Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 975
Filtrar
1.
J Virol ; 97(9): e0055523, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37668370

RESUMO

In vitro investigations of host-virus interactions are reliant on suitable cell and tissue culture models. Results are only as good as the model they are generated in. However, choosing cell models for in vitro work often depends on availability and previous use alone. Despite the vast increase in coronavirus research over the past few years, scientists are still heavily reliant on: non-human, highly heterogeneous or not fully differentiated, or naturally unsusceptible cells requiring overexpression of receptors and other accessory factors. Complex primary or stem cell models are highly representative of human tissues but are expensive and time-consuming to develop and maintain with limited suitability for high-throughput experiments.Using tissue-specific expression patterns, we identified human kidney cells as an ideal target for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and broader coronavirus infection. We show the use of the well-characterized human kidney cell line Caki-1 for infection with three human coronaviruses (hCoVs): Betacoronaviruses SARS-CoV-2 and Middle Eastern respiratory syndrome coronavirus and Alphacoronavirus hCoV 229E. Caki-1 cells show equal or superior susceptibility to all three coronaviruses when compared to other commonly used cell lines for the cultivation of the respective virus. Antibody staining against SARS-CoV-2 N protein shows comparable replication rates. A panel of 26 custom antibodies shows the location of SARS-CoV-2 proteins during replication using immunocytochemistry. In addition, Caki-1 cells were found to be susceptible to two other human respiratory viruses, influenza A virus and respiratory syncytial virus, making them an ideal model for cross-comparison for a broad range of respiratory viruses. IMPORTANCE Cell lines remain the backbone of virus research, but results are only as good as their originating model. Despite increased research into human coronaviruses following the COVID-19 pandemic, researchers continue to rely on suboptimal cell line models of: non-human origin, incomplete differentiation, or lacking active interferon responses. We identified the human kidney Caki-1 cell line as a potential target for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). This cell line could be shown to be infectable with a wide range of coronaviruses including common cold virus hCoV-229E, epidemic virus MERS-CoV, and SARS-CoV-2 as well as other important respiratory viruses influenza A virus and respiratory syncytial virus. We could show the localization of 26 SARS-CoV-2 proteins in Caki-1 cells during natural replication and the cells are competent of forming a cellular immune response. Together, this makes Caki-1 cells a unique tool for cross-virus comparison in one cell line.


Assuntos
Linhagem Celular , Infecções por Coronaviridae , Coronaviridae , Humanos , Coronaviridae/fisiologia , Rim/citologia , Pandemias , Infecções por Coronaviridae/patologia , Infecções por Coronaviridae/virologia
2.
Washington D.C; Organización Panamericana de la Salud; 1 ed; Oct. 2022. 9 p. ilus.
Monografia em Espanhol | MINSAPERÚ, LIPECS | ID: biblio-1397941

RESUMO

La presente guía presenta una serie de recomendaciones dirigidas a facilitar el diálogo entre el personal de salud a cargo de las sesiones de vacunación e información y el público general. Saber permanecer a la escucha de sus inquietudes e informar con claridad y sencillez, en un clima de empatía y respeto, son sin duda las claves de un diálogo que permitirá seguir promoviendo la vacunación entre la población


Assuntos
Imunização , Pessoal de Saúde , Infecções por Coronaviridae , Empatia , Vacinas contra COVID-19 , COVID-19
3.
Viruses ; 14(2)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35215769

RESUMO

Bats have been recognized as an exceptional viral reservoir, especially for coronaviruses. At least three bat zoonotic coronaviruses (SARS-CoV, MERS-CoV and SARS-CoV-2) have been shown to cause severe diseases in humans and it is expected more will emerge. One of the major features of CoVs is that they are all highly prone to recombination. An extreme example is the insertion of the P10 gene from reoviruses in the bat CoV GCCDC1, first discovered in Rousettus leschenaultii bats in China. Here, we report the detection of GCCDC1 in four different bat species (Eonycteris spelaea, Cynopterus sphinx, Rhinolophus shameli and Rousettus sp.) in Cambodia. This finding demonstrates a much broader geographic and bat species range for this virus and indicates common cross-species transmission. Interestingly, one of the bat samples showed a co-infection with an Alpha CoV most closely related to RsYN14, a virus recently discovered in the same genus (Rhinolophus) of bat in Yunnan, China, 2020. Taken together, our latest findings highlight the need to conduct active surveillance in bats to assess the risk of emerging CoVs, especially in Southeast Asia.


Assuntos
Quirópteros/virologia , Infecções por Coronaviridae/veterinária , Coronaviridae/classificação , Coronaviridae/genética , Reservatórios de Doenças/veterinária , Reservatórios de Doenças/virologia , Filogeografia , Recombinação Genética , Animais , Camboja/epidemiologia , China/epidemiologia , Quirópteros/classificação , Coronaviridae/isolamento & purificação , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/transmissão , Evolução Molecular , Genoma Viral , Filogenia
5.
PLoS One ; 16(11): e0260038, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34813610

RESUMO

At present, global immunity to SARS-CoV-2 resides within a heterogeneous combination of susceptible, naturally infected and vaccinated individuals. The extent to which viral shedding and transmission occurs on re-exposure to SARS-CoV-2 is an important determinant of the rate at which COVID-19 achieves endemic stability. We used Sialodacryoadenitis Virus (SDAV) in rats to model the extent to which immune protection afforded by prior natural infection via high risk (inoculation; direct contact) or low risk (fomite) exposure, or by vaccination, influenced viral shedding and transmission on re-exposure. On initial infection, we confirmed that amount, duration and consistency of viral shedding, and seroconversion rates were correlated with exposure risk. Animals were reinfected after 3.7-5.5 months using the same exposure paradigm. 59% of seropositive animals shed virus, although at lower amounts. Previously exposed seropositive reinfected animals were able to transmit virus to 25% of naive recipient rats after 24-hour exposure by direct contact. Rats vaccinated intranasally with a related virus (Parker's Rat Coronavirus) were able to transmit SDAV to only 4.7% of naive animals after a 7-day direct contact exposure, despite comparable viral shedding. Cycle threshold values associated with transmission in both groups ranged from 29-36 cycles. Observed shedding was not a prerequisite for transmission. Results indicate that low-level shedding in both naturally infected and vaccinated seropositive animals can propagate infection in susceptible individuals. Extrapolated to COVID-19, our results suggest that continued propagation of SARS-CoV-2 by seropositive previously infected or vaccinated individuals is possible.


Assuntos
COVID-19/transmissão , Infecções por Coronaviridae/veterinária , Coronavirus do Rato/fisiologia , Modelos Biológicos , Modelos Estatísticos , Doenças dos Roedores/transmissão , Eliminação de Partículas Virais , Animais , COVID-19/virologia , Infecções por Coronaviridae/transmissão , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , SARS-CoV-2/fisiologia , Soroconversão
6.
Cells ; 10(7)2021 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-34359986

RESUMO

A novel coronavirus discovered in 2019 is a new strain of the Coronaviridae family (CoVs) that had not been previously identified in humans. It is known as SARS-CoV-2 for Severe Acute Respiratory Syndrome Coronavirus-2, whilst COVID-19 is the name of the disease associated with the virus. SARS-CoV-2 emerged over one year ago and still haunts the human community throughout the world, causing both healthcare and socioeconomic problems. SARS-CoV-2 is spreading with many uncertainties about treatment and prevention: the data available are limited and there are few randomized controlled trial data on the efficacy of antiviral or immunomodulatory agents. SARS-CoV-2 and its mutants are considered as unique within the Coronaviridae family insofar as they spread rapidly and can have severe effects on health. Although the scientific world has been succeeding in developing vaccines and medicines to combat COVID-19, the appearance and the spread of new, more aggressive mutants are posing extra problems for treatment. Nevertheless, our understanding of pandemics is increasing significantly due to this outbreak and is leading to the development of many different pharmacological, immunological and other treatments. This Review focuses on a subset of COVID-19 research, primarily the cytoskeleton-related physiological and pathological processes in which coronaviruses such as SARS-CoV-2 are intimately involved. The discovery of the exact mechanisms of the subversion of host cells by SARS-CoV-2 is critical to the validation of specific drug targets and effective treatments.


Assuntos
Antivirais/farmacologia , COVID-19/patologia , Infecções por Coronaviridae/patologia , Citoesqueleto/patologia , Animais , Antivirais/uso terapêutico , Infecções por Coronaviridae/tratamento farmacológico , Coronavirus/efeitos dos fármacos , Coronavirus/fisiologia , Citoesqueleto/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/fisiologia , Tratamento Farmacológico da COVID-19
7.
Virology ; 563: 20-27, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34411808

RESUMO

Viruses of the subfamily Orthocoronavirinae can cause mild to severe disease in people, including COVID-19, MERS and SARS. Their most common natural hosts are bat and bird species, which are mostly split across four virus genera. Molecular clock analyses of orthocoronaviruses suggested the most recent common ancestor of these viruses might have emerged either around 10,000 years ago or, using models accounting for selection, many millions of years. Here, we reassess the evolutionary history of these viruses. We present time-aware phylogenetic analyses of a RNA-dependent RNA polymerase locus from 123 orthocoronaviruses isolated from birds and bats, including those in New Zealand, which were geographically isolated from other bats around 35 million years ago. We used this age, as well as the age of the avian-mammals split, to calibrate the molecular clocks, under the assumption that these ages are applicable to the analyzed viruses. We found that the time to the most recent ancestor common for all orthocoronaviruses is likely 150 or more million years, supporting clock analyses that account for selection.


Assuntos
Aves/virologia , Quirópteros/virologia , Infecções por Coronaviridae/virologia , Coronaviridae , Genoma Viral , Animais , Coronaviridae/classificação , Coronaviridae/genética , Evolução Molecular , Nova Zelândia/epidemiologia
8.
Biomed Res Int ; 2021: 8856018, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34239932

RESUMO

Coronaviruses (CoVs) are enveloped nonsegmented positive-sense RNA viruses belonging to the family Coronaviridae that contain the largest genome among RNA viruses. Their genome encodes 4 major structural proteins, and among them, the Spike (S) protein plays a crucial role in determining the viral tropism. It mediates viral attachment to the host cell, fusion to the membranes, and cell entry using cellular proteases as activators. Several in vitro models have been developed to study the CoVs entry, pathogenesis, and possible therapeutic approaches. This article is aimed at summarizing the current knowledge about the use of relevant methodologies and cell lines permissive for CoV life cycle studies. The synthesis of this information can be useful for setting up specific experimental procedures. We also discuss different strategies for inhibiting the binding of the S protein to the cell receptors and the fusion process which may offer opportunities for therapeutic intervention.


Assuntos
Antivirais , Coronaviridae , Modelos Biológicos , Tropismo Viral , Internalização do Vírus , Antivirais/química , Antivirais/farmacologia , COVID-19 , Células Cultivadas , Coronaviridae/efeitos dos fármacos , Coronaviridae/metabolismo , Coronaviridae/patogenicidade , Coronaviridae/fisiologia , Infecções por Coronaviridae , Humanos , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo
9.
Avian Pathol ; 50(4): 295-310, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34126817

RESUMO

Infectious bronchitis virus (IBV) was first isolated in Australia in 1962. Ongoing surveillance and characterization of Australian IBVs have shown that they have evolved separately from strains found throughout the rest of the world, resulting in the evolution of a range of unique strains and changes in the dominant wild-type strains, affecting tissue tropism, pathogenicity, antigenicity, and gene arrangement. Between 1961 and 1976 highly nephropathogenic genotype GI-5 and GI-6 strains, causing mortalities of 40% to 100%, predominated, while strains causing mainly respiratory disease, with lower mortality rates, have predominated since then. Since 1988, viruses belonging to two distinct and novel genotypes, GIII and GV, have been detected. The genome organization of the GIII strains has not been seen in any other gammacoronavirus. Mutations that emerged soon after the introduction of vaccination, incursion of strains with a novel lineage from unknown sources, recombination between IBVs from different genetic lineages, and gene translocations and deletions have contributed to an increasingly complex IBV population. These processes and the consequences of this variation for the biology of these viruses provide an insight into the evolution of endemic coronaviruses during their control by vaccination and may provide a better understanding of the potential for evolution of other coronaviruses, including SARS-CoV-2. Furthermore, the continuing capacity of attenuated IBV vaccines developed over 40 years ago to provide protection against viruses in the same genetic lineage provides some assurance that coronavirus vaccines developed to control other coronaviruses may continue to be effective for an extended period.


Assuntos
Evolução Biológica , Galinhas , Infecções por Coronaviridae/veterinária , Vírus da Bronquite Infecciosa/fisiologia , Doenças das Aves Domésticas/virologia , Animais , Variação Antigênica , Austrália/epidemiologia , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/prevenção & controle , Infecções por Coronaviridae/virologia , Evolução Molecular , Variação Genética , Vírus da Bronquite Infecciosa/classificação , Vírus da Bronquite Infecciosa/genética , Vírus da Bronquite Infecciosa/imunologia , Fenótipo , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais
11.
Cir. Esp. (Ed. impr.) ; 99(5): 368-373, mayo 2021. tab, graf
Artigo em Espanhol | IBECS | ID: ibc-192543

RESUMO

INTRODUCCIÓN: La pandemia por SARS-CoV-2 ha causado un importante impacto en nuestro medio, con la necesidad de demorar la cirugía programada y urgente. Las cifras referentes a la disminución de la actividad quirúrgica y el impacto del periodo aún no se conocen con exactitud. Se estima una mortalidad de hasta un 20% en los pacientes operados con infección peroperatoria por SARS-CoV-2 MÉTODOS: Del 14/03 al 11/05 del 2020 se han recogido los datos de los pacientes ingresados en el servicio de Cirugía General y Digestiva del Hospital Universitari Dr. Josep Trueta de Girona, para analizar el impacto de la pandemia sobre la cirugía electiva y urgente. RESULTADOS: Durante el periodo de ocupación pico de la Unidad de Medicina Intensiva de nuestro centro (con un máximo de 303,8%), la cirugía electiva y la urgente se redujeron un 93,8% y un 72,7%, respectivamente. La mortalidad de los pacientes operados con infección por SARS-CoV-2 en nuestro estudio (n = 10) fue del 10%; las complicaciones fueron del 80% (siendo graves un 30%). CONCLUSIONES: El presente estudio muestra una reducción global de la actividad quirúrgica tanto electiva como urgente durante la pandemia. La mortalidad global de los pacientes operados con infección por SARS-CoV-2 ha sido baja, pero la tasa de complicaciones graves ha sido superior a la global


INTRODUCTION: SARS-CoV-2 pandemic has caused an important impact in our country and elective surgery has been postponed in most cases. There's not known information about the decreasing and impact on surgery. Mortality of surgical patients with SARS-CoV-2 infection is estimated to be around 20%. METHODS: We conducted prospective data recruitment of people inpatient in our Digestive and General Surgery section of Girona's University Hospital Dr. Josep Trueta from 03/14 to 05/11. Our objective is to analyze the impact that SARS-CoV-2 pandemic over elective and urgent surgery. RESULTS: During the peak occupation of our center Intensive Care Unit (303.8%) there was a reduction on elective (93.8%) and urgent (72.7%) surgery. Mortality of patients with SARS-CoV-2 infection who underwent surgery (n=10) is estimated to be a 10%. An 80% of these patients suffer complications (sever complications in 30%). CONCLUSIONS: The actual study shows a global reduction of the surgical activity (elective and urgent) during de SARS-CoV-2 pandemic. Global mortality of patients with SARS-CoV-2 infection are low, but the severe complications have been over the usual


Assuntos
Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Infecções por Coronaviridae/epidemiologia , Pneumonia Viral/epidemiologia , Pandemias , Procedimentos Cirúrgicos Operatórios/estatística & dados numéricos , Emergências , Procedimentos Cirúrgicos Eletivos/estatística & dados numéricos , Infecções por Coronaviridae/mortalidade , Pneumonia Viral/mortalidade , Atenção Terciária à Saúde , Estudos Prospectivos
12.
PLoS Pathog ; 17(4): e1009149, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33878118

RESUMO

The COVID-19 pandemic has demonstrated the serious potential for novel zoonotic coronaviruses to emerge and cause major outbreaks. The immediate animal origin of the causative virus, SARS-CoV-2, remains unknown, a notoriously challenging task for emerging disease investigations. Coevolution with hosts leads to specific evolutionary signatures within viral genomes that can inform likely animal origins. We obtained a set of 650 spike protein and 511 whole genome nucleotide sequences from 222 and 185 viruses belonging to the family Coronaviridae, respectively. We then trained random forest models independently on genome composition biases of spike protein and whole genome sequences, including dinucleotide and codon usage biases in order to predict animal host (of nine possible categories, including human). In hold-one-out cross-validation, predictive accuracy on unseen coronaviruses consistently reached ~73%, indicating evolutionary signal in spike proteins to be just as informative as whole genome sequences. However, different composition biases were informative in each case. Applying optimised random forest models to classify human sequences of MERS-CoV and SARS-CoV revealed evolutionary signatures consistent with their recognised intermediate hosts (camelids, carnivores), while human sequences of SARS-CoV-2 were predicted as having bat hosts (suborder Yinpterochiroptera), supporting bats as the suspected origins of the current pandemic. In addition to phylogeny, variation in genome composition can act as an informative approach to predict emerging virus traits as soon as sequences are available. More widely, this work demonstrates the potential in combining genetic resources with machine learning algorithms to address long-standing challenges in emerging infectious diseases.


Assuntos
Evolução Biológica , Infecções por Coronaviridae/diagnóstico , Infecções por Coronaviridae/virologia , Coronaviridae/patogenicidade , Genoma Viral , Aprendizado de Máquina , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , Infecções por Coronaviridae/genética , Infecções por Coronaviridae/metabolismo , Filogenia , Glicoproteína da Espícula de Coronavírus/genética
13.
J Vet Diagn Invest ; 33(3): 457-468, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33739188

RESUMO

Every day, thousands of samples from diverse populations of animals are submitted to veterinary diagnostic laboratories (VDLs) for testing. Each VDL has its own laboratory information management system (LIMS), with processes and procedures to capture submission information, perform laboratory tests, define the boundaries of test results (i.e., positive or negative), and report results, in addition to internal business and accounting applications. Enormous quantities of data are accumulated and stored within VDL LIMSs. There is a need for platforms that allow VDLs to exchange and share portions of laboratory data using standardized, reliable, and sustainable information technology processes. Here we report concepts and applications for standardization and aggregation of data from swine submissions to multiple VDLs to detect and monitor porcine enteric coronaviruses by RT-PCR. Oral fluids, feces, and fecal swabs were the specimens submitted most frequently for enteric coronavirus testing. Statistical algorithms were used successfully to scan and monitor the overall and state-specific percentage of positive submissions. Major findings revealed a consistently recurrent seasonal pattern, with the highest percentage of positive submissions detected during December-February for porcine epidemic diarrhea virus, porcine deltacoronavirus, and transmissible gastroenteritis virus (TGEV). After 2014, very few submissions tested positive for TGEV. Monitoring VDL data proactively has the potential to signal and alert stakeholders early of significant changes from expected detection. We demonstrate the importance of, and applications for, data organized and aggregated by using LOINC and SNOMED CTs, as well as the use of customized messaging to allow inter-VDL exchange of information.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/isolamento & purificação , Laboratórios/normas , Doenças dos Suínos/virologia , Animais , Teste para COVID-19/veterinária , Infecções por Coronaviridae/diagnóstico , Infecções por Coronaviridae/virologia , Surtos de Doenças , Fezes/virologia , Padrões de Referência , Estações do Ano , Suínos , Doenças dos Suínos/diagnóstico
14.
Cairo; World Health Organization. Regional Office for the Eastern Mediterranean; 2021-03. (WHO-EM/CSR/415/E).
em Inglês | WHO IRIS | ID: who-349418
15.
Cairo; World Health Organization. Regional Office for the Eastern Mediterranean; 2021-02. (WHO-EM/CSR/414/E).
em Inglês | WHO IRIS | ID: who-349419
16.
Biochem Biophys Res Commun ; 538: 24-34, 2021 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-33413979

RESUMO

Two pandemics of respiratory distress diseases associated with zoonotic introductions of the species Severe acute respiratory syndrome-related coronavirus in the human population during 21st century raised unprecedented interest in coronavirus research and assigned it unseen urgency. The two viruses responsible for the outbreaks, SARS-CoV and SARS-CoV-2, respectively, are in the spotlight, and SARS-CoV-2 is the focus of the current fast-paced research. Its foundation was laid down by studies of many corona- and related viruses that collectively form the vast order Nidovirales. Comparative genomics of nidoviruses played a key role in this advancement over more than 30 years. It facilitated the transfer of knowledge from characterized to newly identified viruses, including SARS-CoV and SARS-CoV-2, as well as contributed to the dissection of the nidovirus proteome and identification of patterns of variations between different taxonomic groups, from species to families. This review revisits selected cases of protein conservation and variation that define nidoviruses, illustrates the remarkable plasticity of the proteome during nidovirus adaptation, and asks questions at the interface of the proteome and processes that are vital for nidovirus reproduction and could inform the ongoing research of SARS-CoV-2.


Assuntos
Infecções por Coronaviridae/virologia , Nidovirales/classificação , Nidovirales/genética , Sequência Conservada , Evolução Molecular , Variação Genética , Genômica , Humanos , Filogenia , Proteoma , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , SARS-CoV-2/classificação , SARS-CoV-2/genética , Proteínas Virais
17.
Cairo; World Health Organization. Regional Office for the Eastern Mediterranean; 2021-01. (WHO-EM/CSR/413/E).
em Inglês | WHO IRIS | ID: who-349420
18.
Transbound Emerg Dis ; 68(3): 1097-1110, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32799433

RESUMO

COVID-19 is a highly contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has rapidly spread to 216 countries and territories since first outbreak in December of 2019, posing a substantial economic losses and extraordinary threats to the public health worldwide. Although bats have been suggested as the natural host of SARS-CoV-2, transmission chains of this virus, role of animals during cross-species transmission, and future concerns remain unclear. Diverse animal coronaviruses have extensively been studied since the discovery of avian coronavirus in 1930s. The current article comprehensively reviews and discusses the current understanding about animal coronaviruses and SARS-CoV-2 for their emergence, transmission, zoonotic potential, alteration of tissue/host tropism, evolution, status of vaccines and surveillance. This study aims at providing guidance for control of COVID-19 and preventative strategies for possible future outbreaks of zoonotic coronavirus via cross-species transmission.


Assuntos
COVID-19/virologia , Infecções por Coronaviridae/veterinária , Coronavirus/classificação , SARS-CoV-2/genética , Animais , Infecções por Coronaviridae/virologia , Humanos
19.
Front Immunol ; 12: 804625, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35082791

RESUMO

Porcine hemagglutinating encephalomyelitis virus (PHEV) is a highly neurotropic coronavirus that invades the host central nervous system (CNS) and causes neurological dysfunction. Microglia are key immune cells in the CNS, however, whether and how they response to PHEV infection remains unclear. Herein, microglial activation and proliferation were detected in the CNS of PHEV-infected mice, as along with the proinflammatory response. Moreover, the production of proinflammatory cytokines induced by moderately activated microglia limited viral replication in the early stage of infection. Microglial depletion assays showed that during late infection, excess activation of microglia aggravated neurological symptoms, BBB destruction, and peripheral monocyte/macrophage infiltration into the CNS. Using an in vitro brain slice model, PHEV was identified to specifically and moderately induce microglial activation in the absence of peripheral immune cells infiltration. Consistently, macrophage clearance from circulating blood indicated that peripheral monocytes/macrophages crossing the BBB of mice were responsible for excess activation of microglia and CNS damage in late PHEV infection. Overall, our findings provide evidence supporting a dual role for microglia in the host CNS in response to coronavirus PHEV invasion.


Assuntos
Betacoronavirus 1/imunologia , Barreira Hematoencefálica/imunologia , Infecções por Coronaviridae/imunologia , Macrófagos/imunologia , Microglia/imunologia , Monócitos/imunologia , Animais , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/virologia , Macrófagos/patologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microglia/patologia , Microglia/virologia , Monócitos/patologia , Monócitos/virologia
20.
Pathog Dis ; 79(1)2021 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-33238302

RESUMO

BACKGROUND: biomarkers can be helpful in identifying patients who may profit by explicit treatments or evaluating the reaction to the treatment of specific disease. Finding unique biomarkers in the process of disease could help clinicians in identifying serious disease in the early stage, so as to improve prognosis. OBJECTIVE: these investigations, nonetheless, have made constrained progress. Numerous infections are known to cause intense viral encephalitis (VE) in people which can cause a variable level of meningeal just as parenchymal aggravation. Initial clinical manifestations in most encephalitis are nonspecific, resembling a viral-like illness. However, with disease progression, symptoms can become quite severe and fatal, including prominent cranial hypertension, cognitive problems, cerebral hernia and respiratory failure. Forwards: the clinical and research center discoveries in huge numbers of those viral issues are to a great extent comparable and in this way increasingly explicit biomarkers for indicative and prognostic intentions are justified. These biomarkers are progressively significant in the acknowledgment and treatment of the viral central nervous system (CNS) issue. CONCLUSION: Clinical manifestations have been the indicative approaches for analysis of viral encephalitis. Lots of studies have been endeavored to distinguish progressively objective laboratory-based quantitative CSF biomarkers for VE.


Assuntos
Antivirais/uso terapêutico , Encefalite Viral/tratamento farmacológico , Encefalite Viral/metabolismo , Encefalite Viral/virologia , Vacinas/uso terapêutico , Biomarcadores , Infecções por Coronaviridae/virologia , Encefalite Japonesa/virologia , Infecções por HIV/virologia , Herpes Simples/virologia , Humanos , Influenza Humana/virologia , Prognóstico , Febre do Nilo Ocidental/virologia , Infecção por Zika virus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...