Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78.449
Filter
1.
World J Gastroenterol ; 30(26): 3261-3263, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39086637

ABSTRACT

This letter to the editor relates to the study entitled "Tenofovir amibufenamide vs tenofovir alafenamide for treating chronic hepatitis B: A real-world study", which was recently published by Peng et al. Hepatitis B virus infection represents a significant health burden worldwide and can lead to cirrhosis and even liver cancer. The antiviral drugs currently used to treat patients with chronic hepatitis B infection still have many side effects, so it is crucial to identify safe and effective drugs to inhibit viral replication.


Subject(s)
Antiviral Agents , Hepatitis B virus , Hepatitis B, Chronic , Tenofovir , Humans , Antiviral Agents/therapeutic use , Antiviral Agents/adverse effects , Hepatitis B, Chronic/drug therapy , Hepatitis B, Chronic/virology , Tenofovir/therapeutic use , Tenofovir/analogs & derivatives , Tenofovir/adverse effects , Hepatitis B virus/drug effects , Treatment Outcome , Virus Replication/drug effects , Adenine/analogs & derivatives , Adenine/therapeutic use , Adenine/adverse effects , Alanine/analogs & derivatives , Alanine/therapeutic use , Alanine/adverse effects
2.
Virol J ; 21(1): 170, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090742

ABSTRACT

BACKGROUND: Chronic hepatitis B virus (HBV) infection affects around 250 million people worldwide, causing approximately 887,000 deaths annually, primarily owing to cirrhosis and hepatocellular carcinoma (HCC). The current approved treatments for chronic HBV infection, such as interferon and nucleos(t)ide analogs, have certain limitations as they cannot completely eradicate covalently closed circular DNA (cccDNA). Considering that HBV replication relies on host transcription factors, focusing on host factors in the HBV genome may provide insights into new therapeutic targets against HBV. Therefore, understanding the mechanisms underlying viral persistence and hepatocyte pathogenesis, along with the associated host factors, is crucial. In this study, we investigated novel therapeutic targets for HBV infection by identifying gene and pathway networks involved in HBV replication in primary human hepatocytes (PHHs). Importantly, our study utilized cultured primary hepatocytes, allowing transcriptomic profiling in a biologically relevant context and enabling the investigation of early HBV-mediated effects. METHODS: PHHs were infected with HBV virion particles derived from HepAD38 cells at 80 HBV genome equivalents per cell (Geq/cell). For transcriptomic sequencing, PHHs were harvested 1, 2-, 3-, 5-, and 7 days post-infection (dpi). After preparing the libraries, clustering and sequencing were conducted to generate RNA-sequencing data. This data was processed using Bioinformatics tools and software to analyze DEGs and obtain statistically significant results. Furthermore, qRT-PCR was performed to validate the RNA-sequencing results, ensuring consistent findings. RESULTS: We observed significant alterations in the expression patterns of 149 genes from days 1 to 7 following HBV infection (R2 > 0.7, q < 0.05). Functional analysis of these genes identified RNA-binding proteins involved in mRNA metabolism and the regulation of alternative splicing during HBV infection. Results from qRT-PCR experiments and the analysis of two validation datasets suggest that RBM14 and RPL28 may serve as potential biomarkers for HBV-associated HCC. CONCLUSIONS: Transcriptome analysis of gene expression changes during HBV infection in PHHs provided valuable insights into chronic HBV infection. Additionally, understanding the functional involvement of host factor networks in the molecular mechanisms of HBV replication and transcription may facilitate the development of novel strategies for HBV treatment.


Subject(s)
Hepatitis B virus , Hepatocytes , Virus Replication , Humans , Hepatocytes/virology , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Gene Expression Profiling , Host-Pathogen Interactions , Cells, Cultured , Gene Regulatory Networks , Hepatitis B/virology , Hepatitis B/genetics , Hepatitis B, Chronic/virology
3.
Nat Commun ; 15(1): 6503, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090095

ABSTRACT

The COVID-19 pandemic has led to the deaths of millions of people and severe global economic impacts. Small molecule therapeutics have played an important role in the fight against SARS-CoV-2, the virus responsible for COVID-19, but their efficacy has been limited in scope and availability, with many people unable to access their benefits, and better options are needed. EDP-235 is specifically designed to inhibit the SARS-CoV-2 3CLpro, with potent nanomolar activity against all SARS-CoV-2 variants to date, as well as clinically relevant human and zoonotic coronaviruses. EDP-235 maintains potency against variants bearing mutations associated with nirmatrelvir resistance. Additionally, EDP-235 demonstrates a ≥ 500-fold selectivity index against multiple host proteases. In a male Syrian hamster model of COVID-19, EDP-235 suppresses SARS-CoV-2 replication and viral-induced hamster lung pathology. In a female ferret model, EDP-235 inhibits production of SARS-CoV-2 infectious virus and RNA at multiple anatomical sites. Furthermore, SARS-CoV-2 contact transmission does not occur when naïve ferrets are co-housed with infected, EDP-235-treated ferrets. Collectively, these results demonstrate that EDP-235 is a broad-spectrum coronavirus inhibitor with efficacy in animal models of primary infection and transmission.


Subject(s)
Antiviral Agents , COVID-19 , Coronavirus 3C Proteases , SARS-CoV-2 , Virus Replication , Animals , Cricetinae , Female , Humans , Male , Antiviral Agents/pharmacology , Chlorocebus aethiops , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , COVID-19/virology , COVID-19/transmission , COVID-19 Drug Treatment , Disease Models, Animal , Ferrets , Lactams , Leucine , Lung/virology , Lung/drug effects , Lung/pathology , Mesocricetus , Nitriles , Organic Chemicals , Pandemics/prevention & control , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Pneumonia, Viral/transmission , Pneumonia, Viral/prevention & control , Proline , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Vero Cells , Virus Replication/drug effects
4.
Sci Rep ; 14(1): 18929, 2024 08 15.
Article in English | MEDLINE | ID: mdl-39147857

ABSTRACT

Porcine Epidemic Diarrhea Virus (PEDV) poses a significant threat to neonatal piglets, particularly due to the limited efficacy of existing vaccines and the scarcity of efficacious therapeutic drugs. Gegen Qinlian Decoction (GQD) has been employed for over two millennia in treating infectious diarrhea. Nonetheless, further scrutiny is required to improve the drug's efficacy and elucidate its underlying mechanisms of action. In this study, a modified GQD (MGQD) was developed and demonstrated its capacity to inhibit the replication of PEDV. Animal trials indicated that MGQD effectively alleviated pathological damage in immune tissues and modulated T-lymphocyte subsets. The integration of network analysis with UHPLC-MS/MS facilitated the identification of active ingredients within MGQD and elucidated the molecular mechanisms underlying its therapeutic effects against PEDV infections. In vitro studies revealed that MGQD significantly impeded PEDV proliferation in IPEC-J2 cells, promoting cellular growth via virucidal activity, inhibition of viral attachment, and disruption of viral biosynthesis. Furthermore, MGQD treatment led to increased expression levels of IFN-α, IFN-ß, and IFN-λ3, while concurrently decreasing the expression of TNF-α, thereby enhancing resistance to PEDV infection in IPEC-J2 cells. In conclusion, our findings suggest that MGQD holds promise as a novel antiviral agent for the treatment of PEDV infections.


Subject(s)
Coronavirus Infections , Drugs, Chinese Herbal , Network Pharmacology , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Porcine epidemic diarrhea virus/drug effects , Swine , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Swine Diseases/drug therapy , Swine Diseases/virology , Antiviral Agents/pharmacology , Virus Replication/drug effects , Cell Line , Tandem Mass Spectrometry , Diarrhea/drug therapy , Diarrhea/virology , Diarrhea/veterinary , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
5.
Zool Res ; 45(5): 990-1000, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39147714

ABSTRACT

The von Hippel-Lindau tumor suppressor protein (VHL), an E3 ubiquitin ligase, functions as a critical regulator of the oxygen-sensing pathway for targeting hypoxia-inducible factors. Recent evidence suggests that mammalian VHL may also be critical to the NF-κB signaling pathway, although the specific molecular mechanisms remain unclear. Herein, the roles of mandarin fish ( Siniperca chuatsi) VHL ( scVHL) in the NF-κB signaling pathway and mandarin fish ranavirus (MRV) replication were explored. The transcription of scVHL was induced by immune stimulation and MRV infection, indicating a potential role in innate immunity. Dual-luciferase reporter gene assays and reverse transcription quantitative PCR (RT-qPCR) results demonstrated that scVHL evoked and positively regulated the NF-κB signaling pathway. Treatment with NF-κB signaling pathway inhibitors indicated that the role of scVHL may be mediated through scIKKα, scIKKß, scIκBα, or scp65. Co-immunoprecipitation (Co-IP) analysis identified scIκBα as a novel target protein of scVHL. Moreover, scVHL targeted scIκBα to catalyze the formation of K63-linked polyubiquitin chains to activate the NF-κB signaling pathway. Following MRV infection, NF-κB signaling remained activated, which, in turn, promoted MRV replication. These findings suggest that scVHL not only positively regulates NF-κB but also significantly enhances MRV replication. This study reveals a novel function of scVHL in NF-κB signaling and viral infection in fish.


Subject(s)
Fish Diseases , NF-kappa B , Ranavirus , Signal Transduction , Virus Replication , Animals , NF-kappa B/metabolism , NF-kappa B/genetics , Virus Replication/physiology , Fish Diseases/virology , Ranavirus/physiology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , DNA Virus Infections/veterinary , DNA Virus Infections/virology , Fish Proteins/metabolism , Fish Proteins/genetics , I-kappa B Proteins/metabolism , I-kappa B Proteins/genetics , Gene Expression Regulation
6.
Proc Natl Acad Sci U S A ; 121(35): e2406421121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39159381

ABSTRACT

Viral infection is frequently assayed by ongoing expression of viral genes. These assays fail to identify cells that have been exposed to the virus but limit or inhibit viral replication. To address this limitation, we used a dual-labeling vesicular stomatitis virus (DL-VSV), which has a deletion of the viral glycoprotein gene, to allow evaluation of primary infection outcomes. This virus encodes Cre, which can stably mark any cell with even a minimal level of viral gene expression. Additionally, the virus encodes GFP, which distinguishes cells with higher levels of viral gene expression, typically due to genome replication. Stereotactic injections of DL-VSV into the murine brain showed that different cell types had very different responses to the virus. Almost all neurons hosted high levels of viral gene expression, while glial cells varied in their responses. Astrocytes (Sox9+) were predominantly productively infected, while oligodendrocytes (Sox10+) were largely abortively infected. Microglial cells (Iba1+) were primarily uninfected. Furthermore, we monitored the early innate immune response to viral infection and identified unique patterns of interferon (IFN) induction. Shortly after infection, microglia were the main producers of IFNb, whereas later, oligodendrocytes were the main producers. IFNb+ cells were primarily abortively infected regardless of cell type. Last, we investigated whether IFN signaling had any impact on the outcome of primary infection and did not observe significant changes, suggesting that intrinsic factors are likely responsible for determining the outcome of primary infection.


Subject(s)
Astrocytes , Animals , Mice , Astrocytes/virology , Astrocytes/metabolism , Virus Replication , Microglia/virology , Microglia/metabolism , Microglia/immunology , Neurons/virology , Neurons/metabolism , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Vesiculovirus/physiology , Vesiculovirus/immunology , Vesiculovirus/genetics , Oligodendroglia/virology , Oligodendroglia/metabolism , Vesicular Stomatitis/virology , Vesicular Stomatitis/immunology , Immunity, Innate , Mice, Inbred C57BL , Brain/virology , Brain/metabolism , Brain/immunology , Neuroglia/virology , Neuroglia/metabolism
7.
Front Cell Infect Microbiol ; 14: 1383917, 2024.
Article in English | MEDLINE | ID: mdl-39119292

ABSTRACT

Introduction: Oxysterol-binding protein (OSBP) is known for its crucial role in lipid transport, facilitating cholesterol exchange between the Golgi apparatus and endoplasmic reticulum membranes. Despite its established function in cellular processes, its involvement in coronavirus replication remains unclear. Methods: In this study, we investigated the role of OSBP in coronavirus replication and explored the potential of a novel OSBP-binding compound, ZJ-1, as an antiviral agent against coronaviruses, including SARS-CoV-2. We utilized a combination of biochemical and cellular assays to elucidate the interactions between OSBP and SARS-CoV-2 non-structural proteins (Nsps) and other viral proteins. Results: Our findings demonstrate that OSBP positively regulates coronavirus replication. Moreover, treatment with ZJ-1 resulted in reduced OSBP levels and exhibited potent antiviral effects against multiple coronaviruses. Through our investigation, we identified specific interactions between OSBP and SARS-CoV-2 Nsps, particularly Nsp3, Nsp4, and Nsp6, which are involved in double-membrane vesicle formation-a crucial step in viral replication. Additionally, we observed that Nsp3 a.a.1-1363, Nsp4, and Nsp6 target vesicle-associated membrane protein (VAMP)-associated protein B (VAP-B), which anchors OSBP to the ER membrane. Interestingly, the interaction between OSBP and VAP-B is disrupted by Nsp3 a.a.1-1363 and partially impaired by Nsp6. Furthermore, we identified SARS-CoV-2 orf7a, orf7b, and orf3a as additional OSBP targets, with OSBP contributing to their stabilization. Conclusion: Our study highlights the significance of OSBP in coronavirus replication and identifies it as a promising target for the development of antiviral therapies against SARS-CoV-2 and other coronaviruses. These findings underscore the potential of OSBP-targeted interventions in combating coronavirus infections.


Subject(s)
Antiviral Agents , Receptors, Steroid , SARS-CoV-2 , Viral Nonstructural Proteins , Virus Replication , Virus Replication/drug effects , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Antiviral Agents/pharmacology , Receptors, Steroid/metabolism , Viral Nonstructural Proteins/metabolism , COVID-19/virology , COVID-19/metabolism , Chlorocebus aethiops , Vero Cells , Viral Proteins/metabolism , HEK293 Cells , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Viroporin Proteins/metabolism , Coronavirus Papain-Like Proteases/metabolism , Protein Binding
8.
Sci Adv ; 10(32): eadn9519, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110796

ABSTRACT

While the significance of N6-methyladenosine (m6A) in viral regulation has been extensively studied, the functions of 5-methylcytosine (m5C) modification in viral biology remain largely unexplored. In this study, we demonstrate that m5C is more abundant than m6A in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and provide a comprehensive profile of the m5C landscape of SARS-CoV-2 RNA. Knockout of NSUN2 reduces m5C levels in SARS-CoV-2 virion RNA and enhances viral replication. Nsun2 deficiency mice exhibited higher viral burden and more severe lung tissue damages. Combined RNA-Bis-seq and m5C-MeRIP-seq identified the NSUN2-dependent m5C-methylated cytosines across the positive-sense genomic RNA of SARS-CoV-2, and the mutations of these cytosines enhance RNA stability. The progeny SARS-CoV-2 virions from Nsun2 deficiency mice with low levels of m5C modification exhibited a stronger replication ability. Overall, our findings uncover the vital role played by NSUN2-mediated m5C modification during SARS-CoV-2 replication and propose a host antiviral strategy via epitranscriptomic addition of m5C methylation to SARS-CoV-2 RNA.


Subject(s)
COVID-19 , RNA, Viral , SARS-CoV-2 , Virus Replication , Virus Replication/genetics , Animals , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , SARS-CoV-2/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , COVID-19/virology , COVID-19/pathology , Mice , Humans , Methylation , Virulence/genetics , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , Epigenesis, Genetic , Mice, Knockout , Adenosine/analogs & derivatives , Adenosine/metabolism , Transcriptome
9.
Virol J ; 21(1): 181, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118175

ABSTRACT

The orf virus (ORFV) poses a serious threat to the health of domestic small ruminants (i.e., sheep and goats) and humans on a global scale, causing around $150 million in annual losses to livestock industry. However, the host factors involved in ORFV infection and replication are still elusive. In this study, we compared the RNA-seq profiles of ORFV-infected or non-infected sheep testicular interstitial cells (STICs) and identified a novel host gene, potassium voltage-gated channel subfamily E member 4 (KCNE4), as a key host factor involved in the ORFV infection. Both RNA-seq data and RT-qPCR assay revealed a significant increase in the expression of KCNE4 in the infected STICs from 9 to 48 h post infection (hpi). On the other hand, the RT-qPCR assay detected a decrease in ORFV copy number in both the STICs transfected by KCNE4 siRNA and the KCNE4 knockout (KO) HeLa cells after the ORFV infection, together with a reduced fluorescence ratio of ORFV-GFP in the KO HeLa cells at 24 hpi, indicating KCNE4 to be critical for the ORFV infection. Furthermore, the attachment and internalization assays showed decreased ORFV attachment, internalization, replication, and release by the KO HeLa cells, demonstrating a potential inhibition of ORFV entry into the cells by KCNE4. Pretreatment with the KCNE4 inhibitors such as quinidine and fluoxetine significantly repressed the ORFV infection. All our findings reveal KCNE4 as a novel host regulator of the ORFV entry and replication, shedding new insight into the interactive mechanism of ORFV infection. The study also highlights the K+ channels as possible druggable targets to impede viral infection and disease.


Subject(s)
Orf virus , Potassium Channels, Voltage-Gated , Virus Internalization , Animals , Humans , Sheep , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , HeLa Cells , Orf virus/genetics , Orf virus/physiology , Virus Replication , Host-Pathogen Interactions , Male , Ecthyma, Contagious/virology
10.
Virology ; 598: 110192, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39106585

ABSTRACT

In vitro studies have shown that deletion of nef and deleterious mutation in the Nef dimerization interface attenuates HIV replication and associated pathogenesis. Humanized rodents with human immune cells and lymphoid tissues are robust in vivo models for investigating the interactions between HIV and the human immune system. Here, we demonstrate that nef deletion impairs HIV replication and HIV-induced immune dysregulation in the blood and human secondary lymphoid tissue (human spleen) in bone marrow-liver-thymus-spleen (BLTS) humanized mice. Furthermore, we also show that nef defects (via deleterious mutations in the dimerization interface) impair HIV replication and HIV-induced immune dysregulation in the blood and human spleen in BLTS-humanized mice. We demonstrate that the reduced replication of nef-deleted and nef-defective HIV is associated with robust antiviral innate immune response, and T helper 1 response. Our results support the proposition that Nef may be a therapeutic target for adjuvants in HIV cure strategies.


Subject(s)
Disease Models, Animal , HIV Infections , HIV-1 , Liver , Spleen , Viremia , Virus Replication , nef Gene Products, Human Immunodeficiency Virus , Animals , nef Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/immunology , HIV Infections/immunology , HIV Infections/virology , Mice , Humans , Viremia/immunology , Spleen/immunology , Spleen/virology , HIV-1/immunology , HIV-1/genetics , HIV-1/physiology , Liver/virology , Liver/immunology , Liver/pathology , Bone Marrow/virology , Bone Marrow/immunology , Thymus Gland/immunology , Thymus Gland/virology , Immunity, Innate
11.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125583

ABSTRACT

Coronaviruses constitute a global threat to human and animal health. It is essential to investigate the long-distance RNA-RNA interactions that approximate remote regulatory elements in strategies, including genome circularization, discontinuous transcription, and transcriptional enhancers, aimed at the rapid replication of their large genomes, pathogenicity, and immune evasion. Based on the primary sequences and modeled RNA-RNA interactions of two experimentally defined coronaviral enhancers, we detected via an in silico primary and secondary structural analysis potential enhancers in various coronaviruses, from the phylogenetically ancient avian infectious bronchitis virus (IBV) to the recently emerged SARS-CoV-2. These potential enhancers possess a core duplex-forming region that could transition between closed and open states, as molecular switches directed by viral or host factors. The duplex open state would pair with remote sequences in the viral genome and modulate the expression of downstream crucial genes involved in viral replication and host immune evasion. Consistently, variations in the predicted IBV enhancer region or its distant targets coincide with cases of viral attenuation, possibly driven by decreased open reading frame (ORF)3a immune evasion protein expression. If validated experimentally, the annotated enhancer sequences could inform structural prediction tools and antiviral interventions.


Subject(s)
Enhancer Elements, Genetic , Genome, Viral , Infectious bronchitis virus , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Infectious bronchitis virus/genetics , Humans , Enhancer Elements, Genetic/genetics , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , COVID-19/virology , COVID-19/genetics , Betacoronavirus/genetics , Virus Replication/genetics , Coronavirus Infections/virology , Transcription, Genetic , Gene Expression Regulation, Viral , Pneumonia, Viral/virology
12.
Science ; 385(6709): eadn5866, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39116226

ABSTRACT

Antiviral therapies with reduced frequencies of administration and high barriers to resistance remain a major goal. For HIV, theories have proposed that viral-deletion variants, which conditionally replicate with a basic reproductive ratio [R0] > 1 (termed "therapeutic interfering particles" or "TIPs"), could parasitize wild-type virus to constitute single-administration, escape-resistant antiviral therapies. We report the engineering of a TIP that, in rhesus macaques, reduces viremia of a highly pathogenic model of HIV by >3log10 following a single intravenous injection. Animal lifespan was significantly extended, TIPs conditionally replicated and were continually detected for >6 months, and sequencing data showed no evidence of viral escape. A single TIP injection also suppressed virus replication in humanized mice and cells from persons living with HIV. These data provide proof of concept for a potential new class of single-administration antiviral therapies.


Subject(s)
Artificial Virus-Like Particles , Gene Deletion , HIV Infections , HIV-1 , Viral Interference , Virus Replication , Animals , Humans , Mice , Basic Reproduction Number , Disease Models, Animal , Genetic Engineering , HIV Infections/therapy , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Macaca mulatta , Proof of Concept Study , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Viremia/therapy , Viremia/virology
14.
Sci Rep ; 14(1): 18509, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122768

ABSTRACT

Rabies virus (RABV) is the causative agent of rabies, a lethal neurological disease in mammals. RABV strains can be classified into fixed strains (laboratory strains) and street strains (field/clinical strains), which have different properties including cell tropism and neuroinvasiveness. RABV Toyohashi strain is a street strain isolated in Japan from an imported case which had been bitten by rabid dog in the Philippines. In order to facilitate molecular studies of RABV, we established a reverse genetics (RG) system for the study of the Toyohashi strain. The recombinant virus was obtained from a cDNA clone of Toyohashi strain and exhibited similar growth efficiency as the original virus in cultured cell lines. Both the original and recombinant strains showed similar pathogenicity with high neuroinvasiveness in mice, and the infected mice developed a long and inconsistent incubation period, which is characteristic of street strains. We also generated a recombinant Toyohashi strain expressing viral phosphoprotein (P protein) fused with the fluorescent protein mCherry, and tracked the intracellular dynamics of the viral P protein using live-cell imaging. The presented reverse genetics system for Toyohashi strain will be a useful tool to explore the fundamental molecular mechanisms of the replication of RABV street strains.


Subject(s)
Rabies virus , Rabies , Reverse Genetics , Rabies virus/genetics , Rabies virus/pathogenicity , Animals , Reverse Genetics/methods , Mice , Rabies/virology , Dogs , Humans , Cell Line , Virus Replication/genetics , Philippines
15.
Arch Virol ; 169(9): 175, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117748

ABSTRACT

Newcastle disease virus (NDV), an avian paramyxovirus, causes major economic losses in the poultry industry worldwide. NDV strains are classified as avirulent, moderately virulent, or virulent according to the severity of the disease they cause. In order to gain a deeper understanding of the molecular mechanisms of virus-host interactions, we conducted Illumina HiSeq-based RNA-Seq analysis on chicken embryo fibroblast (DF1) cells during the first 24 hours of infection with NDV strain Komarov. Comparative analysis of uninfected DF1 cells versus NDV-infected DF1 cells at 6, 12, and 24 h postinfection identified 462, 459, and 410 differentially expressed genes, respectively. The findings revealed an increase in the expression of genes linked to the MAPK signalling pathway in the initial stages of NDV infection. This overexpression potentially aids viral multiplication while hindering pathogen detection and subsequent immune responses from the host. Our findings provide initial insights into the early responses of DF1 cells to NDV infection.


Subject(s)
Chickens , Fibroblasts , Gene Expression Profiling , Host-Pathogen Interactions , Newcastle Disease , Newcastle disease virus , Newcastle disease virus/genetics , Newcastle disease virus/pathogenicity , Newcastle disease virus/physiology , Animals , Newcastle Disease/virology , Newcastle Disease/immunology , Chickens/virology , Fibroblasts/virology , Host-Pathogen Interactions/genetics , Chick Embryo , Cell Line , Transcriptome , Poultry Diseases/virology , Poultry Diseases/genetics , Virus Replication/genetics
16.
PLoS Pathog ; 20(8): e1012388, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102425

ABSTRACT

Enteroviruses are a vast genus of positive-sense RNA viruses that cause diseases ranging from common cold to poliomyelitis and viral myocarditis. They encode a membrane-bound AAA+ ATPase, 2C, that has been suggested to serve several roles in virus replication, e.g. as an RNA helicase and capsid assembly factor. Here, we report the reconstitution of full-length, poliovirus 2C's association with membranes. We show that the N-terminal membrane-binding domain of 2C contains a conserved glycine, which is suggested by structure predictions to divide the domain into two amphipathic helix regions, which we name AH1 and AH2. AH2 is the main mediator of 2C oligomerization, and is necessary and sufficient for its membrane binding. AH1 is the main mediator of a novel function of 2C: clustering of membranes. Cryo-electron tomography reveal that several 2C copies mediate this function by localizing to vesicle-vesicle interfaces. 2C-mediated clustering is partially outcompeted by RNA, suggesting a way by which 2C can switch from an early role in coalescing replication organelles and lipid droplets, to a later role where 2C assists RNA replication and particle assembly. 2C is sufficient to recruit RNA to membranes, with a preference for double-stranded RNA (the replicating form of the viral genome). Finally, the in vitro reconstitution revealed that full-length, membrane-bound 2C has ATPase activity and ATP-independent, single-strand ribonuclease activity, but no detectable helicase activity. Together, this study suggests novel roles for 2C in membrane clustering, RNA membrane recruitment and cleavage, and calls into question a role of 2C as an RNA helicase. The reconstitution of functional, 2C-decorated vesicles provides a platform for further biochemical studies into this protein and its roles in enterovirus replication.


Subject(s)
RNA, Viral , Viral Proteins , Virus Replication , RNA, Viral/metabolism , RNA, Viral/genetics , Humans , Virus Replication/physiology , Viral Proteins/metabolism , Viral Proteins/genetics , Poliovirus/metabolism , Poliovirus/physiology , Cell Membrane/metabolism , Enterovirus/physiology , Adenosine Triphosphatases/metabolism , Carrier Proteins , Viral Nonstructural Proteins
17.
Vet Res ; 55(1): 101, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143491

ABSTRACT

Senecavirus A (SVA) is an emerging virus that poses a threat to swine herds worldwide. To date, the role of tripartite motif 5 (TRIM5) in the replication of viruses has not been evaluated. Here, TRIM5 was reported to inhibit SVA replication by promoting the type I interferon (IFN) antiviral response mediated by retinoic acid-inducible gene I (RIG-I). TRIM5 expression was significantly upregulated in SVA-infected cells, and TRIM5 overexpression inhibited viral replication and promoted IFN-α, IFN-ß, interleukin-1beta (IL-1ß), IL-6, and IL-18 expression. Conversely, interfering with the expression of TRIM5 had the opposite effect. Viral adsorption and entry assays showed that TRIM5 did not affect the adsorption of SVA but inhibited its entry. In addition, TRIM5 promoted the expression of RIG-I and RIG-I-mediated IFNs and proinflammatory cytokines, and this effect was also proven by inhibiting the expression of TRIM5. These findings expand the scope of knowledge on host factors inhibiting the replication of SVA and indicate that targeting TRIM5 may aid in the development of new agents against SVA.


Subject(s)
Interferon Type I , Picornaviridae , Virus Replication , Animals , Interferon Type I/metabolism , Swine , Picornaviridae/physiology , Picornaviridae/immunology , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Swine Diseases/virology , Swine Diseases/immunology
18.
Emerg Microbes Infect ; 13(1): 2387439, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39139051

ABSTRACT

Avian influenza viruses (AIVs) are the origin of multiple mammal influenza viruses. The genetic determinants of AIVs adapted to humans have been widely elucidated, however, the molecular mechanism of cross-species transmission and adaptation of AIVs to canines are still poorly understood. In this study, two H3N2 influenza viruses isolated from a live poultry market (A/environment/Guangxi/13431/2018, GX13431) and a swab sample from a canine (A/canine/Guangdong/0601/2019, GD0601) were used to investigate the possible molecular basis that determined H3N2 AIV adapting to canine. We found that GD0601 exhibited more robust polymerase activity in cells and higher pathogenicity in mice compared with its evolution ancestor H3N2 AIV GX13431. A series of reassortments of the ribonucleoprotein (RNP) complex showed that the PB2 subunit was the crucial factor that conferred high polymerase activity of GD0601, and the substitution of I714S in the PB2 subunit of GD0601 attenuated the replication and pathogenicity in mammal cells and the mouse model. Mechanistically, the reverse mutation of I714S in the PB2 polymerase subunit which was identified in AIV GX13431 reduced the nuclear import efficiency of PB2 protein and interfered with the interactions of PB2-PA/NP that affected the assembly of the viral RNP complex. Our study reveals amino acid mutation at the position of 714 in the nuclear localization signal (NLS) area in PB2 plays an important role in overcoming the barrier from poultry to mammals of the H3N2 canine influenza virus and provides clues for further study of mammalian adaptation mechanism of AIVs.


Subject(s)
Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections , RNA-Dependent RNA Polymerase , Ribonucleoproteins , Viral Proteins , Animals , Dogs , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Mice , Viral Proteins/genetics , Viral Proteins/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Orthomyxoviridae Infections/virology , Humans , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Active Transport, Cell Nucleus , Virus Replication , Mutation , Madin Darby Canine Kidney Cells , Dog Diseases/virology , Mice, Inbred BALB C , HEK293 Cells , Reassortant Viruses/genetics
19.
Nat Commun ; 15(1): 6664, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164284

ABSTRACT

Hyperglycemia, and exacerbation of pre-existing deficits in glucose metabolism, are manifestations of the post-acute sequelae of SARS-CoV-2. Our understanding of metabolic decline after acute COVID-19 remains unclear due to the lack of animal models. Here, we report a non-human primate model of metabolic post-acute sequelae of SARS-CoV-2 using SARS-CoV-2 infected African green monkeys. Using this model, we identify a dysregulated blood chemokine signature during acute COVID-19 that correlates with elevated and persistent hyperglycemia four months post-infection. Hyperglycemia also correlates with liver glycogen levels, but there is no evidence of substantial long-term SARS-CoV-2 replication in the liver and pancreas. Finally, we report a favorable glycemic effect of the SARS-CoV-2 mRNA vaccine, administered on day 4 post-infection. Together, these data suggest that the African green monkey model exhibits important similarities to humans and can be utilized to assess therapeutic candidates to combat COVID-related metabolic defects.


Subject(s)
COVID-19 , Disease Models, Animal , Hyperglycemia , Liver , SARS-CoV-2 , Animals , Hyperglycemia/immunology , COVID-19/immunology , COVID-19/virology , COVID-19/blood , Chlorocebus aethiops , SARS-CoV-2/immunology , Liver/virology , Liver/metabolism , Liver/immunology , Glycogen/metabolism , Blood Glucose/metabolism , Humans , Male , Pancreas/virology , Pancreas/immunology , Pancreas/pathology , Pancreas/metabolism , Chemokines/metabolism , Chemokines/blood , Female , Virus Replication
20.
Sci Rep ; 14(1): 18470, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122799

ABSTRACT

The microbial communities residing in the mosquito midgut play a key role in determining the outcome of mosquito pathogen infection. Elizabethkingia anophelis, originally isolated from the midgut of Anopheles gambiae possess a broad-spectrum antiviral phenotype, yet a gap in knowledge regarding the mechanistic basis of its interaction with viruses exists. The current study aims to identify pathways and genetic factors linked to E. anophelis antiviral activity. The understanding of E. anophelis antiviral mechanism could lead to novel transmission barrier tools to prevent arboviral outbreaks. We utilized a non-targeted multi-omics approach, analyzing extracellular lipids, proteins, metabolites of culture supernatants coinfected with ZIKV and E. anophelis. We observed a significant decrease in arginine and phenylalanine levels, metabolites that are essential for viral replication and progression of viral infection. This study provides insights into the molecular basis of E. anophelis antiviral phenotype. The findings lay a foundation for in-depth mechanistic studies.


Subject(s)
Flavobacteriaceae , Zika Virus , Zika Virus/physiology , Animals , Flavobacteriaceae/metabolism , Flavobacteriaceae/genetics , Anopheles/virology , Anopheles/microbiology , Zika Virus Infection/virology , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Virus Replication , Phenylalanine/metabolism , Arginine/metabolism , Multiomics
SELECTION OF CITATIONS
SEARCH DETAIL