Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 477
Filter
1.
PLoS Negl Trop Dis ; 18(7): e0012008, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949988

ABSTRACT

BACKGROUND: Hand, foot, and mouth disease (HFMD) is a major public health issue in China while temperature and humidity are well-documented predictors. However, evidence on the combined effect of temperature and humidity is still limited. It also remains unclear whether such an effect could be modified by the enterovirus 71 (EV71) vaccination. METHODS: Based on 320,042 reported HFMD cases during the summer months between 2012 and 2019, we conducted a study utilizing Distributed Lag Non-Linear Models (DLNM) and time-varying DLNM to examine how China's HFMD EV71 vaccine strategy would affect the correlation between meteorological conditions and HFMD risk. RESULTS: The incidence of HFMD changed with the Discomfort Index in an arm-shaped form. The 14-day cumulative risk of HFMD exhibited a statistically significant increase during the period of 2017-2019 (following the implementation of the EV71 vaccine policy) compared to 2012-2016 (prior to the vaccine implementation). For the total population, the range of relative risk (RR) values for HFMD at the 75th, 90th, and 99th percentiles increased from 1.082-1.303 in 2012-2016 to 1.836-2.022 in 2017-2019. In the stratified analyses, Han Chinese areas show stronger relative growth, with RR values at the 75th, 90th, and 99th percentiles increased by 14.3%, 39.1%, and 134.4% post-vaccination, compared to increases of 22.7%, 41.6%, and 38.8% in minority areas. Similarly, boys showed greater increases (24.4%, 47.7%, 121.5%) compared to girls (8.1%, 28.1%, 58.3%). Additionally, the central Guizhou urban agglomeration displayed a tendency for stronger relative growth compared to other counties. CONCLUSIONS: Although the EV71 vaccine policy has been implemented, it hasn't effectively controlled the overall risk of HFMD. There's been a shift in the main viral subtypes, potentially altering population susceptibility and influencing HFMD occurrences. The modulating effects of vaccine intervention may also be influenced by factors such as race, sex, and economic level.


Subject(s)
Enterovirus A, Human , Hand, Foot and Mouth Disease , Vaccination , Hand, Foot and Mouth Disease/epidemiology , Hand, Foot and Mouth Disease/prevention & control , Humans , China/epidemiology , Male , Female , Vaccination/statistics & numerical data , Infant , Child, Preschool , Enterovirus A, Human/immunology , Incidence , Viral Vaccines/administration & dosage , Humidity , Temperature , Child
2.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891876

ABSTRACT

Enterovirus A71 (EV-A71) is a major pathogen causing hand, foot, and mouth disease (HFMD) in children worldwide. It can lead to severe gastrointestinal, pulmonary, and neurological complications. The innate immune system, which rapidly detects pathogens via pathogen-associated molecular patterns or pathogen-encoded effectors, serves as the first defensive line against EV-A71 infection. Concurrently, the virus has developed various sophisticated strategies to evade host antiviral responses and establish productive infection. Thus, the virus-host interactions and conflicts, as well as the ability to govern biological events at this first line of defense, contribute significantly to the pathogenesis and outcomes of EV-A71 infection. In this review, we update recent progress on host innate immune responses to EV-A71 infection. In addition, we discuss the underlying strategies employed by EV-A71 to escape host innate immune responses. A better understanding of the interplay between EV-A71 and host innate immunity may unravel potential antiviral targets, as well as strategies that can improve patient outcomes.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Host-Pathogen Interactions , Immune Evasion , Immunity, Innate , Humans , Immune Evasion/immunology , Enterovirus A, Human/immunology , Enterovirus A, Human/pathogenicity , Host-Pathogen Interactions/immunology , Enterovirus Infections/immunology , Enterovirus Infections/virology , Animals , Hand, Foot and Mouth Disease/immunology , Hand, Foot and Mouth Disease/virology
3.
Front Cell Infect Microbiol ; 14: 1393680, 2024.
Article in English | MEDLINE | ID: mdl-38938877

ABSTRACT

Hand, foot, and mouth disease (HFMD) is a common infectious disease caused by enterovirus 71 (EV71) that frequently affects children, leading to severe infections in some cases. In general, when infection occurs, the body upregulates inflammatory responses to eliminate pathogenic microorganisms to protect the host from infection. However, EV71 may inhibit host's innate immunity to promote virus infection. At present, it is not fully understood how EV71 hijack the host cells for its own replication. Toll-like receptor 4 (TLR4), a natural immune receptor, historically associated with bacterial endotoxin-induced inflammatory responses. However, it is still unclear whether and how TLR4 is altered during EV71 infection. In this study, we observed a reduction in both TLR4 protein and gene transcript levels in RD, GES-1, and Vero cells following EV71 infection, as detected by RT-qPCR, immunofluorescence staining and western blot. Furthermore, we observed that the TLR4 downstream molecules of MYD88, p-NF-κB p65, p-TBK1 and related inflammatory cytokines were also reduced, suggesting that antiviral innate immune and inflammatory response were suppressed. To determine the impact of TLR4 changes on EV71 infection, we interfered EV71-infected RD cells with TLR4 agonist or inhibitor and the results showed that activation of TLR4 inhibited EV71 replication, while inhibition of TLR4 promote EV71 replication. Besides, EV71 replication was also promoted in TLR4 siRNA-transfected and EV71-infected RD cells. This suggests that down-regulation the expression of TLR4 by EV71 can inhibit host immune defense to promote EV71 self-replication. This novel mechanism may be a strategy for EV71 to evade host immunity.


Subject(s)
Enterovirus A, Human , Immunity, Innate , Signal Transduction , Toll-Like Receptor 4 , Virus Replication , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Enterovirus A, Human/immunology , Humans , Animals , Vero Cells , Chlorocebus aethiops , Host-Pathogen Interactions/immunology , Inflammation/metabolism , Inflammation/immunology , Myeloid Differentiation Factor 88/metabolism , Myeloid Differentiation Factor 88/genetics , Cell Line , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cytokines/metabolism , NF-kappa B/metabolism , Hand, Foot and Mouth Disease/immunology , Hand, Foot and Mouth Disease/virology
4.
Vaccine ; 42(17): 3733-3743, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38705805

ABSTRACT

Hand, foot, and mouth disease (HFMD) poses a significant public health threat primarily caused by four major enteroviruses: enterovirus 71 (EV71), coxsackieviruses A16, A10, and A6. Broadly protective immune responses are essential for complete protection against these major enteroviruses. In this study, we designed a new tetravalent immunogen for HFMD, validated it in silico, in vivo evaluated the immunogenicity of the DNA-based tetravalent vaccine in mice, and identified immunogenic B-cell and T-cell epitopes. A new tetravalent immunogen, VP1me, was designed based on the chimeric protein and epitope-based vaccine principles. It contains a complete EV71 VP1 protein and six reported neutralizing B-cell epitopes derived from the four major enteroviruses causing HFMD. In silico validation using multiple immunoinformatic tools indicated good attributes of the VP1me immunogen suitable for vaccine development. The VP1me-based DNA vaccine efficiently induced both humoral and cellular immune responses in BALB/cAJcl mice. A combination of in silico prediction and immunoassays enabled the identification of immunogenic linear B-cell and CD8 T-cell epitopes within the VP1me immunogen. Immunodominant linear B-cell epitopes were identified in six regions of VP1me, with one epitope located at the N-terminus of the VP1 protein (aa 9-23) regarded as a novel epitope. Interestingly, some B-cell epitopes could also induce the CD8 T-cell response, suggesting their dual functions in immune stimulation. These results lay the groundwork for further development of VP1me as a new vaccine candidate.


Subject(s)
Antibodies, Viral , Epitopes, B-Lymphocyte , Hand, Foot and Mouth Disease , Immunodominant Epitopes , Mice, Inbred BALB C , Vaccines, DNA , Viral Vaccines , Animals , Vaccines, DNA/immunology , Epitopes, B-Lymphocyte/immunology , Hand, Foot and Mouth Disease/prevention & control , Hand, Foot and Mouth Disease/immunology , Mice , Viral Vaccines/immunology , Immunodominant Epitopes/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Female , Epitopes, T-Lymphocyte/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Enterovirus/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Enterovirus A, Human/immunology , Enterovirus A, Human/genetics , Immunogenicity, Vaccine , Immunity, Cellular , Immunity, Humoral
5.
Viruses ; 16(4)2024 04 08.
Article in English | MEDLINE | ID: mdl-38675915

ABSTRACT

The enterovirus A71 (EV71) inactivated vaccine is an effective intervention to control the spread of the virus and prevent EV71-associated hand, foot, and mouth disease (HFMD). It is widely administered to infants and children in China. The empty particles (EPs) and full particles (FPs) generated during production have different antigenic and immunogenic properties. However, the antigen detection methods currently used were established without considering the differences in antigenicity between EPs and FPs. There is also a lack of other effective analytical methods for detecting the different particle forms, which hinders the consistency between batches of products. In this study, we analyzed the application of sedimentation velocity analytical ultracentrifugation (SV-AUC) in characterizing the EPs and FPs of EV71. Our results showed that the proportions of the two forms could be quantified simultaneously by SV-AUC. We also determined the repeatability and accuracy of this method and found that both parameters were satisfactory. We assessed SV-AUC for bulk vaccine quality control, and our findings indicated that SV-AUC can be used effectively to analyze the percentage of EPs and FPs and monitor the consistency of the process to ensure the quality of the vaccine.


Subject(s)
Enterovirus A, Human , Ultracentrifugation , Enterovirus A, Human/immunology , Enterovirus A, Human/isolation & purification , Ultracentrifugation/methods , Humans , Viral Vaccines/immunology , Vaccines, Inactivated/immunology , Virion/immunology , Virion/isolation & purification , Hand, Foot and Mouth Disease/virology , Hand, Foot and Mouth Disease/prevention & control , China , Quality Control
7.
J Immunol ; 212(11): 1782-1790, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38629901

ABSTRACT

Enterovirus 71 (EV71) is a significant causative agent of hand, foot, and mouth disease, with potential serious neurologic complications or fatal outcomes. The lack of effective treatments for EV71 infection is attributed to its elusive pathogenicity. Our study reveals that human plasmacytoid dendritic cells (pDCs), the main type I IFN-producing cells, selectively express scavenger receptor class B, member 2 (SCARB2) and P-selectin glycoprotein ligand 1 (PSGL-1), crucial cellular receptors for EV71. Some strains of EV71 can replicate within pDCs and stimulate IFN-α production. The activation of pDCs by EV71 is hindered by Abs to PSGL-1 and soluble PSGL-1, whereas Abs to SCARB2 and soluble SCARB2 have a less pronounced effect. Our data suggest that only strains binding to PSGL-1, more commonly found in severe cases, can replicate in pDCs and induce IFN-α secretion, highlighting the importance of PSGL-1 in these processes. Furthermore, IFN-α secretion by pDCs can be triggered by EV71 or UV-inactivated EV71 virions, indicating that productive infection is not necessary for pDC activation. These findings provide new insights into the interaction between EV71 and pDCs, suggesting that pDC activation could potentially mitigate the severity of EV71-related diseases.


Subject(s)
Dendritic Cells , Enterovirus A, Human , Interferon-alpha , Lysosomal Membrane Proteins , Membrane Glycoproteins , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Enterovirus A, Human/immunology , Enterovirus A, Human/physiology , Membrane Glycoproteins/metabolism , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/immunology , Interferon-alpha/metabolism , Interferon-alpha/immunology , Receptors, Scavenger/metabolism , Enterovirus Infections/immunology , Enterovirus Infections/virology , Virus Replication
8.
Arch Virol ; 167(12): 2483-2501, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36171507

ABSTRACT

Enterovirus A71 (EV-A71) is one of the major pathogens responsible for hand, foot, and mouth disease (HFMD). Many HFMD outbreaks have been reported throughout the world in the past decades. Compared with other viruses, EV-A71 infection is more frequently associated with severe neurological complications and even death in children. EV-A71 can also infect adults and cause severe complications and death, although such cases are very uncommon. Although fatal cases of EV-A71 infection have been reported, the underlying mechanisms of EV-A71 infection, especially the mode of viral spread into the central nervous system (CNS) and mechanisms of pulmonary edema, which is considered to be the direct cause of death, have not yet been fully clarified, and more studies are needed. Here, we first summarize the pathological findings in various systems of patients with fatal EV-A71 infections, focussing in detail on gross changes, histopathological examination, tissue distribution of viral antigens and nucleic acids, systemic inflammatory cell infiltration, and tissue distribution of viral receptors and their co-localization with viral antigens. We then present our conclusions about viral dissemination, neuropathogenesis, and the mechanism of pulmonary edema in EV-A71 infection, based on pathological findings.


Subject(s)
Enterovirus A, Human , Enterovirus Infections , Child , Humans , Antigens, Viral/metabolism , Enterovirus/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/complications , Enterovirus Infections/pathology , Pulmonary Edema/virology
9.
Commun Biol ; 4(1): 663, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34079066

ABSTRACT

The reciprocal interactions between pathogens and hosts are complicated and profound. A comprehensive understanding of these interactions is essential for developing effective therapies against infectious diseases. Interferon responses induced upon virus infection are critical for establishing host antiviral innate immunity. Here, we provide a molecular mechanism wherein isoform switching of the host IKKε gene, an interferon-associated molecule, leads to alterations in IFN production during EV71 infection. We found that IKKε isoform 2 (IKKε v2) is upregulated while IKKε v1 is downregulated in EV71 infection. IKKε v2 interacts with IRF7 and promotes IRF7 activation through phosphorylation and translocation of IRF7 in the presence of ubiquitin, by which the expression of IFNß and ISGs is elicited and virus propagation is attenuated. We also identified that IKKε v2 is activated via K63-linked ubiquitination. Our results suggest that host cells induce IKKε isoform switching and result in IFN production against EV71 infection. This finding highlights a gene regulatory mechanism in pathogen-host interactions and provides a potential strategy for establishing host first-line defense against pathogens.


Subject(s)
Enterovirus A, Human/immunology , Enterovirus A, Human/pathogenicity , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Alternative Splicing , Cell Line , Genes, Switch , HEK293 Cells , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , I-kappa B Kinase/metabolism , Immunity, Innate/genetics , Interferon Regulatory Factor-7/metabolism , Interferon-beta/biosynthesis , Isoenzymes/genetics , Isoenzymes/immunology , Phosphorylation , Ubiquitin/metabolism
10.
Front Immunol ; 12: 665197, 2021.
Article in English | MEDLINE | ID: mdl-34054834

ABSTRACT

Coxsackievirus A6 (CVA6) is recognized as a major enterovirus type that can cause severe hand, foot, and mouth disease and spread widely among children. Vaccines and antiviral drugs may be developed more effectively based on a stable and easy-to-operate CVA6 mouse infection model. In this study, a wild CVA6-W strain was sub-cultured in newborn mice of different ages (in days), for adaptation. Therefore, a CVA6-A mouse-adapted strain capable of stably infecting the mice was generated, and a fatal model was built. As the result indicated, CVA6-A could infect the 10-day-old mice to generate higher levels of IFN-γ, IL-6, and IL-10. The mice infected with CVA6-A were treated with IFN-α1b at a higher dose, with complete protection. Based on this strain, an animal model with active immunization was built to evaluate antiviral protection by active immunization. The three-day-old mice were pre-immunized with inactivated CVA6 thereby generating IgM and IgG antibodies within 7 days that enabled complete protection of the pre-immunized mice following the CVA6 virus challenge. There were eight mutations in the genome of CVA6-A than in that of CVA6-W, possibly attributed to the virulence of CVA6 in mice. Briefly, the CVA6 infection model of the 10-day-old mice built herein, may serve as an applicable preclinical evaluation model for CVA6 antiviral drugs and vaccine study.


Subject(s)
Antibodies, Viral/therapeutic use , Antiviral Agents/therapeutic use , Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/immunology , Hand, Foot and Mouth Disease/prevention & control , Viral Vaccines/immunology , Animals , Animals, Newborn , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Disease Models, Animal , Enterovirus A, Human/drug effects , Enterovirus A, Human/pathogenicity , Hand, Foot and Mouth Disease/drug therapy , Hand, Foot and Mouth Disease/virology , Interferon-gamma/blood , Interferon-gamma/pharmacology , Interleukin-10/blood , Interleukin-10/pharmacology , Interleukin-6/blood , Interleukin-6/pharmacology , Male , Mice , Mice, Inbred BALB C , Vaccination , Vaccines, Inactivated/immunology , Viral Load/drug effects
11.
Emerg Microbes Infect ; 10(1): 1180-1190, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34044752

ABSTRACT

Hand-foot-and-mouth disease is a contagious disease common among children under 5 years old worldwide. It is caused by strains of enterovirus, especially EV-A71, which can lead to severe disease. Vaccines are the only way to fight this disease. Accordingly, it is necessary to establish an efficient and accurate methodology to evaluate vaccine efficacy in vivo. Here, we established a practical method using a hSCARB2 knock-in mouse model, which was susceptible to EV-A71 infection at 5-6 weeks of age, to directly determine the efficacy of vaccines. Unlike traditional approaches, one-week-old hSCARB2 mice were immunized twice with a licensed vaccine, with an interval of a week. The titre of antibodies was measured after 1 week. Mice at 4 weeks of age were challenged with EV-A71 intraperitoneally and intracranially, respectively. The unimmunized hSCARB2 mice displayed systemic clinical symptoms and succumbed to the disease at a rate of approximately 50%. High viral loads were detected in the lungs, brain, and muscles, accompanied by clear pathological changes. The expression of IL-1ß, IL-13, IL-17, and TNF-α was significantly upregulated. By contrast, the immunized group was practically normal and indistinguishable from the control mice. These results indicate that the hSCARB2 knock-in mouse is susceptible to infection in adulthood, and the in vivo efficacy of EV-A71 vaccine could be directly evaluated in this mouse model. The method developed here may be used in the development of new vaccines against HFMD or quality control of licensed vaccines.


Subject(s)
Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/prevention & control , Lysosomal Membrane Proteins/genetics , Receptors, Scavenger/genetics , Vaccines, Inactivated/administration & dosage , Animals , Disease Models, Animal , Enterovirus A, Human/physiology , Gene Knock-In Techniques , Hand, Foot and Mouth Disease/immunology , Humans , Immunization , Mice , Vaccines, Inactivated/immunology , Viral Load , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
12.
EBioMedicine ; 68: 103398, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34049245

ABSTRACT

BACKGROUND: Hand, foot, and mouth disease (HFMD) caused by enterovirus A71 (EV-A71) poses a serious threat to children's health. Kinetics of the neutralising antibody (NAb) response in EV-A71 infected HFMD patients remains unclear. The ideal sampling time of paired serum samples for serological diagnosis of EV-A71 infection is not well defined. METHODS: HFMD inpatients admitted to Henan Children's Hospital between February 15, 2017 and February 15, 2018 were enrolled. Serial serum samples collected during hospitalisation and up to 1.5 years after discharge were tested for NAb against EV-A71. Random intercept modelling with B-spline was conducted to characterize the kinetics of the EV-A71 NAb response over time after illness onset. FINDINGS: A total of 524 serum samples from 264 EV-A71 RNA positive HFMD inpatients were collected. NAb titres of EV-A71 infected patients were estimated to increase from 40 (95% CI: 9-180) at the day of onset to the peak of 2417 (95% CI: 1859-3143) at day 13, then remained above 1240 until 26 months. For serological diagnosis of EV-A71 infection, if at least a 4-fold rise in titre was used as the criteria, the acute phase serum should be collected at 0-4 days, the corresponding convalescent serum should be collected 14.9 days (95% CI: 9.1-23.8) after illness onset. INTERPRETATION: EV-A71 infection induced a strong and persistent humoral immune response in HFMD patients. The findings provide a scientific support for determining the collection time of paired serum samples for serological diagnosis of EV-A71 infected HFMD patients. FUNDING: National Science Fund for Distinguished Young Scholars.


Subject(s)
Antibodies, Neutralizing/blood , Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/immunology , Child , Child, Preschool , China , Enterovirus A, Human/genetics , Female , Humans , Immunity, Humoral , Infant , Inpatients , Longitudinal Studies , Male , Prospective Studies , RNA, Viral/genetics , Time Factors
13.
Viruses ; 13(3)2021 03 09.
Article in English | MEDLINE | ID: mdl-33803356

ABSTRACT

Enterovirus A71 (EV-A71) and coxsackievirus A16 (CA16) are major etiological agents of hand foot and mouth disease (HFMD) in children, which may result in fatal neurological complications. The development of safe, cost effective vaccines against HFMD, especially for use in developing countries, is still a top public health priority. We have successfully generated a stable, cold-adapted, temperature sensitive/conditional lethal EV-A71 through adaptive culturing in Vero cells at incrementally lower cultivation temperatures. An additional 40 passages at an incubation temperature of 28 °C, and a temperature reversion study at an incubation temperature of 37 °C and 39.5 °C, reveals the virus's phenotypic and genetic stability at the predefined culture conditions. Six unique mutations (two in noncoding regions and four in nonstructural protein-coding genes) in combination may have contributed to its stable phenotype and inability to fully revert to its original wild phenotype. The safety and immunogenicity of this stable, cold-adapted, temperature sensitive/conditional lethal EV-A71 was performed in six monkeys. None of the inoculated monkeys developed any obvious clinical illness except one which developed a transient spike of fever. No gross postmortem lesion or abnormal histological finding was noted for all monkeys at autopsy. No virus was reisolated although EV-A71 specific RNA was detected in serum samples collected on both day 4 and day 8 postinoculation. Only EV-A71 RNA and viral antigen were detected in the spleen homogenate and peripheral blood mononuclear cells, respectively, collected on day 4. The two remaining monkeys developed good humoral immune response on day 14 and day 30 post-inoculation.


Subject(s)
Antibodies, Viral/blood , Enterovirus A, Human/genetics , Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/prevention & control , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , Animals , Antigens, Viral/blood , Cell Line , Chlorocebus aethiops , Female , Hand, Foot and Mouth Disease/virology , Macaca fascicularis , Male , RNA, Viral/blood , Vero Cells , Viral Vaccines/adverse effects
14.
Viruses ; 13(5)2021 04 21.
Article in English | MEDLINE | ID: mdl-33919184

ABSTRACT

Enterovirus A-71 (EV71) is a global, highly contagkkious pathogen responsible for severe cases of hand-food-mouth-disease (HFMD). The use of vaccines eliciting cross neutralizing antibodies (NTAbs) against the different circulating EV71 sub-genotypes is important for preventing HFMD outbreaks. Here, we tested the cross-neutralizing activities induced by EV71 genotype/sub-genotype A, B0-B4, C1, C2, C4, and C5 viruses using rats. Differences were noted in the cross-neutralization of the 10 sub-genotypes tested but there were generally good levels of cross-neutralization except against genotype A virus, against which neutralization antibody titres (NTAb) where the lowest with NTAbs being the highest against sub-genotype B4. Moreover, NTAb responses induced by C4, B4, C1, and C2 viruses were homogenous, with values of maximum/minimum NTAb ratios (MAX/MIN) against all B and C viruses ranging between 4.0 and 6.0, whereas MAX/MIN values against B3 and A viruses were highly variable, 48.0 and 256.0, respectively. We then dissected the cross-neutralizing ability of sera from infants and children and rats immunized with C4 EV71 vaccines. Cross-neutralizing titers against the 10 sub-genotypes were good in both vaccinated infants and children and rats with the MAX/MIN ranging from 1.8-3.4 and 5.1-7.1, respectively, which were similar to those found in naturally infected patients (2.8). Therefore, we conclude that C4 EV71 vaccines can provide global protection to infants and children against HFMD caused by different sub-genotypes.


Subject(s)
Antigens, Viral/immunology , Cross Reactions/immunology , Enterovirus A, Human/genetics , Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Enterovirus Infections/virology , Genotype , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Enterovirus Infections/prevention & control , Humans , Immunogenicity, Vaccine , Neutralization Tests , Rats , Vaccines, Inactivated/immunology , Viral Vaccines/immunology
15.
Emerg Microbes Infect ; 10(1): 619-628, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33682641

ABSTRACT

Enterovirus A71 (EV-A71) inactivated vaccines have been widely inoculated among children in Kunming City after it was approved. However, there was a large-scale outbreak of Enteroviruses (EVs) infection in Kunming, 2018. The epidemiological characteristics of HFMD and EVs were analysed during 2008-2018, which are before and three years after EV-A71 vaccine starting to use. The changes in infection spectrum were also investigated, especially for severe HFMD in 2018. The incidence of EV-A71 decreased dramatically after the EV-A71 vaccine starting use. The proportion of non-CV-A16/EV-A71 EVs positive patients raised to 77.17-85.82%, while, EV-A71 and CV-A16 only accounted for 3.41-7.24% and 6.94-19.42% in 2017 and 2018, respectively. CV-A6 was the most important causative agent in all clinical symptoms (severe HFMD, HFMD, Herpangina and fever), accounting from 42.13% to 62.33%. EV-A71 only account for 0.36-2.05%. In severe HFMD, CV-A6 (62.33%), CV-A10 (11.64%), and CV-A16 (10.96%) were the major causative agent in 2018. EV-A71 inactivated vaccine has a good protective effect against EV-A71 and induced EVs infection spectrum changefully. EV-A71 vaccine has no or insignificant cross-protection effect on CV-A6, CV-A10, and CV-A16. Herein, developing 4-valent combined vaccines is urgently needed.


Subject(s)
Enterovirus A, Human/immunology , Enterovirus Infections/epidemiology , Enterovirus Infections/prevention & control , Hand, Foot and Mouth Disease/epidemiology , Hand, Foot and Mouth Disease/prevention & control , Vaccines, Inactivated/immunology , Adolescent , Child , Child, Preschool , China/epidemiology , Disease Outbreaks , Enterovirus A, Human/classification , Enterovirus A, Human/genetics , Enterovirus Infections/immunology , Feces/microbiology , Female , Hand, Foot and Mouth Disease/immunology , Humans , Infant , Infant, Newborn , Inpatients , Male , RNA, Viral , Reverse Transcriptase Polymerase Chain Reaction , Serogroup , Tertiary Care Centers
16.
Emerg Microbes Infect ; 10(1): 763-773, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33739899

ABSTRACT

Coxsackievirus A6 (CV-A6) has been emerging as a major pathogen of hand, foot and mouth disease (HFMD). Study on the pathogenesis of CV-A6 infection and development of vaccines is hindered by a lack of appropriate animal models. Here, we report an actively immunized-challenged mouse model to evaluate the efficacy of a Vero-cell-based, inactivated CV-A6 vaccine candidate. The neonatal Kunming mice were inoculated with a purified, formaldehyde-inactivated CV-A6 vaccine on days 3 and 9, followed by challenging on day 14 with a naturally selected virulent strain at a lethal dose. Within 14 days postchallenge, all mice in the immunized groups survived, while 100% of the Alum-only inoculated mice died. Neutralizing antibodies (NtAbs) were detected in the serum of immunized suckling mice, and the NtAb levels correlated with the survival rate of the challenged mice. The virus loads in organs were reduced, and pathological changes and viral protein expression were weak in the immunized mice compared with those in Alum-only inoculated control mice. Elevated levels of interleukin-4, 6, interferon γ and tumour necrosis factor α were also observed in Alum-only control mice compared with immunized mice. Importantly, the virulent CV-A6 challenge strain was selected quickly and conveniently from a RD cell virus stock characterized with the natural multi-genotypes. The virulent determinants were mapped to V124M and I242 V at VP1. Together, our results indicated that this actively immunized mouse model is invaluable for future studies to develop multivalent vaccines containing the major component of CV-A6 against HFMD.


Subject(s)
Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/virology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chlorocebus aethiops , Disease Models, Animal , Enterovirus A, Human/genetics , Hand, Foot and Mouth Disease/genetics , Hand, Foot and Mouth Disease/immunology , Humans , Immunization , Interleukin-4/genetics , Interleukin-4/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Vero Cells , Viral Vaccines/administration & dosage
17.
Virulence ; 12(1): 704-722, 2021 12.
Article in English | MEDLINE | ID: mdl-33517839

ABSTRACT

Toll-like receptors (TLRs) are essential for the protection of the host from pathogen infections by initiating the integration of contextual cues to regulate inflammation and immunity. However, without tightly controlled immune responses, the host will be subjected to detrimental outcomes. Therefore, it is important to balance the positive and negative regulations of TLRs to eliminate pathogen infection, yet avert harmful immunological consequences. This study revealed a distinct mechanism underlying the regulation of the TLR network. The expression of sex-determining region Y-box 4 (Sox4) is induced by virus infection in viral infected patients and cultured cells, which subsequently represses the TLR signaling network to facilitate viral replication at multiple levels by a distinct mechanism. Briefly, Sox4 inhibits the production of myeloid differentiation primary response gene 88 (MyD88) and most of the TLRs by binding to their promoters to attenuate gene transcription. In addition, Sox4 blocks the activities of the TLR/MyD88/IRAK4/TAK1 and TLR/TRIF/TRAF3/TBK1 pathways by repressing their key components. Moreover, Sox4 represses the activation of the nuclear factor kappa-B (NF-κB) through interacting with IKKα/α, and attenuates NF-kB and IFN regulatory factors 3/7 (IRF3/7) abundances by promoting protein degradation. All these contributed to the down-regulation of interferons (IFNs) and IFN-stimulated gene (ISG) expression, leading to facilitate the viral replications. Therefore, we reveal a distinct mechanism by which viral pathogens evade host innate immunity and discover a key regulator in host defense.


Subject(s)
Immunity, Innate/genetics , SOXC Transcription Factors/genetics , SOXC Transcription Factors/immunology , Signal Transduction/immunology , Toll-Like Receptors/metabolism , Viruses/immunology , Enterovirus A, Human/immunology , Enterovirus A, Human/pathogenicity , Hep G2 Cells , Humans , Immunity, Innate/immunology , Influenza A virus/immunology , Influenza A virus/pathogenicity , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Virus Replication , Viruses/pathogenicity
18.
J Neuroophthalmol ; 41(3): e351-e353, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-33417416

ABSTRACT

ABSTRACT: A 33-year-old man presented with acute painless loss of vision in his right eye after hand-foot-mouth disease (HFMD). Examination confirmed a right optic neuropathy. Neuroimaging and routine evaluations for alternative causes for an optic neuropathy were negative. He was treated with high dose corticosteroids and made an almost complete visual recovery. Postinfectious optic neuritis has been reported after a vast array of infections including: varicella zoster virus, influenza virus, herpes simplex virus, Epstein-Barr Virus, Lyme disease, and many others. Although Coxsackie virus infections are a known cause of HFMD and have been reported to cause maculopathy, to the best of our knowledge, this is the first reported case of optic neuritis after HFMD in the English language ophthalmic literature.


Subject(s)
Antibodies, Viral/analysis , Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/complications , Optic Nerve/diagnostic imaging , Optic Neuritis/etiology , Adult , Follow-Up Studies , Hand, Foot and Mouth Disease/diagnosis , Hand, Foot and Mouth Disease/virology , Humans , Magnetic Resonance Imaging/methods , Male , Optic Neuritis/diagnosis , Time Factors
19.
Virology ; 555: 1-9, 2021 03.
Article in English | MEDLINE | ID: mdl-33418337

ABSTRACT

In this study, we analyzed ten CVA10 strains were genotyped and cultured for 10 generations to detect plaque morphology, pathogenicity, growth and other characteristics. Mice were injected with live and inactivated virus to detect neutralizing antibody titers. The results suggested that all CVA10 strains fell into Genotype C. Each strain cultured on KMB17 and Vero cells, increased from 1st generation onwards to peak in the 3rd and 4th, and the titer at which each became infectious ranged from 5.0 to 6.5 and 6.0 to 7.0 lgCCID50/ml, respectively. Two-day-old BALB/c mice were selected and inoculated intracerebral with the CVA10 strains, Limb paralysis was significant as early as 3 d; paralysis of all limbs for 50% of affected mice. LT50 was approximately 6 d, all died within 8 d. Ten strains induced good immune response, the GMT value of booster immunizations was higher than that of initial immunization. This provide reference points for selecting CVA10 vaccine candidates.


Subject(s)
Enterovirus A, Human , Hand, Foot and Mouth Disease/virology , Vaccine Development/methods , Viral Vaccines/immunology , Animals , Chlorocebus aethiops , Enterovirus A, Human/growth & development , Enterovirus A, Human/immunology , Enterovirus A, Human/physiology , Humans , Male , Mice , Mice, Inbred BALB C , Vero Cells
20.
J Virol ; 95(6)2021 02 24.
Article in English | MEDLINE | ID: mdl-33408178

ABSTRACT

Coxsackievirus A5 (CV-A5) has recently emerged as a main hand, foot, and mouth disease (HFMD) pathogen. Following a large-scale vaccination campaign against enterovirus 71 (EV-71) in China, the number of HFMD-associated cases with EV-71 was reduced, especially severe and fatal cases. However, the total number of HFMD cases remains high, as HFMD is also caused by other enterovirus serotypes. A multivalent HFMD vaccine containing 4 or 6 antigens of enterovirus serotypes is urgently needed. A formaldehyde-inactivated CV-A5 vaccine derived from Vero cells was used to inoculate newborn Kunming mice on days 3 and 10. The mice were challenged on day 14 with a mouse-adapted CV-A5 strain at a dose that was lethal for 14-day-old suckling mice. Within 14 days postchallenge, groups of mice immunized with three formulations, empty particles (EPs), full particles (FPs), and a mixture of the EP and FP vaccine candidates, all survived, while 100% of the mock-immunized mice died. Neutralizing antibodies (NtAbs) were detected in the sera of immunized mice, and the NtAb levels were correlated with the survival rate of the challenged mice. The virus loads in organs were reduced, and pathological changes and viral protein expression were weak or not observed in the immunized mice compared with those in alum-inoculated control mice. Another interesting finding was the identification of CV-A5 dense particles (DPs), facilitating morphogenesis study. These results demonstrated that the Vero cell-adapted CV-A5 strain is a promising vaccine candidate and could be used as a multivalent HFMD vaccine component in the future.IMPORTANCE The vaccine candidate strain CV-A5 was produced with a high infectivity titer and a high viral particle yield. Three particle forms, empty particles (EPs), full particles (FPs), and dense particles (DPs), were obtained and characterized after purification. The immunogenicities of EP, FP, and the EP and FP mixture were evaluated in mice. Mouse-adapted CV-A5 was generated as a challenge strain to infect 14-day-old mice. An active immunization challenge mouse model was established to evaluate the efficacy of the inactivated vaccine candidate. This animal model mimics vaccination, similar to immune responses of the vaccinated. The animal model also tests protective efficacy in response to the vaccine against the disease. This work is important for the preparation of multivalent vaccines against HFMD caused by different emerging strains.


Subject(s)
Enterovirus A, Human/immunology , Hand, Foot and Mouth Disease/prevention & control , Vaccination/methods , Viral Vaccines/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Chlorocebus aethiops , Disease Models, Animal , Hand, Foot and Mouth Disease/virology , Mice , Serogroup , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Vero Cells , Viral Load , Viral Vaccines/immunology , Virion/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...