Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 493
Filtrar
1.
J Virol ; 98(2): e0135823, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38226810

RESUMEN

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.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Enfermedad de Boca, Mano y Pie , Animales , Niño , Humanos , Ratones , Anticuerpos Antivirales , Modelos Animales de Enfermedad , Enterovirus/patogenicidad , Enterovirus/fisiología , Enterovirus Humano A , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Expresión Génica , Enfermedad de Boca, Mano y Pie/genética , Fosfatidilinositol 3-Quinasas , Virulencia
2.
J Med Case Rep ; 17(1): 310, 2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37464446

RESUMEN

BACKGROUND: Enterovirus A71 is one of the causative agents of hand, foot, and mouth disease, which is usually a self-limiting disease. Complications of enterovirus infection are also very rare. However, when such complications occur, they can lead to serious neurological diseases or even death. CASE PRESENTATION: In this report, we describe a case of enterovirus A71-associated acute flaccid paralysis in a 13-month-old Caucasian girl that was managed in our hospital. The patient presented with sudden onset of left arm paresis that could not be attributed to any other cause. Establishing a diagnosis was furthermore complicated by negative virological investigations of cerebrospinal fluid and non-pathological radiological findings. A polymerase chain reaction test of the child's stool sample however tested positive for enterovirus and sequencing results revealed the presence of enterovirus A71. A previous history of febrile gastroenteritis just before the paresis started also supported the suspected diagnosis of enterovirus-associated acute flaccid paralysis. Following this, the child was treated with intravenous immunoglobulin over 5 days and a remarkable improvement was observed in the child's paresis. CONCLUSION: This case report describes a possible complication of enterovirus A71 infection in a child. It also highlights the prolonged detection of enterovirus in the child's stool sample as compared with cerebrospinal fluid weeks after the primary infection occurred. Finally, it shows the need for increased clinical and diagnostic awareness especially in the management of sudden and unknown causes of paresis or paralysis in children.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Mielitis , Femenino , Niño , Humanos , Lactante , Infecciones por Enterovirus/complicaciones , Infecciones por Enterovirus/diagnóstico , Infecciones por Enterovirus/patología , Paresia
3.
J Virol ; 97(5): e0015623, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37154751

RESUMEN

In 2014, 2016, and 2018, the United States experienced unprecedented spikes in pediatric cases of acute flaccid myelitis (AFM), which is a poliomyelitis-like paralytic illness. Accumulating clinical, immunological, and epidemiological evidence has identified enterovirus D68 (EV-D68) as a major causative agent of these biennial AFM outbreaks. There are currently no available FDA-approved antivirals that are effective against EV-D68, and the treatment for EV-D68-associated AFM is primarily supportive. Telaprevir is an food and drug administration (FDA)-approved protease inhibitor that irreversibly binds the EV-D68 2A protease and inhibits EV-D68 replication in vitro. Here, we utilize a murine model of EV-D68 associated AFM to show that early telaprevir treatment improves paralysis outcomes in Swiss Webster (SW) mice. Telaprevir reduces both viral titer and apoptotic activity in both muscles and spinal cords at early disease time points, which results in improved AFM outcomes in infected mice. Following intramuscular inoculation in mice, EV-D68 infection results in a stereotypic pattern of weakness that is reflected by the loss of the innervating motor neuron population, in sequential order, of the ipsilateral (injected) hindlimb, the contralateral hindlimb, and then the forelimbs. Telaprevir treatment preserved motor neuron populations and reduced weakness in limbs beyond the injected hindlimb. The effects of telaprevir were not seen when the treatment was delayed, and toxicity limited doses beyond 35 mg/kg. These studies are a proof of principle, provide the first evidence of benefit of an FDA-approved antiviral drug with which to treat AFM, and emphasize both the need to develop better tolerated therapies that remain efficacious when administered after viral infections and the development of clinical symptoms. IMPORTANCE Recent outbreaks of EV-D68 in 2014, 2016, and 2018 have resulted in over 600 cases of a paralytic illness that is known as AFM. AFM is a predominantly pediatric disease with no FDA-approved treatment, and many patients show minimal recovery from limb weakness. Telaprevir is an FDA-approved antiviral that has been shown to inhibit EV-D68 in vitro. Here, we demonstrate that a telaprevir treatment that is given concurrently with an EV-D68 infection improves AFM outcomes in mice by reducing apoptosis and viral titers at early time points. Telaprevir also protected motor neurons and improved paralysis outcomes in limbs beyond the site of viral inoculation. This study improves understanding of EV-D68 pathogenesis in the mouse model of AFM. This study serves as a proof of principle for the first FDA-approved drug that has been shown to improve AFM outcomes and have in vivo efficacy against EV-D68 as well as underlines the importance of the continued development of EV-D68 antivirals.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central , Enterovirus Humano D , Infecciones por Enterovirus , Animales , Estados Unidos , Ratones , Enterovirus Humano D/fisiología , Modelos Animales de Enfermedad , Parálisis/tratamiento farmacológico , Parálisis/etiología , Infecciones por Enterovirus/patología , Antivirales/farmacología , Antivirales/uso terapéutico
4.
J Virol ; 96(24): e0136722, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36448797

RESUMEN

Coxsackievirus A9 (CVA9), an enterovirus, is a common cause of pediatric aseptic meningitis and neonatal sepsis. During cell entry, enterovirus capsids undergo conformational changes leading to expansion, formation of large pores, externalization of VP1 N termini, and loss of the lipid factor from VP1. Factors such as receptor binding, heat, and acidic pH can trigger capsid expansion in some enteroviruses. Here, we show that fatty acid-free bovine serum albumin or neutral endosomal ionic conditions can independently prime CVA9 for expansion and genome release. Our results showed that CVA9 treatment with albumin or endosomal ions generated a heterogeneous population of virions, which could be physically separated by asymmetric flow field flow fractionation and computationally by cryo-electron microscopy (cryo-EM) and image processing. We report cryo-EM structures of CVA9 A-particles obtained by albumin or endosomal ion treatment and a control nonexpanded virion to 3.5, 3.3, and 2.9 Å resolution, respectively. Whereas albumin promoted stable expanded virions, the endosomal ionic concentrations induced unstable CVA9 virions which easily disintegrated, losing their genome. Loss of most of the VP4 molecules and exposure of negatively charged amino acid residues in the capsid's interior after expansion created a repulsive viral RNA-capsid interface, aiding genome release. IMPORTANCE Coxsackievirus A9 (CVA9) is a common cause of meningitis and neonatal sepsis. The triggers and mode of action of RNA release into the cell unusually do not require receptor interaction. Rather, a slow process in the endosome, independent of low pH, is required. Here, we show by biophysical separation, cryogenic electron microscopy, and image reconstruction that albumin and buffers mimicking the endosomal ion composition can separately and together expand and prime CVA9 for uncoating. Furthermore, we show in these expanded particles that VP4 is present at only ~10% of the occupancy found in the virion, VP1 is externalized, and the genome is repelled by the negatively charged, repulsive inner surface of the capsid that occurs due to the expansion. Thus, we can now link observations from cell biology of infection with the physical processes that occur in the capsid to promote genome uncoating.


Asunto(s)
Cationes , Enterovirus Humano B , Humanos , Albúminas/farmacología , Proteínas de la Cápside/metabolismo , Cationes/farmacología , Microscopía por Crioelectrón , Endosomas/metabolismo , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestructura , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , ARN/metabolismo , Virión/efectos de los fármacos , Virión/metabolismo , Virión/ultraestructura , Genoma Viral
5.
Arch Virol ; 167(12): 2483-2501, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36171507

RESUMEN

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.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Niño , Humanos , Antígenos Virales/metabolismo , Enterovirus/inmunología , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/complicaciones , Infecciones por Enterovirus/patología , Edema Pulmonar/virología
6.
Pediatr Transplant ; 26(5): e14275, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35340121

RESUMEN

BACKGROUND: Enteroviruses can cause severe infections, including viral myocarditis, meningitis, acute flaccid myelitis, and viral myositis. METHODS/RESULTS: We report a 3-year-old female renal transplant recipient who presented to a tertiary care hospital with elevated serum liver aminotransferases and subsequently developed proximal muscle pain, weakness, and respiratory distress during the first week of hospitalization. Imaging of the lower extremities revealed diffuse myositis of the proximal thigh and pelvic muscles. A muscle biopsy was obtained and revealed necrotizing myositis with immunostaining positive for enterovirus, consistent with a diagnosis of enterovirus necrotizing myositis. She had complete resolution of symptoms with steroids, intravenous immune globulin, reduced tacrolimus dose, and physical therapy. CONCLUSIONS: Enterovirus myositis should be included in the differential diagnosis for necrotizing myositis following renal transplantation in children.


Asunto(s)
Infecciones por Enterovirus , Enterovirus , Fascitis Necrotizante , Trasplante de Riñón , Mielitis , Miositis , Niño , Preescolar , Infecciones por Enterovirus/diagnóstico , Infecciones por Enterovirus/patología , Femenino , Humanos , Trasplante de Riñón/efectos adversos , Mielitis/complicaciones , Miositis/diagnóstico , Miositis/tratamiento farmacológico , Miositis/etiología
7.
Viruses ; 13(11)2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34835023

RESUMEN

Recently, bovine enterovirus (BEV) has caused several respiratory and gastrointestinal diseases outbreaks in cattle. Monitoring the epidemiological and pathogenic characteristics of this virus is crucial to controlling its spread. We isolated a BEV strain with typical cytopathic effects from the faeces of cows with significant diarrhoeal symptoms in China and observed the viral particles within 20-30 nm through transmission electron microscopy. Then, we designated this strain as HB19-1 in this study. The multistep growth curves showed that the virus propagated well in the MDBK cells. Molecular genetic analysis of VP1 indicated that HB19-1 belonged to the BEV-F1 group. Although the challenged ICR mice did not exhibit typical disease symptoms in animal infection assay, we observed significant pathological damage in the lungs, intestines, and muscle tissues. In summary, we isolated a BEV strain HB19-1 causing severe diarrhoea in cattle and proposed reinforcing the epidemiological surveillance of this virus.


Asunto(s)
Enfermedades de los Bovinos/virología , Diarrea/veterinaria , Diarrea/virología , Enterovirus Bovino/clasificación , Enterovirus Bovino/aislamiento & purificación , Animales , Bovinos , Enfermedades de los Bovinos/diagnóstico , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/patología , China , Diarrea/diagnóstico , Diarrea/epidemiología , Brotes de Enfermedades , Infecciones por Enterovirus/diagnóstico , Infecciones por Enterovirus/epidemiología , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Enterovirus Bovino/genética , Heces/virología , Femenino , Genoma Viral , Ratones , Ratones Endogámicos ICR , Filogenia , Alineación de Secuencia , Secuenciación Completa del Genoma
8.
PLoS One ; 16(11): e0258727, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34735494

RESUMEN

INTRODUCTION: Patients with hemoglobinopathies have been reported to have higher rates of pulmonary complications. Few studies have investigated the association between thalassemia and asthma in children. METHODS: We used the data of one million individuals randomly selected from the Registry for Beneficiaries of the National Health Insurance Research Database. One thalassemic child was matched with four control children without thalassemia according to sex, birth year, birth season, prematurity, and previous enteroviral infection. RESULTS: A total of 800 hundred thalassemic children and 3200 controls were included. Children with thalassemia had higher rates of developing asthma (41.81 vs 25.70 per 1000 person-years, P < 0.001) than the non-thalassemia controls with an adjusted hazard ratio of 1.37 (95% confidence interval [CI] = 1.19-1.58). Boys in the thalassemia cohort had a significantly higher adjusted incidence hazard ratio (IRR) of asthma than those in the non-thalassemia cohort (adjusted IRR = 1.45, 95% CI = 1.02-1.73). The risk of atopic and nonatopic asthma was higher in the thalassemia cohort than in the non-thalassemia cohort (IRR = 1.3, 1.61, respectively). CONCLUSIONS: Children with thalassemia were more likely to develop asthma. More attention should be paid to the early diagnosis of asthma and prevention of asthma attacks.


Asunto(s)
Asma/epidemiología , Infecciones por Enterovirus/epidemiología , Infecciones del Sistema Respiratorio/epidemiología , Talasemia/epidemiología , Adolescente , Adulto , Asma/complicaciones , Asma/patología , Asma/virología , Niño , Preescolar , Estudios de Cohortes , Bases de Datos Factuales , Infecciones por Enterovirus/complicaciones , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Femenino , Humanos , Lactante , Masculino , Hombres , Nacimiento Prematuro , Modelos de Riesgos Proporcionales , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Factores de Riesgo , Talasemia/complicaciones , Talasemia/patología , Talasemia/virología , Adulto Joven
9.
PLoS One ; 16(11): e0258680, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34731182

RESUMEN

OBJECTIVE: To evaluate the pathogenicity of a broad range of 11 possible gastroenteritis viruses, by means of statistical relationships with cases vs. controls, or Ct-values, in order to establish the most appropriate diagnostic panel for our general practitioner (GP) patients in the Netherlands (2010-2012). METHODS: Archived stool samples from 1340 cases and 1100 controls were retested using internally controlled multiplex real-time PCRs for putative pathogenic gastroenteritis viruses: adenovirus, astrovirus, bocavirus, enterovirus, norovirus GI and GII, human parechovirus, rotavirus, salivirus, sapovirus, and torovirus. RESULTS: The prevalence of any virus in symptomatic cases and asymptomatic controls was 16.6% (223/1340) and 10.2% (112/1100), respectively. Prevalence of astrovirus (adjusted odds ratio (aOR) 10.37; 95% confidence interval (CI) 1.34-80.06) and norovirus GII (aOR 3.10; CI 1.62-5.92) was significantly higher in cases versus controls. Rotavirus was encountered only in cases. We did not find torovirus and there was no statistically significant relationship with cases for salivirus (aOR 1,67; (CI) 0.43-6.54)), adenovirus non-group F (aOR 1.20; CI 0.75-1.91), bocavirus (aOR 0.85; CI 0.05-13.64), enterovirus (aOR 0.83; CI 0.50-1.37), human parechovirus (aOR 1.61; CI 0.54-4.77) and sapovirus (aOR 1.15; CI 0.67-1.98). Though adenovirus group F (aOR 6.37; CI 0.80-50.92) and norovirus GI (aOR 2.22, CI: 0.79-6.23) are known enteropathogenic viruses and were more prevalent in cases than in controls, this did not reach significance in this study. The Ct value did not discriminate between carriage and disease in PCR-positive subjects. CONCLUSIONS: In our population, diagnostic gastroenteritis tests should screen for adenovirus group F, astrovirus, noroviruses GI and GII, and rotavirus. Case-control studies as ours are lacking and should also be carried out in populations from other epidemiological backgrounds.


Asunto(s)
Infecciones por Enterovirus/diagnóstico , Heces/virología , Gastroenteritis/diagnóstico , Adenoviridae/genética , Adenoviridae/aislamiento & purificación , Adenoviridae/patogenicidad , Bocavirus/genética , Bocavirus/aislamiento & purificación , Bocavirus/patogenicidad , Preescolar , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Femenino , Gastroenteritis/genética , Gastroenteritis/patología , Gastroenteritis/virología , Médicos Generales , Humanos , Lactante , Masculino , Norovirus/genética , Norovirus/aislamiento & purificación , Norovirus/patogenicidad , Pacientes , Rotavirus/genética , Rotavirus/aislamiento & purificación , Rotavirus/patogenicidad , Sapovirus/genética , Sapovirus/aislamiento & purificación , Sapovirus/patogenicidad
10.
Front Immunol ; 12: 700903, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566960

RESUMEN

The activation of the sympathetic nervous system, release of norepinephrine (NE), and adrenergic receptor signaling participate in and regulate the complicated enterovirus 71 (EV71) brainstem encephalitis (BE). The neurotoxin 6-hydroxydopamine (6-OHDA) selectively ablates sympathetic nerves and markedly depletes NE in innervated organs. Changes in the plasma levels of NE, severity score, cytokine profiles, and percentages of immunophenotype expression in 7-day-old Bltw : CD1 (ICR) mice infected with EV71, with or without 6-OHDA treatment, were compared. The survival rate (76.9%) of EV71-infected and 6-OHDA (30 µg/g)-treated mice was increased significantly. The clinical scores were decreased markedly on days 8-12 in MP4-infected and 6-OHDA-treated mice compared to those without treatment. The results showed that the plasma levels of NE, epinephrine, and dopamine were decreased on days 4-8 after 6-OHDA treatment and at most on day 8. The plasma levels of interleukin (IL)-12p70, tumor necrosis factor, IL-6, and IL-10 did not change significantly after 6-OHDA treatment. Interferon-γ levels decreased evidently on days 4, 6, and 8 after 6-OHDA treatment. The absolute events of CD3+CD4+, CD3+CD8+, and CD3+NK1.1+ cells of peripheral blood mononuclear cells were increased significantly in MP4-infected and 6-OHDA-treated mice compared to those without treatment. In splenocytes, the absolute cells of CD3-NK1.1+, CD3+NK1.1+ and CD11b+Gr-1+ cells of EV71-infected mice were increased significantly after 6-OHDA treatment. These findings suggested that 6-OHDA may be used a probe to explore clinical improvements and immune responses in the complicated EV71 infection. Taken together, peripheral chemical sympathectomy contribute to further understand the immunopathogenesis of EV71 BE with autonomic nervous system dysregulation.


Asunto(s)
Encefalitis Viral/inmunología , Infecciones por Enterovirus/inmunología , Simpatectomía Química/métodos , Animales , Tronco Encefálico/inmunología , Tronco Encefálico/patología , Encefalitis Viral/patología , Enterovirus Humano A , Infecciones por Enterovirus/patología , Ratones , Ratones Endogámicos ICR , Oxidopamina
11.
J Virol ; 95(21): e0089721, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34379497

RESUMEN

Enterovirus A71 (EV-A71) and many members of the Picornaviridae family are neurotropic pathogens of global concern. These viruses are primarily transmitted through the fecal-oral route, and thus suitable animal models of oral infection are needed to investigate viral pathogenesis. An animal model of oral infection was developed using transgenic mice expressing human SCARB2 (hSCARB2 Tg), murine-adapted EV-A71/MP4 virus, and EV-A71/MP4 virus with an engineered nanoluciferase gene that allows imaging of viral replication and spread in infected mice. Next-generation sequencing of EV-A71 genomes in the tissues and organs of infected mice was also performed. Oral inoculation of EV-A71/MP4 or nanoluciferase-carrying MP4 virus stably induced neurological symptoms and death in infected 21-day-old weaned mice. In vivo bioluminescence imaging of infected mice and tissue immunostaining of viral antigens indicated that orally inoculated virus can spread to the central nervous system (CNS) and other tissues. Next-generating sequencing further identified diverse mutations in viral genomes that can potentially contribute to viral pathogenesis. This study presents an EV-A71 oral infection murine model that efficiently infects weaned mice and allows tracking of viral spread, features that can facilitate research into viral pathogenesis and neuroinvasion via the natural route of infection. IMPORTANCE Enterovirus A71 (EV-A71), a positive-strand RNA virus of the Picornaviridae, poses a persistent global public health problem. EV-A71 is primarily transmitted through the fecal-oral route, and thus suitable animal models of oral infection are needed to investigate viral pathogenesis. We present an animal model of EV-A71 infection that enables the natural route of oral infection in weaned and nonimmunocompromised 21-day-old hSCARB2 transgenic mice. Our results demonstrate that severe disease and death could be stably induced, and viral invasion of the CNS could be replicated in this model, similar to severe real-world EV-A71 infections. We also developed a nanoluciferase-containing EV-A71 virus that can be used with this animal model to track viral spread after oral infection in real time. Such a model offers several advantages over existing animal models and can facilitate future research into viral spread, tissue tropism, and viral pathogenesis, all pressing issues that remain unaddressed for EV-A71 infections.


Asunto(s)
Sistema Nervioso Central/virología , Enterovirus Humano A/patogenicidad , Infecciones por Enterovirus/complicaciones , Proteínas de Membrana de los Lisosomas/genética , Boca/virología , Enfermedades del Sistema Nervioso/virología , Receptores Depuradores/genética , Animales , Modelos Animales de Enfermedad , Enterovirus Humano A/genética , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Genoma Viral , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Tropismo Viral , Replicación Viral , Destete
12.
Arch Virol ; 166(10): 2683-2692, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34268639

RESUMEN

Porcine sapelovirus (PSV) infections have been associated with a wide spectrum of symptoms, ranging from asymptomatic infection to clinical signs including diarrhoea, pneumonia, reproductive disorders, and polioencephalomyelitis. Although it has a global distribution, there have been relatively few studies on PSV in domestic animals. We isolated a PSV strain, SHCM2019, from faecal specimens from swine, using PK-15 cells. To investigate its molecular characteristics and pathogenicity, the genomic sequence of strain SHCM2019 was analysed, and clinical manifestations and pathological changes occurring after inoculation of neonatal piglets were observed. The virus isolated using PK-15 cells was identified as PSV using RT-PCR, transmission electron microscopy (TEM), and immunofluorescence assay (IFA). Sequencing results showed that the full-length genome of the SHCM2019 strain was 7,567 nucleotides (nt) in length, including a 27-nucleotide poly(A) tail. Phylogenetic analysis demonstrated that this virus was a PSV isolate belonging to the Chinese strain cluster. Recombination analysis indicated that there might be a recombination breakpoint upstream of the 3D region of the genome. Pathogenicity experiments demonstrated that the virus isolate could cause diarrhoea and pneumonia in piglets. In breif, a recombinant PSV strain, SHCM2019, was isolated and shown to be pathogenic. Our results may provide a reference for future research on the pathogenic mechanism and evolutionary characteristics of PSV.


Asunto(s)
Infecciones por Enterovirus/veterinaria , Enterovirus Porcinos/genética , Enterovirus Porcinos/aislamiento & purificación , Enfermedades de los Porcinos/virología , Animales , Línea Celular , China , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Enterovirus Porcinos/clasificación , Enterovirus Porcinos/patogenicidad , Heces/virología , Genoma Viral/genética , Filogenia , Recombinación Genética , Porcinos , Enfermedades de los Porcinos/patología , Virulencia
13.
mSphere ; 6(2)2021 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692197

RESUMEN

Enterovirus 71 (EV71) can cause a severe hand-foot-mouth disease in children. However, the precise mechanism of EV71-associated disease, particularly the neuropathogenesis and pulmonary disorder, is still not fully understood because no suitable animal models are available. The human scavenger receptor class B, member 2 (hSCARB2), is a cellular receptor for EV71. Here, we generated a novel knock-in (KI) mouse model using the CRISPR/Cas9 system to insert the hSCARB2 gene into the mouse Rosa26 locus to study the pathogenesis of EV71. The hSCARB2 KI mice infected with clinical isolates of EV71 showed neurological symptoms, such as ataxia, paralysis, and death. Viral replication was detected in mainly astrocytes and a limited number of neurons and microglia, accompanied by gliosis. Vascular leakage and alveoli filled with erythrocytes were detected, suggesting that edema and hemorrhage, which are observed in human patients, also occurred in EV71-infected KI mice. In addition, proinflammatory cytokines and chemokines were significantly increased in the serum of infected KI mice. These pathological features of the KI mice after infection resembled those of EV71 encephalomyelitis in humans. Therefore, our KI mouse model is suitable to study the pathogenesis of EV71 and is of great significance for development of antiviral drugs and vaccines to treat or prevent EV71 infection.IMPORTANCE Enterovirus 71 (EV71) is associated with severe hand-foot-mouth disease. Recently, outbreaks of EV71 infection with high mortality have been reported in the Asia-Pacific region, posing a great challenge for global public health. To date, the precise mechanism of EV71-induced disease, particularly the neuropathogenesis and respiratory disorders, is still not fully understood because no suitable animal models are available. Human scavenger receptor class B, member 2 (hSCARB2), has been identified as a cellular receptor for EV71. Here, we introduce a novel CRISPR/Cas9-mediated hSCARB2 knock-in (KI) mouse model for the study of EV71 pathogenesis, which is of great significance for the development of antiviral drugs and vaccines.


Asunto(s)
Enterovirus Humano A/genética , Enterovirus Humano A/patogenicidad , Infecciones por Enterovirus/patología , Proteínas de Membrana de los Lisosomas/genética , Receptores Depuradores/genética , Animales , Astrocitos/virología , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Infecciones por Enterovirus/inmunología , Femenino , Técnicas de Sustitución del Gen , Enfermedad de Boca, Mano y Pie/complicaciones , Enfermedad de Boca, Mano y Pie/virología , Humanos , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/virología
14.
Arch Virol ; 166(4): 1133-1140, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33590343

RESUMEN

Severe hand, foot, and mouth disease (HFMD) caused by enterovirus 71 (EV71) infection is associated with high mortality and disability. DC-SIGN, a receptor for EV71, is widely distributed in dendritic cells and may influence the severity of HFMD caused by EV71 infection. This observational study attempts to explore whether single-nucleotide polymorphisms (SNPs) in DC-SIGN are related to the severity of EV71-associated HFMD. Based on linkage disequilibrium and functional predictions, two DC-SIGN SNPs were selected and tested to explore their potential association with the severity of HFMD caused by EV71 infection. Two hundred sixteen Han Chinese children with HFMD caused by EV71 were enrolled to obtain clinical data, including the severity of HFMD, serum DC-SIGN levels, and DC-SIGN SNPs. We found a significant association between the rs7248637 polymorphism (A vs. G: OR = 0.644, 95% CI = 0.515-0.806) and the severity of HFMD caused by EV71 infection, as well as the rs4804800 polymorphism (A vs. G: OR = 1.539, 95% CI =1.229-1.928). These two DC-SIGN SNPs may have an effect on the severity of HFMD caused by EV71 infection.


Asunto(s)
Moléculas de Adhesión Celular/genética , Infecciones por Enterovirus/genética , Predisposición Genética a la Enfermedad/genética , Enfermedad de Boca, Mano y Pie/genética , Lectinas Tipo C/genética , Receptores de Superficie Celular/genética , Pueblo Asiatico/genética , Moléculas de Adhesión Celular/sangre , Niño , Preescolar , China/epidemiología , Enterovirus Humano A , Infecciones por Enterovirus/epidemiología , Infecciones por Enterovirus/patología , Femenino , Estudios de Asociación Genética , Genotipo , Enfermedad de Boca, Mano y Pie/epidemiología , Enfermedad de Boca, Mano y Pie/patología , Humanos , Lactante , Lectinas Tipo C/sangre , Masculino , Polimorfismo de Nucleótido Simple , Receptores de Superficie Celular/sangre , Índice de Severidad de la Enfermedad
15.
Life Sci ; 271: 119188, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33581126

RESUMEN

AIMS: Enterovirus 71 (EV71) is one of the main viruses that cause hand-foot-mouth disease; however, its pathogenic mechanism remains unclear. This study characterized the relationship between EV71 infection and autophagy in vivo and explored the molecular mechanism underlying EV71-induced autophagy. MATERIALS AND METHODS: A mouse model of EV71 infection was prepared by intraperitoneally injecting one-day-old BALB/c suckling mice with 30 µL/g of EV71 virus stock solution for 3 days. The behavior, fur condition, weight, and mice mortality were monitored, and disease scores were calculated. The pathological damage to the brain, lung, and muscle tissues after the viral infection was assessed by hematoxylin and eosin staining. Western blot and immunofluorescence analyses were used to detect the expression levels of viral protein 1, Beclin-1, microtubule-associated protein light chain 3B, mammalian target of rapamycin (mTOR), phosphorylated (p)-mTOR, extracellular signal-regulated protein kinase (ERK) 1/2, and p-ERK. KEY FINDINGS: EV71 infection can trigger autophagy in the brains, lungs, and muscles of infected mice. The autophagy response triggered by EV71 is achieved by the simultaneous mTOR inhibition and the ERK pathway activation. Blocking the mTOR pathway may aggravate autophagy, whereas ERK inhibition alleviates autophagy but cannot completely prevent it. SIGNIFICANCE: EV71 infection can induce autophagy in mice, involving mTOR and ERK signaling pathways. These two signaling pathways are independent and do not interfere with each other.


Asunto(s)
Autofagia/fisiología , Enterovirus Humano A/metabolismo , Infecciones por Enterovirus/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Infecciones por Enterovirus/patología , Activación Enzimática/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C
16.
RNA Biol ; 18(5): 796-808, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33406999

RESUMEN

The pathogenic human enterovirus EV-A71 has raised serious public health concerns. A hallmark of EV-A71 infection is the distortion of host transcriptomes in favour of viral replication. While high-throughput approaches have been exploited to dissect these gene dysregulations, they do not fully capture molecular perturbations at the single-cell level and in a physiologically relevant context. In this study, we applied a single-cell RNA sequencing approach on infected differentiated enterocyte cells (C2BBe1), which model the gastrointestinal epithelium targeted initially by EV-A71. Our single-cell analysis of EV-A71-infected culture provided several lines of illuminating observations: 1) This systems approach demonstrated extensive cell-to-cell variation in a single culture upon viral infection and delineated transcriptomic differences between the EV-A71-infected and bystander cells. 2) By analysing expression profiles of known EV-A71 receptors and entry facilitation factors, we found that ANXA2 was closely correlated in expression with the viral RNA in the infected population, supporting its role in EV-A71 entry in the enteric cells. 3) We further catalogued dysregulated lncRNAs elicited by EV-A71 infection and demonstrated the functional implication of lncRNA CYTOR in promoting EV-A71 replication. Viewed together, our single-cell transcriptomic analysis illustrated at the single-cell resolution the heterogeneity of host susceptibility to EV-A71 and revealed the involvement of lncRNAs in host antiviral response.


Asunto(s)
Enterovirus Humano A/patogenicidad , Interacciones Huésped-Patógeno/genética , Transcriptoma , Células Cultivadas , Enterocitos/metabolismo , Enterocitos/patología , Enterocitos/virología , Enterovirus Humano A/genética , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/virología , ARN Largo no Codificante/genética , Análisis de la Célula Individual , Replicación Viral/genética
17.
J Virol ; 95(3)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33148796

RESUMEN

Human enterovirus D68 (EV-D68) has received considerable attention recently as a global reemergent pathogen because it causes severe respiratory tract infections and acute flaccid myelitis (AFM). The nonstructural protein 2A protease (2Apro) of EVs, which functions in the cleavage of host proteins, comprises a pivotal part of the viral immune evasion process. However, the pathogenic mechanism of EV-D68 is not fully understood. In this study, we found that EV-D68 inhibited antiviral type I interferon responses by cleaving tumor necrosis factor receptor-associated factor 3 (TRAF3), which is the key factor for type I interferon production. EV-D68 inhibited Sendai virus (SEV)-induced interferon regulatory factor 3 (IRF3) activation and beta interferon (IFN-ß) expression in HeLa and HEK293T cells. Furthermore, we demonstrated that EV-D68 and 2Apro were able to cleave the C-terminal region of TRAF3 in HeLa and HEK293T cells, respectively. A cysteine-to-alanine substitution at amino acid 107 (C107A) in the 2Apro protease resulted in the loss of cleavage activity to TRAF3, and mutation of glycine at amino acid 462 to alanine (G462A) in TRAF3 conferred resistance to 2Apro These results suggest that control of TRAF3 by 2Apro may be a mechanism EV-D68 utilizes to subvert host innate immune responses.IMPORTANCE Human enterovirus 68 (EV-D68) has received considerable attention recently as a global reemergent pathogen because it causes severe respiratory tract infections and acute flaccid myelitis. The nonstructural protein 2A protease (2Apro) of EV, which functions in cleavage of host proteins, comprises an essential part of the viral immune evasion process. However, the pathogenic mechanism of EV-D68 is not fully understood. Here, we show for the first time that EV-D68 inhibited antiviral type I interferon responses by cleaving tumor necrosis factor receptor-associated factor 3 (TRAF3). Furthermore, we identified the key cleavage site in TRAF3. Our study may suggest a new mechanism by which the 2Apro of EV facilitates subversion of host innate immune responses. These findings increase our understanding of EV-D68 infection and may help identify new antiviral targets against EV-D68.


Asunto(s)
Enterovirus Humano D/enzimología , Infecciones por Enterovirus/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Péptido Hidrolasas/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Proteínas Virales/metabolismo , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/metabolismo , Péptido Hidrolasas/genética , Proteolisis , Factor 3 Asociado a Receptor de TNF/genética , Proteínas Virales/genética
18.
J Microbiol ; 58(9): 812-820, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32870487

RESUMEN

Enterovirus D68 (EVD68) is an emerging pathogen that recently caused a large worldwide outbreak of severe respiratory disease in children. However, the relationship between EVD68 and host cells remains unclear. Caspases are involved in cell death, immune response, and even viral production. We found that caspase-3 was activated during EVD68 replication to induce apoptosis. Caspase-3 inhibitor (Z-DEVD-FMK) inhibited viral production, protected host cells from the cytopathic effects of EVD68 infection, and prevented EVD68 from regulating the host cell cycle at G0/G1. Meanwhile, caspase-3 activator (PAC-1) increased EVD68 production. EVD68 infection therefore activates caspase-3 for virus production. This knowledge provides a potential direction for the prevention and treatment of disease related to EVD68.


Asunto(s)
Antivirales/farmacología , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Enterovirus Humano D/efectos de los fármacos , Enterovirus Humano D/crecimiento & desarrollo , Apoptosis/fisiología , Línea Celular Tumoral , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/prevención & control , Infecciones por Enterovirus/virología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Hidrazonas/farmacología , Oligopéptidos/farmacología , Piperazinas/farmacología
19.
J Neurovirol ; 26(5): 790-792, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32671810

RESUMEN

Enteroviruses are one of the most important causes of viral encephalitis in the neonatal period. However, the non-specificity of the symptoms presented renders its diagnosis challenging. Intracranial MRI has been reported to be a very useful imaging modality that can detect the characteristic white matter lesions around the periventricular regions. In this study, we report a case of a patient with neonatal encephalitis who presented with normal white blood cell counts in the initial cerebrospinal fluid analysis. A lumbar puncture retap identified pleocytosis, and polymerase chain reaction assays detected enterovirus 71 in the blood and stool samples. Furthermore, MRI revealed atypical disseminated cortical and subcortical white matter lesions on diffusion weighted images, and neuroradiological re-evaluation showed necrotic changes 2 weeks later. This unique case expands our knowledge of the spectrum of neurological disorders due to enterovirus 71 infection in neonatal period.


Asunto(s)
Encefalitis Viral/diagnóstico por imagen , Enterovirus Humano A/patogenicidad , Infecciones por Enterovirus/diagnóstico por imagen , Sustancia Blanca/diagnóstico por imagen , Aciclovir/uso terapéutico , Antivirales/uso terapéutico , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/patología , Corteza Cerebral/virología , Imagen de Difusión por Resonancia Magnética/métodos , Encefalitis Viral/tratamiento farmacológico , Encefalitis Viral/patología , Encefalitis Viral/virología , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/genética , Enterovirus Humano A/crecimiento & desarrollo , Infecciones por Enterovirus/tratamiento farmacológico , Infecciones por Enterovirus/patología , Infecciones por Enterovirus/virología , Humanos , Recién Nacido , Masculino , Neuroimagen/métodos , Punción Espinal/métodos , Sustancia Blanca/patología , Sustancia Blanca/virología
20.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31666382

RESUMEN

The gastrointestinal tract presents a formidable barrier for pathogens to initiate infection. Despite this barrier, enteroviruses, including coxsackievirus B3 (CVB3), successfully penetrate the intestine to initiate infection and spread systemically prior to shedding in stool. However, the effect of the gastrointestinal barrier on CVB3 population dynamics is relatively unexplored, and the selective pressures acting on CVB3 in the intestine are not well characterized. To examine viral population dynamics in orally infected mice, we produced over 100 CVB3 clones harboring nine unique nucleotide "barcodes." Using this collection of barcoded viruses, we found diverse viral populations throughout each mouse within the first day postinfection, but by 48 h the viral populations were dominated by fewer than three barcoded viruses in intestinal and extraintestinal tissues. Using light-sensitive viruses to track replication status, we found that diverse viruses had replicated prior to loss of diversity. Sequencing whole viral genomes from samples later in infection did not reveal detectable viral adaptations. Surprisingly, orally inoculated CVB3 was detectable in pancreas and liver as soon as 20 min postinoculation, indicating rapid systemic dissemination. These results suggest rapid dissemination of diverse viral populations, followed by a major restriction in population diversity and monopolization in all examined tissues. These results underscore a complex dynamic between dissemination and clearance for an enteric virus.IMPORTANCE Enteric viruses initiate infection in the gastrointestinal tract but can disseminate to systemic sites. However, the dynamics of viral dissemination are unclear. In this study, we created a library of 135 barcoded coxsackieviruses to examine viral population diversity across time and space following oral inoculation of mice. Overall, we found that the broad population of viruses disseminates early, followed by monopolization of mouse tissues with three or fewer pool members at later time points. Interestingly, we detected virus in systemic tissues such as pancreas and liver just 20 min after oral inoculation. These results suggest rapid dissemination of diverse viral populations, followed by a major restriction in population diversity and monopolization in all examined tissues.


Asunto(s)
Código de Barras del ADN Taxonómico , Enterovirus Humano B/fisiología , Infecciones por Enterovirus , Replicación Viral , Animales , Infecciones por Enterovirus/genética , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/patología , Células HeLa , Humanos , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...