Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 484
Filter
1.
PLoS Pathog ; 20(9): e1012511, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39298524

ABSTRACT

Molecular details of genome packaging are little understood for the majority of viruses. In enteroviruses (EVs), cleavage of the structural protein VP0 into VP4 and VP2 is initiated by the incorporation of RNA into the assembling virion and is essential for infectivity. We have applied a combination of bioinformatic, molecular and structural approaches to generate the first high-resolution structure of an intermediate in the assembly pathway, termed a provirion, which contains RNA and intact VP0. We have demonstrated an essential role of VP0 E096 in VP0 cleavage independent of RNA encapsidation and generated a new model of capsid maturation, supported by bioinformatic analysis. This provides a molecular basis for RNA-dependence, where RNA induces conformational changes required for VP0 maturation, but that RNA packaging itself is not sufficient to induce maturation. These data have implications for understanding production of infectious virions and potential relevance for future vaccine and antiviral drug design.


Subject(s)
Capsid Proteins , Virus Assembly , Virus Assembly/physiology , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , Virion/metabolism , Enterovirus/physiology , Capsid/metabolism , Enterovirus Infections/virology , Enterovirus Infections/metabolism
2.
Microb Pathog ; 195: 106901, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39218378

ABSTRACT

Neurotropic viruses, characterized by their capacity to invade the central nervous system, present a considerable challenge to public health and are responsible for a diverse range of neurological disorders. This group includes a diverse array of viruses, such as herpes simplex virus, varicella zoster virus, poliovirus, enterovirus and Japanese encephalitis virus, among others. Some of these viruses exhibit high neuroinvasiveness and neurovirulence, while others demonstrate weaker neuroinvasive and neurovirulent properties. The clinical manifestations of infections caused by neurotropic viruses can vary significantly, ranging from mild symptoms to severe life-threatening conditions. Extracellular vesicles (EVs) have garnered considerable attention due to their pivotal role in intracellular communication, which modulates the biological activity of target cells via the transport of biomolecules in both health and disease. Investigating EVs in the context of virus infection is crucial for elucidating their potential role contribution to viral pathogenesis. This is because EVs derived from virus-infected cells frequently transfer viral components to uninfected cells. Importantly, EVs released by virus-infected cells have the capacity to traverse the blood-brain barrier (BBB), thereby impacting neuronal activity and inducing neuroinflammation. In this review, we explore the roles of EVs during neurotropic virus infections in either enhancing or inhibiting viral pathogenesis. We will delve into our current comprehension of the molecular mechanisms that underpin these roles, the potential implications for the infected host, and the prospective diagnostic applications that could arise from this understanding.


Subject(s)
Blood-Brain Barrier , Extracellular Vesicles , Extracellular Vesicles/virology , Extracellular Vesicles/metabolism , Humans , Blood-Brain Barrier/virology , Animals , Viruses/pathogenicity , Viruses/classification , Virus Diseases/virology , Encephalitis Virus, Japanese/pathogenicity , Encephalitis Virus, Japanese/physiology , Herpesvirus 3, Human/pathogenicity , Herpesvirus 3, Human/physiology , Enterovirus/pathogenicity , Enterovirus/physiology
3.
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
4.
Viruses ; 16(8)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39205256

ABSTRACT

Rhinovirus C (RV-C) infects airway epithelial cells and is an important cause of acute respiratory disease in humans. To interrogate the mechanisms of RV-C-mediated disease, animal models are essential. Towards this, RV-C infection was recently reported in wild-type (WT) mice, yet, titers were not sustained. Therefore, the requirements for RV-C infection in mice remain unclear. Notably, prior work has implicated human cadherin-related family member 3 (CDHR3) and stimulator of interferon genes (STING) as essential host factors for virus uptake and replication, respectively. Here, we report that even though human (h) and murine (m) CDHR3 orthologs have similar tissue distribution, amino acid sequence homology is limited. Further, while RV-C can replicate in mouse lung epithelial type 1 (LET1) cells and produce infectious virus, we observed a significant increase in the frequency and intensity of dsRNA-positive cells following hSTING expression. Based on these findings, we sought to assess the impact of hCDHR3 and hSTING on RV-C infection in mice in vivo. Thus, we developed hCDHR3 transgenic mice, and utilized adeno-associated virus (AAV) to deliver hSTING to the murine airways. Subsequent challenge of these mice with RV-C15 revealed significantly higher titers 24 h post-infection in mice expressing both hCDHR3 and hSTING-compared to either WT mice, or mice with hCDHR3 or hSTING alone, indicating more efficient infection. Ultimately, this mouse model can be further engineered to establish a robust in vivo model, recapitulating viral dynamics and disease.


Subject(s)
Cadherin Related Proteins , Cadherins , Mice, Transgenic , Virus Replication , Animals , Mice , Humans , Cadherins/genetics , Cadherins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Epithelial Cells/virology , Disease Models, Animal , Enterovirus/physiology , Enterovirus/genetics , Cell Line , Picornaviridae Infections/virology , Mice, Inbred C57BL , Lung/virology
5.
Commun Biol ; 7(1): 969, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122806

ABSTRACT

Serine proteases are important environmental contributors of enterovirus biocontrol. However, the structural features of molecular interaction accounting for the susceptibility of enteroviruses to proteases remains unexplained. Here, we describe the molecular mechanisms involved in the recruitment of serine proteases to viral capsids. Among the virus types used, coxsackievirus A9 (CVA9), but not CVB5 and echovirus 11 (E11), was inactivated by Subtilisin A in a host-independent manner, while Bovine Pancreatic Trypsin (BPT) only reduced CVA9 infectivity in a host-dependent manner. Predictive interaction models of each protease with capsid protomers indicate the main targets as internal disordered protein (IDP) segments exposed either on the 5-fold vertex (DE loop VP1) or at the 5/2-fold intersection (C-terminal end VP1) of viral capsids. We further show that a functional binding protease/capsid depends on both the strength and the evolution over time of protease-VP1 complexes, and lastly on the local adaptation of proteases on surrounding viral regions. Finally, we predicted three residues on CVA9 capsid that trigger cleavage by Subtilisin A, one of which may act as a sensor residue contributing to enzyme recognition on the DE loop. Overall, this study describes an important biological mechanism involved in enteroviruses biocontrol.


Subject(s)
Capsid Proteins , Capsid , Serine Proteases , Capsid/metabolism , Serine Proteases/metabolism , Serine Proteases/chemistry , Serine Proteases/genetics , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Humans , Enterovirus/enzymology , Enterovirus/physiology , Animals , Enterovirus B, Human/physiology , Enterovirus B, Human/enzymology
6.
Viruses ; 16(7)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-39066172

ABSTRACT

Enterovirus genomic replication initiates at a predicted RNA cloverleaf (5'CL) at the 5' end of the RNA genome. The 5'CL contains one stem (SA) and three stem-loops (SLB, SLC, SLD). Here, we present an analysis of 5'CL conservation and divergence for 209 human health-related serotypes from the enterovirus genus, including enterovirus and rhinovirus species. Phylogenetic analysis indicates six distinct 5'CL serotypes that only partially correlate with the species definition. Additional findings include that 5'CL sequence conservation is higher between the EV species than between the RV species, the 5'CL of EVA and EVB are nearly identical, and RVC has the lowest 5'CL conservation. Regions of high conservation throughout all species include SA and the loop and nearby bases of SLB, which is consistent with known protein interactions at these sites. In addition to the known protein binding site for the Poly-C binding protein in the loop of SLB, other conserved consecutive cytosines in the stems of SLB and SLC provide additional potential interaction sites that have not yet been explored. Other sites of conservation, including the predicted bulge of SLD and other conserved stem, loop, and junction regions, are more difficult to explain and suggest additional interactions or structural requirements that are not yet fully understood. This more intricate understanding of sequence and structure conservation and variability in the 5'CL may assist in the development of broad-spectrum antivirals against a wide range of enteroviruses, while better defining the range of virus isotypes expected to be affected by a particular antiviral.


Subject(s)
Antiviral Agents , Enterovirus , Phylogeny , RNA, Viral , Virus Replication , Virus Replication/drug effects , Antiviral Agents/pharmacology , Enterovirus/genetics , Enterovirus/drug effects , Enterovirus/classification , Enterovirus/physiology , Humans , RNA, Viral/genetics , Nucleic Acid Conformation , Conserved Sequence , 5' Untranslated Regions , Genome, Viral
7.
J Virol ; 98(7): e0081324, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38904364

ABSTRACT

Enteroviruses are single-stranded, positive-sense RNA viruses causing endoplasmic reticulum (ER) stress to induce or modulate downstream signaling pathways known as the unfolded protein responses (UPR). However, viral and host factors involved in the UPR related to viral pathogenesis remain unclear. In the present study, we aimed to identify the major regulator of enterovirus-induced UPR and elucidate the underlying molecular mechanisms. We showed that host Golgi-specific brefeldin A-resistant guanine nucleotide exchange factor 1 (GBF1), which supports enteroviruses replication, was a major regulator of the UPR caused by infection with enteroviruses. In addition, we found that severe UPR was induced by the expression of 3A proteins encoded in human pathogenic enteroviruses, such as enterovirus A71, coxsackievirus B3, poliovirus, and enterovirus D68. The N-terminal-conserved residues of 3A protein interact with the GBF1 and induce UPR through inhibition of ADP-ribosylation factor 1 (ARF1) activation via GBF1 sequestration. Remodeling and expansion of ER and accumulation of ER-resident proteins were observed in cells infected with enteroviruses. Finally, 3A induced apoptosis in cells infected with enteroviruses via activation of the protein kinase RNA-like endoplasmic reticulum kinase (PERK)/C/EBP homologous protein (CHOP) pathway of UPR. Pharmaceutical inhibition of PERK suppressed the cell death caused by infection with enteroviruses, suggesting the UPR pathway is a therapeutic target for treating diseases caused by infection with enteroviruses.IMPORTANCEInfection caused by several plus-stranded RNA viruses leads to dysregulated ER homeostasis in the host cells. The mechanisms underlying the disruption and impairment of ER homeostasis and its significance in pathogenesis upon enteroviral infection remain unclear. Our findings suggested that the 3A protein encoded in human pathogenic enteroviruses disrupts ER homeostasis by interacting with GBF1, a major regulator of UPR. Enterovirus-mediated infections drive ER into pathogenic conditions, where ER-resident proteins are accumulated. Furthermore, in such scenarios, the PERK/CHOP signaling pathway induced by an unresolved imbalance of ER homeostasis essentially drives apoptosis. Therefore, elucidating the mechanisms underlying the virus-induced disruption of ER homeostasis might be a potential target to mitigate the pathogenesis of enteroviruses.


Subject(s)
Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Guanine Nucleotide Exchange Factors , Homeostasis , Unfolded Protein Response , Humans , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Guanine Nucleotide Exchange Factors/metabolism , Guanine Nucleotide Exchange Factors/genetics , Enterovirus Infections/virology , Enterovirus Infections/metabolism , Apoptosis , Enterovirus/physiology , Enterovirus/metabolism , HeLa Cells , Virus Replication , ADP-Ribosylation Factor 1/metabolism , ADP-Ribosylation Factor 1/genetics , HEK293 Cells , Host-Pathogen Interactions , Signal Transduction , eIF-2 Kinase/metabolism
8.
Virus Res ; 345: 199386, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705479

ABSTRACT

Coxsackievirus A16 (CV-A16) and coxsackievirus A10 (CV-A10), more commonly etiological agents of hand, foot and mouth disease (HFMD), are capable of causing severe neurological syndromes with high fatalities, but their neuropathogenesis has rarely been studied. Mounting evidence indicated that pyroptosis is an inflammatory form of cell death that might be widely involved in the pathogenic mechanisms of neurotropic viruses. Our study was designed to examine the effects of NLRP3-mediated pyroptosis in CV-A16- and CV-A10-induced inflammatory neuropathologic formation. In this work, it was showed that SH-SY5Y cells were susceptible to CV-A16 and CV-A10, and meanwhile their infections could result in a decreasing cell viability and an increasing LDH release as well as Caspase1 activation. Moreover, CV-A16 and CV-A10 infections triggered NLRP3-mediated pyroptosis and promoted the release of inflammatory cytokines. Additionally, activated NLRP3 accelerated the pyroptosis formation and aggravated the inflammatory response, but inhibited NLRP3 had a dampening effect on the above situation. Finally, it was further revealed that NLRP3 agonist enhanced the viral replication, but NLRP3 inhibitor suppressed the viral replication, suggesting that NLRP3-driven pyroptosis might support CV-A16 and CV-A10 production in SH-SY5Y cells. Together, our findings demonstrated a mechanism by which CV-A16 and CV-A10 induce inflammatory responses by evoking NLRP3 inflammasome-regulated pyroptosis, which in turn further stimulated the viral replication, providing novel insights into the pathogenesis of CV-A16 and CV-A10 infections.


Subject(s)
NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Virus Replication , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Cytokines/metabolism , Cytokines/genetics , Inflammation/virology , Enterovirus/physiology , Enterovirus/pathogenicity , Cell Line, Tumor , Inflammasomes/metabolism , Enterovirus A, Human/physiology , Enterovirus A, Human/pathogenicity , Cell Survival
9.
J Virol ; 98(6): e0026824, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38775480

ABSTRACT

Enteroviruses are the causative agents associated with several human and animal diseases, posing a significant threat to human and animal health. As one of the host immune defense strategies, innate immunity plays a crucial role in defending against invading pathogens, where the host utilizes a variety of mechanisms to inhibit or eliminate the pathogen. Here, we report a new strategy for the host to repress enterovirus replication by the 78 kDa glucose-regulated protein (GRP78), also known as heat shock protein family A member 5 (HSPA5). The GRP78 recognizes the EV-encoded RNA-dependent RNA polymerases (RdRPs) 3D protein and interacts with the nuclear factor kappa B kinase complex (CHUK) and subunit beta gene (IKBKB) to facilitate the phosphorylation and nuclear translocation of NF-κB, which induces the production of inflammatory factors and leads to a broad inhibition of enterovirus replication. These findings demonstrate a new role of GRP78 in regulating host innate immunity in response to viral infection and provide new insights into the mechanism underlying enterovirus replication and NF-κB activation.IMPORTANCEGRP78 is known as a molecular chaperone for protein folding and plays a critical role in maintaining protein folding and participating in cell proliferation, cell survival, apoptosis, and metabolism. However, the functions of GRP78 to participate in enterovirus genome replication and innate immune responses are rarely documented. In this study, we explored the functions of the EV-3D-interacting protein GRP78 and found that GRP78 inhibits enterovirus replication by activating NF-κB through binding to EV-F 3D and interacting with the NF-κB signaling molecules CHUK/IKBKB. This is the first report that GRP78 interacts with CHUK/IKBKB to activate the NF-κB signaling pathway, which leads to the expression of the proinflammatory cytokines and inhibition of enterovirus replication. These results demonstrate a unique mechanism of virus replication regulation by GRP78 and provide insights into the prevention and treatment of viral infections.


Subject(s)
Endoplasmic Reticulum Chaperone BiP , I-kappa B Kinase , NF-kappa B , Viral Proteins , Virus Replication , Animals , Humans , Chlorocebus aethiops , Endoplasmic Reticulum Chaperone BiP/metabolism , Enterovirus/growth & development , Enterovirus/immunology , Enterovirus/metabolism , Enterovirus/physiology , Enterovirus Infections/virology , Enterovirus Infections/metabolism , Enterovirus Infections/immunology , Heat-Shock Proteins/metabolism , HEK293 Cells , Host-Pathogen Interactions/immunology , I-kappa B Kinase/metabolism , Immunity, Innate , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Phosphorylation , Protein Binding , RNA-Dependent RNA Polymerase/metabolism , Signal Transduction , Vero Cells , Viral Proteins/metabolism
10.
Virus Res ; 345: 199388, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38714218

ABSTRACT

Human enteroviruses (EVs) represent a global public health concern due to their association with a range of serious pediatric illnesses. Despite the high morbidity and mortality exerted by EVs, no broad-spectrum antivirals are currently available. Herein, we presented evidence that doxycycline can inhibit in vitro replication of various neurotropic EVs, including enterovirus A71 (EV-A71), enterovirus D68 (EV-D68), and coxsackievirus (CV)-A6, in a dose-dependent manner. Further investigations indicated that the drug primarily acted at the post-entry stage of virus infection in vitro, with inhibitory effects reaching up to 89 % for EV-A71 when administered two hours post-infection. These findings provide valuable insights for the development of antiviral drugs against EV infections.


Subject(s)
Antiviral Agents , Doxycycline , Enterovirus , Virus Replication , Humans , Doxycycline/pharmacology , Virus Replication/drug effects , Antiviral Agents/pharmacology , Enterovirus/drug effects , Enterovirus/physiology , Enterovirus Infections/virology , Enterovirus Infections/drug therapy , Enterovirus A, Human/drug effects , Enterovirus A, Human/physiology , Cell Line , Enterovirus D, Human/drug effects , Enterovirus D, Human/physiology , Animals , Virus Internalization/drug effects
11.
Indian J Gastroenterol ; 43(2): 475-484, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38460057

ABSTRACT

BACKGROUND: Sepsis remains a global health burden associated with significant morbidity and mortality. Bacteria are known to be the predominant pathogens in sepsis; however, viral etiologies in sepsis are still under diagnosed. Respiratory viral pathogens have been previously linked to sepsis, but the knowledge of incidence, disease burden and mortality of viral-induced sepsis remains limited. This study aimed at understanding the role of respiratory viral infections in the causation of sepsis in liver disease patients. METHODS: In this retrospective study, the clinical records of liver disease patients with influenza-like illness, whose requests for respiratory viral testing were received from January 2019 to December 2022, were reviewed. Respiratory viruses were identified using FilmArray 2.0 respiratory panel (BioFire Diagnostics, Utah, USA). RESULTS: Of 1391 patients tested, a respiratory viral etiology was detected in 23%. The occurrence of sepsis was seen in 35%. Among these, isolated viral etiology with no other bacterial/fungal coinfection was found in 55% of patients. Rhinovirus/Enterovirus was found as the most common underlying viral etiology (23.4%). The sepsis prevalence was higher among patients with associated comorbidities (45%) and decompensated cirrhosis (84%). On multi-variable analysis, no factor was found independently associated with sepsis-related mortality. CONCLUSION: This study underlines the importance of isolated viral etiology in causation of sepsis among liver disease patients. Patients with comorbidities, older age and decompensated cirrhosis are at an increased risk of developing sepsis and are associated with poorer outcomes. Accurate and timely identification of the viral etiology in sepsis would prevent the misuse of antibiotics and improve overall patient care.


Subject(s)
Liver Diseases , Respiratory Tract Infections , Sepsis , Virus Diseases , Humans , Liver Diseases/complications , Respiratory Tract Infections/virology , Sepsis/mortality , Sepsis/virology , Retrospective Studies , Male , Female , Adult , Middle Aged , Comorbidity , Virus Diseases/complications , Rhinovirus/isolation & purification , Rhinovirus/physiology , Enterovirus/isolation & purification , Enterovirus/physiology
12.
PLoS Pathog ; 20(3): e1012036, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38457376

ABSTRACT

Viruses actively reprogram the metabolism of the host to ensure the availability of sufficient building blocks for virus replication and spreading. However, relatively little is known about how picornaviruses-a large family of small, non-enveloped positive-strand RNA viruses-modulate cellular metabolism for their own benefit. Here, we studied the modulation of host metabolism by coxsackievirus B3 (CVB3), a member of the enterovirus genus, and encephalomyocarditis virus (EMCV), a member of the cardiovirus genus, using steady-state as well as 13C-glucose tracing metabolomics. We demonstrate that both CVB3 and EMCV increase the levels of pyrimidine and purine metabolites and provide evidence that this increase is mediated through degradation of nucleic acids and nucleotide recycling, rather than upregulation of de novo synthesis. Finally, by integrating our metabolomics data with a previously acquired phosphoproteomics dataset of CVB3-infected cells, we identify alterations in phosphorylation status of key enzymes involved in nucleotide metabolism, providing insight into the regulation of nucleotide metabolism during infection.


Subject(s)
Cardiovirus , Enterovirus Infections , Enterovirus , Picornaviridae , Humans , Enterovirus/physiology , Encephalomyocarditis virus/physiology , Virus Replication , Enterovirus B, Human/physiology , HeLa Cells
13.
Curr Cancer Drug Targets ; 24(9): 967-974, 2024.
Article in English | MEDLINE | ID: mdl-38310465

ABSTRACT

BACKGROUND: Cancer is a major cause of death worldwide. Colorectal cancer is the second most common type. Additional treatments like chemotherapy and radiation therapy may be recommended. Developing new techniques is vital due to drug resistance and a lack of targeted therapies. OBJECTIVE: In this study, the effects of mesenchymal stem cells (MSCs) loaded with oncolytic Coxsackievirus A21 (CVA21) on a mouse model of CRC were investigated. METHODS: The therapeutic potency of MSCs loaded with oncolytic CVA21 were evaluated in an experimental mouse model of colorectal cancer which received an injection CT26 cells per mouse subcutaneously. Splenocyte proliferation index, lactate dehydrogenase (LDH) assay, nitric oxide (NO) production assessment, and cytokine assay (IFN-γ, IL-4, IL-10, and TGF-ß) in the splenocyte supernatant were all used to evaluate the impact of MSCs loaded with CVA21. RESULTS: The results of this study showed that the treatment of a mouse model of colorectal cancer with MSCs loaded with oncolytic CVA21 could significantly suppress the tumor growth, which was accompanied by stimulation of splenocytes proliferation index, an increase of NO and LDH. Also, MSCs loaded with oncolytic CVA21 increased the secretion of IFN-γ and decreased the secretion of IL-4, IL-10, and TGF-ß. CONCLUSION: The results of the current study suggest that MSCs loaded with oncolytic CVA21 therapy for the CRC mouse model may have some potential advantages. On the other hand, the results of the study showed that, in addition to activating the acquired immune system, the use of MSCs loaded with oncolytic CVA21 also stimulates the innate immune system by increasing level of nitric oxide.


Subject(s)
Colorectal Neoplasms , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals , Colorectal Neoplasms/therapy , Colorectal Neoplasms/pathology , Mice , Mesenchymal Stem Cell Transplantation/methods , Mice, Inbred BALB C , Disease Models, Animal , Oncolytic Virotherapy/methods , Cell Proliferation , Oncolytic Viruses/physiology , Humans , Cell Line, Tumor , Cytokines/metabolism , Enterovirus/physiology , Female
14.
J Virol ; 98(2): e0135823, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38226810

ABSTRACT

Hand, foot, and mouth disease (HFMD) is caused by more than 20 pathogenic enteroviruses belonging to the Picornaviridae family and Enterovirus genus. Since the introduction of the enterovirus-71 (EV71) vaccine in 2016, the number of HFMD cases caused by EV71 has decreased. However, cases of infections caused by other enteroviruses, such as coxsackievirus A6 (CA6) and coxsackievirus A10, have been increasing accordingly. In this study, we used a clinical isolate of CA6 to establish an intragastric infection mouse model using 7-day-old mice to mimic the natural transmission route, by which we investigated the differential gene expression profiles associated with virus infection and pathogenicity. After intragastric infection, mice exhibited hind limb paralysis symptoms and weight loss, similar to those reported for EV71 infection in mice. The skeletal muscle was identified as the main site of virus replication, with a peak viral load reaching 2.31 × 107 copies/mg at 5 dpi and increased infiltration of inflammatory cells. RNA sequencing analysis identified differentially expressed genes (DEGs) after CA6 infection. DEGs in the blood, muscle, brain, spleen, and thymus were predominantly enriched in immune system responses, including pathways such as Toll-like receptor signaling and PI3K-Akt signaling. Our study has unveiled the genes involved in the host immune response during CA6 infection, thereby enhancing our comprehension of the pathological mechanism of HFMD.IMPORTANCEThis study holds great significance for the field of hand, foot, and mouth disease (HFMD). It not only delves into the disease's etiology, transmission pathways, and severe complications but also establishes a novel mouse model that mimics the natural coxsackievirus A6 infection process, providing a pivotal platform to delve deeper into virus replication and pathogenic mechanisms. Additionally, utilizing RNA-seq technology, it unveils the dynamic gene expression changes during infection, offering valuable leads for identifying novel therapeutic drug targets. This research has the potential to enhance our understanding of HFMD, offering fresh perspectives for disease prevention and treatment and positively impacting children's health worldwide.


Subject(s)
Enterovirus Infections , Enterovirus , Hand, Foot and Mouth Disease , Animals , Child , Humans , Mice , Antibodies, Viral , Disease Models, Animal , Enterovirus/pathogenicity , Enterovirus/physiology , Enterovirus A, Human , Enterovirus Infections/pathology , Enterovirus Infections/virology , Gene Expression , Hand, Foot and Mouth Disease/genetics , Phosphatidylinositol 3-Kinases , Virulence
15.
Virology ; 591: 109989, 2024 03.
Article in English | MEDLINE | ID: mdl-38219371

ABSTRACT

Enteroviruses (EVs), comprise a genus in the Picornaviridae family, which have been shown to be neurotropic and can cause various neurological disorders or long-term neurological condition, placing a huge burden on society and families. The blood-brain barrier (BBB) is a protective barrier that prevents dangerous substances from entering the central nervous system (CNS). Recently, numerous EVs have been demonstrated to have the ability to disrupt BBB, and further lead to severe neurological damage. However, the precise mechanisms of BBB disruption associated with these EVs remain largely unknown. In this Review, we focus on the molecular mechanisms of BBB dysfunction caused by EVs, emphasizing the invasiveness of enterovirus A71 (EVA71), which will provide a research direction for further treatment and prevention of CNS disorders.


Subject(s)
Enterovirus Infections , Enterovirus , Humans , Blood-Brain Barrier , Enterovirus/physiology , Central Nervous System , Biological Transport
16.
J Virol ; 97(12): e0092823, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38047713

ABSTRACT

IMPORTANCE: Most protease-targeted antiviral development evaluates the ability of small molecules to inhibit the cleavage of artificial substrates. However, before they can cleave any other substrates, viral proteases need to cleave themselves out of the viral polyprotein in which they have been translated. This can occur either intra- or inter-molecularly. Whether this process occurs intra- or inter-molecularly has implications for the potential for precursors to accumulate and for the effectiveness of antiviral drugs. We argue that evaluating candidate antivirals for their ability to block these cleavages is vital to drug development because the buildup of uncleaved precursors can be inhibitory to the virus and potentially suppress the selection of drug-resistant variants.


Subject(s)
Antiviral Agents , Enterovirus , Viral Protease Inhibitors , Viral Proteases , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Proteolysis , Viral Proteases/metabolism , Viral Protease Inhibitors/pharmacology , Enterovirus/drug effects , Enterovirus/physiology , Polyproteins/metabolism
17.
Elife ; 122023 10 18.
Article in English | MEDLINE | ID: mdl-37850626

ABSTRACT

Enterovirus D68 (EV-D68) is a re-emerging enterovirus that causes acute respiratory illness in infants and has recently been linked to Acute Flaccid Myelitis. Here, we show that the histone deacetylase, SIRT-1, is essential for autophagy and EV-D68 infection. Knockdown of SIRT-1 inhibits autophagy and reduces EV-D68 extracellular titers. The proviral activity of SIRT-1 does not require its deacetylase activity or functional autophagy. SIRT-1's proviral activity is, we demonstrate, mediated through the repression of endoplasmic reticulum stress (ER stress). Inducing ER stress through thapsigargin treatment or SERCA2A knockdown in SIRT-1 knockdown cells had no additional effect on EV-D68 extracellular titers. Knockdown of SIRT-1 also decreases poliovirus and SARS-CoV-2 titers but not coxsackievirus B3. In non-lytic conditions, EV-D68 is primarily released in an enveloped form, and SIRT-1 is required for this process. Our data show that SIRT-1, through its translocation to the cytosol, is critical to promote the release of enveloped EV-D68 viral particles.


Subject(s)
Enterovirus D, Human , Enterovirus Infections , Sirtuin 1 , Virus Activation , Humans , COVID-19 , Enterovirus/genetics , Enterovirus/physiology , Enterovirus D, Human/genetics , Enterovirus D, Human/physiology , Enterovirus Infections/genetics , Enterovirus Infections/physiopathology , Neuromuscular Diseases , Proviruses , SARS-CoV-2 , Viral Envelope/metabolism , Viral Envelope/physiology , Virus Activation/genetics , Virus Activation/physiology , Sirtuin 1/genetics , Sirtuin 1/physiology
18.
Virol J ; 20(1): 178, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37559147

ABSTRACT

Coxsackievirus A16 (CV-A16) is still an important pathogen that causes hand, foot and mouth disease (HFMD) in young children and infants worldwide. Previous studies indicated that CV-A16 infection is usually mild or self-limiting, but it was also found that CV-A16 infection can trigger severe neurological complications and even death. However, there are currently no vaccines or antiviral compounds available to either prevent or treat CV-A16 infection. Therefore, investigation of the virus‒host interaction and identification of host proteins that play a crucial regulatory role in the pathogenesis of CV-A16 infection may provide a novel strategy to develop antiviral drugs. Here, to increase our understanding of the interaction of CV-A16 with the host cell, we analyzed changes in the proteome of 16HBE cells in response to CV-A16 using tandem mass tag (TMT) in combination with LC‒MS/MS. There were 6615 proteins quantified, and 172 proteins showed a significant alteration during CV-A16 infection. These differentially regulated proteins were involved in fundamental biological processes and signaling pathways, including metabolic processes, cytokine‒cytokine receptor interactions, B-cell receptor signaling pathways, and neuroactive ligand‒receptor interactions. Further bioinformatics analysis revealed the characteristics of the protein domains and subcellular localization of these differentially expressed proteins. Then, to validate the proteomics data, 3 randomly selected proteins exhibited consistent changes in protein expression with the TMT results using Western blotting and immunofluorescence methods. Finally, among these differentially regulated proteins, we primarily focused on HMGB1 based on its potential effects on viral replication and virus infection-induced inflammatory responses. It was demonstrated that overexpression of HMGB1 could decrease viral replication and upregulate the release of inflammatory cytokines, but deletion of HMGB1 increased viral replication and downregulated the release of inflammatory cytokines. In conclusion, the results from this study have helped further elucidate the potential molecular pathogenesis of CV-A16 based on numerous protein changes and the functions of HMGB1 Found to be involved in the processes of viral replication and inflammatory response, which may facilitate the development of new antiviral therapies as well as innovative diagnostic methods.


Subject(s)
Enterovirus , HMGB1 Protein , Virus Replication , Humans , Chromatography, Liquid , Cytokines/metabolism , Enterovirus/physiology , Hand, Foot and Mouth Disease , HMGB1 Protein/metabolism , Proteomics , Tandem Mass Spectrometry , Cell Line
19.
Nucleic Acids Res ; 51(16): 8850-8863, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37486760

ABSTRACT

The genomes of positive-strand RNA viruses serve as a template for both protein translation and genome replication. In enteroviruses, a cloverleaf RNA structure at the 5' end of the genome functions as a switch to transition from viral translation to replication by interacting with host poly(C)-binding protein 2 (PCBP2) and the viral 3CDpro protein. We determined the structures of cloverleaf RNA from coxsackievirus and poliovirus. Cloverleaf RNA folds into an H-type four-way junction and is stabilized by a unique adenosine-cytidine-uridine (A•C-U) base triple involving the conserved pyrimidine mismatch region. The two PCBP2 binding sites are spatially proximal and are located on the opposite end from the 3CDpro binding site on cloverleaf. We determined that the A•C-U base triple restricts the flexibility of the cloverleaf stem-loops resulting in partial occlusion of the PCBP2 binding site, and elimination of the A•C-U base triple increases the binding affinity of PCBP2 to the cloverleaf RNA. Based on the cloverleaf structures and biophysical assays, we propose a new mechanistic model by which enteroviruses use the cloverleaf structure as a molecular switch to transition from viral protein translation to genome replication.


Subject(s)
Enterovirus , Genome, Viral , Poliovirus , RNA, Viral , Humans , Enterovirus/genetics , Enterovirus/physiology , HeLa Cells , Nucleic Acid Conformation , Poliovirus/genetics , Poliovirus/physiology , Protein Biosynthesis , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/genetics
20.
Adv Biol (Weinh) ; 7(7): e2200336, 2023 07.
Article in English | MEDLINE | ID: mdl-37132155

ABSTRACT

Colorectal cancer (CRC) is an intestinal malignant tumor with high morbidity and mortality worldwide. Inoperability or resistanance to radiation and chemotherapy occur in the conventional treatments against CRC. Oncolytic viruses (OVs) are one kind of virus that selectively infects and lyses cancer cells, which is considered to be a new anticancer therapy with biological and immune-based approaches. Enterovirus 71 (EV71), belonging to the enterovirus genus in the family Picornaviridae, is a single positive-stranded RNA virus. EV71 is transmitted in a fetal-oral route and infects gastrointestinal tract in infants. Here, EV71 is exploited to be a novel oncolytic virus in colorectal cancer. It is revealed that EV71 infection can selectively cause colorectal cancer cells cytotoxicity but not primary intestinal epithelial cells. Consistently, EV71 injection significantly inhibits tumor growth in nude mice xenografted colorectal cancer cells. In detail, EV71 infects colorectal cancer cells to repress the expression of Ki67 and B-cell leukemia 2 (Bcl-2) leading to the inhibition of cell proliferation, while activating the cleavage of poly-adenosine diphosphatase-ribose polymerase and Caspase-3 protein resulting in the promotion of cell apoptosis. The findings demonstrate the oncolytic feature of EV71 in CRC treatment and may provide a potential clue for clinical anticancer therapy.


Subject(s)
Colorectal Neoplasms , Enterovirus A, Human , Enterovirus Infections , Enterovirus , Oncolytic Viruses , Mice , Animals , Enterovirus A, Human/genetics , Mice, Nude , Enterovirus/physiology , Enterovirus Infections/therapy , Colorectal Neoplasms/therapy
SELECTION OF CITATIONS
SEARCH DETAIL