was confirmed by immunofluorescence assay targeting HEV capsid protein and sequencing of HEV isolate retrieved from cell cultures. CONCLUSIONS: This study showed that wild boar-derived HEV genotype 3 subtype 3i strain was capable of infecting cell lines of animal origin, including primate and porcine kidney cells (MARC-145, PK-15 and Vero), and mouse neuroblastoma cells (Neuro-2a), supporting the notion of the capacity of HEV genotype 3 to cross the species barrier and extra-hepatic localisation of the virus. These findings warrant further studies of tested cell lines to investigate their capacity as an efficient system for HEV propagation. HEV isolates from other wild animal hosts should be isolated on tested cell lines in order to generate more data on HEV transmission between wild animal populations and their role as sources of human infections.


Assuntos
Vírus da Hepatite E/genética , Hepatite E/veterinária , Rim/citologia , Neuroblastoma/virologia , Animais , Linhagem Celular , Chlorocebus aethiops , Genótipo , Hepatite E/virologia , Camundongos , Filogenia , Sus scrofa , Suínos , Cultura de Vírus
14.
Virus Res ; 282: 197945, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32220619

RESUMO

Enterovirus A71 (EV71) remains the most common causative agent of hand, foot, and mouth disease (HFMD), and the neurological complications induced by EV71 are usually the leading cause of death in children with HFMD. However, the mechanism of nervous system changes caused by EV71 infection is still unclear. Therefore, in the current study, EV71 was inoculated into the human neuroblastoma cell line SH-SY5Y and subsequent transcriptome sequencing was used to examine the alterations of the transcriptome in infected SH-SY5Y cells. It is expected to determine the underlying mechanism of neurological diseases in response to EV71 infection. As a result, a total of 82,406,974, 112,410,808 and 87,780,371 clean reads were found in the control, EV71-12 h and EV71-24 h groups, respectively. Moreover, 160 and 745 differentially expressed genes were identified in the EV71-12 h and EV71-24 h groups, respectively, as compared to the control group. Next, to further explore the pathogenic mechanism triggered by EV71 infection, we mainly focused on the common differentially expressed genes at different time points of EV71 infection. And it was discovered that there were 95 common differentially expressed genes, which were used to conduct GO and pathway analysis. GO enrichment analysis demarcated related biological processes, molecular functions and cellular components, and KEGG pathway analysis enabled annotations of metabolic pathways and revealed interactions among the significantly enriched pathways. The results showed that the enriched GO term "Nervous system development" and enriched pathway "CCKR signaling map" might be important contributors to EV71-induced neuropathological mechanisms. In addition, we also screened 10 up- and down-regulated non-protein coding genes with significantly different expression in our transcriptome profiling, which suggested that these abnormally regulated non-protein-encoding genes might also play important roles in the pathogenesis of EV71 infection. Eventually, RT-qPCR technology was adopted to validate the transcriptome sequencing data and the experiment demonstrated that the RT-qPCR and transcriptome sequencing results were basically consistent. In summary, this is the first transcriptome analysis of SH-SY5Y cells in response to EV71 infection and provides valuable cues for further exploring the mechanism of nervous system changes caused by EV71 infection.


Assuntos
Enterovirus Humano A/genética , Enterovirus Humano A/patogenicidade , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Transcriptoma , Linhagem Celular Tumoral , Redes Reguladoras de Genes , Doença de Mão, Pé e Boca/virologia , Humanos , Sistema Nervoso/patologia , Neuroblastoma/virologia
15.
Int J Mol Sci ; 20(20)2019 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-31658698

RESUMO

Japanese encephalitis virus (JEV) infection induces uncontrolled neuronal apoptosis, leading to irreversible brain damage. However, the mechanism of JEV-induced neuronal apoptosis has not been clearly elucidated. This study aimed to investigate both virus replication and neuronal cell apoptosis during JEV infection in human neuroblastoma SH-SY5Y cells. As a result, the kinetic productions of new viral progeny were time- and dose-dependent. The stimulation of SH-SY5Y cell apoptosis was dependent on the multiplicity of infections (MOIs) and infection periods, particularly during the late period of infection. Interestingly, we observed that of full-length Bax (p21 Bax) level started to decrease, which corresponded to the increased level of its cleaved form (p18 Bax). The formation of p18 Bax resulting in cytochrome c release into the cytosol appeared to correlate with JEV-induced apoptotic cell death together with the activation of caspase-3/7 activity, especially during the late stage of a robust viral infection. Therefore, our results suggest another possible mechanism of JEV-induced apoptotic cell death via the induction of the proteolysis of endogenous p21 Bax to generate p18 Bax. This finding could be a new avenue to facilitate novel drug discovery for the further development of therapeutic treatments that could relieve neuronal damage from JEV infection.


Assuntos
Morte Celular/fisiologia , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/metabolismo , Neuroblastoma/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Sobrevivência Celular , Chlorocebus aethiops , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Encefalite Japonesa/virologia , Humanos , Cinética , Neuroblastoma/virologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Vero , Replicação Viral
16.
Microb Pathog ; 128: 106-111, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30579945

RESUMO

As one of the major causative agents of hand, foot and mouth disease (HFMD), enterovirus 71 (EV71) is a small, non-enveloped positive stranded RNA virus. Children suffering EV71 infection may cause severe symptoms including neurological complications, pulmonary edema and aseptic meningitis. EV71 is a neurotropic virus and it can cause the damage of nervous cells, cytokine storm and toxic substance. Identifying the factors that mediate viral binding or entry to host cells is important to uncover the mechanisms which viruses utilize to cause diseases in human body. Heat shock protein 70 (HSP70) is induced during virus infection and facilitates proper protein folding during viral propagation. The role that HSP70 plays during EV71 infection is still unclear. In this study, siRNA interference technique and transgenic technique were used to investigate the interaction between HSP70 and EV71 virus. The result demonstrated that the cell surface HSP70 is not essential for EV71 infection but helps the initial binding of virus to host cells and that multiple receptors are involved during EV71 infection. In addition, HSP70 was upregulated in human neuroblastoma cells (SK-N-SH) infected with EV71.


Assuntos
Enterovirus Humano A/metabolismo , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/virologia , Proteínas de Choque Térmico HSP70/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Linhagem Celular , Técnicas de Silenciamento de Genes , Proteínas de Choque Térmico HSP70/genética , Humanos , Neuroblastoma/virologia , Neurônios/virologia , RNA Interferente Pequeno , Regulação para Cima , Ligação Viral , Internalização do Vírus , Replicação Viral/fisiologia
17.
Virology ; 521: 69-76, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29886343

RESUMO

Enterovirus A71 (EV-A71) infection can cause hand, foot and mouth disease (HFMD), and even fatal meningoencephalitis. Unfortunately, there is currently no effective treatment for EV-A71 infection due to the lack of understanding of the mechanism of neurological diseases. In this study, we employed SH-SY5Y human neuroblastoma cells to explore the roles of caspase-1 in neuropathogenesis. The expression and activity of caspase-1 were analyzed. The potential immuneconsequences mediated by caspase-1 including cell death, lysis, DNA degradation, and secretion of pro-inflammatory were also examined. We found the gene expression levels of caspase-1, IL-1ß, IL-18 and active caspase-1 were markedly increased in the SH-SY5Y cells at 48 h post EV-A71 infection. The cell death, lysis, and DNA degradation were also increased during infection, which could be significantly alleviated by caspase-1 inhibition. These observations provided additional experimental evidence supporting caspase-1-mediated pyroptosis as a novel pathway of inflammatory programmed cell death.


Assuntos
Caspase 1/metabolismo , Enterovirus Humano A/patogenicidade , Doença de Mão, Pé e Boca/patologia , Piroptose , Células Cultivadas , Fragmentação do DNA , Humanos , Inflamação , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Neuroblastoma/patologia , Neuroblastoma/virologia , Replicação Viral
18.
Viruses ; 9(11)2017 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-29156632

RESUMO

Enterovirus 71 (EV71) is a common etiological agent of hand, foot, and mouth disease and fatal neurological diseases in children. The neuropathogenicity of severe EV71 infection has been documented, but studies comparing mouse models of severe and mild EV71 infection are lacking. The aim of the study was to investigate the neurovirulence of EV71 strains and the differences in serum cytokine and chemokine levels in mouse models of severe and mild EV71 infection. Nine EV71 isolates belonging to the C4 subgenogroup (proposed as genotype D) displayed infectivity in human neuroblastoma SK-N-SH cells; moreover, ultrastructural observation confirmed viral particle replication. The survival rate of the severe model was 71.43% (5/7), and 60% (3/5) of the surviving severe model mice displayed sequelae of paralysis, whereas the only symptom in mild model mice was ruffled fur. Dynamic detection of serum cytokine and chemokine levels demonstrated that interleukin (IL)-5, IL-13, IL-6, monocyte chemotactic protein 1 (MCP-1), and chemokine (C-C motif) ligand 5 (also called Regulated upon Activation, Normal T-cell Expressed, and Secreted (CCL5/RANTES) were significantly up-regulated at the early period of infection, indicating that these factors might herald a severe outcome. Our findings suggest that elevated cytokines and chemokines may have potential value as prognostic markers in mouse models.


Assuntos
Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/virologia , Tropismo Viral , Animais , Biomarcadores/sangue , Linhagem Celular , Quimiocinas/sangue , Citocinas/sangue , Modelos Animais de Doenças , Enterovirus Humano A/imunologia , Infecções por Enterovirus/imunologia , Humanos , Camundongos , Neuroblastoma/virologia , Carga Viral , Virulência , Replicação Viral
19.
J Virol ; 91(21)2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28794031

RESUMO

Bovine herpesvirus 1 (BoHV-1), an important bovine pathogen, establishes lifelong latency in sensory neurons. Latently infected calves consistently reactivate from latency following a single intravenous injection of the synthetic corticosteroid dexamethasone. The immediate early transcription unit 1 (IEtu1) promoter, which drives bovine ICP0 (bICP0) and bICP4 expression, is stimulated by dexamethasone because it contains two glucocorticoid receptor (GR) response elements (GREs). Several Krüppel-like transcription factors (KLF), including KLF15, are induced during reactivation from latency, and they stimulate certain viral promoters and productive infection. In this study, we demonstrate that the GR and KLF15 were frequently expressed in the same trigeminal ganglion (TG) neuron during reactivation and cooperatively stimulated productive infection and IEtu1 GREs in mouse neuroblastoma cells (Neuro-2A). We further hypothesized that additional regions in the BoHV-1 genome are transactivated by the GR or stress-induced transcription factors. To test this hypothesis, BoHV-1 DNA fragments (less than 400 bp) containing potential GR and KLF binding sites were identified and examined for transcriptional activation by stress-induced transcription factors. Intergenic regions within the unique long 52 gene (UL52; a component of the DNA primase/helicase complex), bICP4, IEtu2, and the unique short region were stimulated by KLF15 and the GR. Chromatin immunoprecipitation studies revealed that the GR and KLF15 interacted with sequences within IEtu1 GREs and the UL52 fragment. Coimmunoprecipitation studies demonstrated that KLF15 and the GR were associated with each other in transfected cells. Since the GR stimulates KLF15 expression, we suggest that these two transcription factors form a feed-forward loop that stimulates viral gene expression and productive infection following stressful stimuli.IMPORTANCE Bovine herpesvirus 1 (BoHV-1) is an important viral pathogen that causes respiratory disease and suppresses immune responses in cattle; consequently, life-threatening bacterial pneumonia can occur. Following acute infection, BoHV-1 establishes lifelong latency in sensory neurons. Reactivation from latency is initiated by the synthetic corticosteroid dexamethasone. Dexamethasone stimulates lytic cycle viral gene expression in sensory neurons of calves latently infected with BoHV-1, culminating in virus shedding and transmission. Two stress-induced cellular transcription factors, Krüppel-like transcription factor 15 (KLF15) and the glucocorticoid receptor (GR), cooperate to stimulate productive infection and viral transcription. Additional studies demonstrated that KLF15 and the GR form a stable complex and that these stress-induced transcription factors bind to viral DNA sequences, which correlates with transcriptional activation. The ability of the GR and KLF15 to synergistically stimulate viral gene expression and productive infection may be critical for the ability of BoHV-1 to reactivate from latency following stressful stimuli.


Assuntos
Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/fisiologia , Fatores de Transcrição Kruppel-Like/metabolismo , Receptores de Glucocorticoides/metabolismo , Transcrição Gênica , Ativação Viral , Animais , Sítios de Ligação , Bovinos , Doenças dos Bovinos/virologia , Células Cultivadas , Imunoprecipitação da Cromatina , Regulação Viral da Expressão Gênica , Infecções por Herpesviridae/metabolismo , Rim/metabolismo , Rim/virologia , Camundongos , Neuroblastoma/metabolismo , Neuroblastoma/virologia , Regiões Promotoras Genéticas , Coelhos , Elementos de Resposta , Pele/metabolismo , Pele/virologia , Transativadores , Gânglio Trigeminal/metabolismo , Gânglio Trigeminal/virologia , Latência Viral
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(6): 761-766, 2017 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-28615098

RESUMO

Objective To study the mechanism ofhuman enterovirus 71 (EV71) entering human neuroblastoma SK-N-SH cells. Methods After the SK-N-SH cells were pretreated with chlorpromazine (CPZ) or nystatin (NT), real-time quantitative PCR (qRT-PCR) was employed to measure EV71 mRNA level, and indirect immunofluorescence microscopy was used to detect the expression level of viral protein 1 (VP1) in the target cells. In order to reveal the colocalization of EV71 with clathrin, laser confocal microscopy was performed on the infected cells. Results CPZ could significantly inhibit EV71 mRNA level and the expression of VP1 in the target cells, while NT had no effect on EV71 infection. Confocal microscopy showed that EV71 was colocalize with clathrin. Conclusion EV71 infects human neuroblastoma SK-N-SH cells by the clathrin-mediated endocytosis.


Assuntos
Clatrina/fisiologia , Endocitose , Enterovirus Humano A/genética , Neuroblastoma/virologia , Proteínas do Capsídeo/análise , Linhagem Celular Tumoral , Clorpromazina/farmacologia , Humanos , Neuroblastoma/patologia , Nistatina/farmacologia , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA