Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.285
Filtrar
1.
J Virol ; 97(1): e0144222, 2023 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-36541803

RESUMO

Pathological effects of apoptosis associated with viral infections of the central nervous system are an important cause of morbidity and mortality. Reovirus is a neurotropic virus that causes apoptosis in neurons, leading to lethal encephalitis in newborn mice. Reovirus-induced encephalitis is diminished in mice with germ line ablation of NF-κB subunit p50. It is not known whether the proapoptotic function of NF-κB is mediated by neural-cell-intrinsic (neural-intrinsic) processes, NF-κB-regulated cytokine production by inflammatory cells, or a combination of both. To determine the contribution of cell type-specific NF-κB signaling in reovirus-induced neuronal injury, we established mice that lack NF-κB p65 expression in neural cells using the Cre/loxP recombination system. Following intracranial inoculation of reovirus, 50% of wild-type (WT) mice succumbed to infection, whereas more than 90% of mice lacking neural cell NF-κB p65 (Nsp65-/-) survived. While viral loads in brains of WT and Nsp65-/- mice were comparable, histological analysis revealed that reovirus antigen-positive areas in the brains of WT mice displayed increased immunoreactivity for cleaved caspase-3, a marker of apoptosis, relative to Nsp65-/- mice. These data suggest that neural-intrinsic NF-κB-dependent factors are essential mediators of reovirus neurovirulence. RNA sequencing analysis of reovirus-infected brain cortices of WT and Nsp65-/- mice suggests that NF-κB activation in neuronal cells upregulates genes involved in innate immunity, inflammation, and cell death following reovirus infection. A better understanding of the contribution of cell type-specific NF-κB-dependent signaling to viral neuropathogenesis could inform development of new therapeutics that target and protect highly vulnerable cell populations. IMPORTANCE Viral encephalitis contributes to illness and death in children and adults worldwide and has limited treatment options. Identifying common host factors upregulated by neurotropic viruses can enhance an understanding of virus-induced neuropathogenesis and aid in development of therapeutics. Although many neurotropic viruses activate NF-κB during infection, mechanisms by which NF-κB regulates viral neuropathogenesis and contributes to viral encephalitis are not well understood. We established mice in which NF-κB expression is ablated in neural tissue to study the function of NF-κB in reovirus neurovirulence and identify genes activated by NF-κB in response to reovirus infection in the central nervous system. Encephalitis following reovirus infection was dampened in mice lacking neural cell NF-κB. Reovirus induced a chemokine profile in the brain that was dependent on NF-κB signaling and was similar to chemokine profiles elicited by other neurotropic viruses. These data suggest common underlying mechanisms of encephalitis caused by neurotropic viruses and potentially shared therapeutic targets.


Assuntos
Encefalite Viral , Neurônios , Infecções por Reoviridae , Reoviridae , Animais , Camundongos , Apoptose/genética , Apoptose/imunologia , Quimiocinas/imunologia , Encefalite Viral/imunologia , Encefalite Viral/virologia , Neurônios/imunologia , NF-kappa B/genética , NF-kappa B/metabolismo , Reoviridae/imunologia , Reoviridae/patogenicidade , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/virologia , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia
2.
Emerg Microbes Infect ; 11(1): 6-13, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34783638

RESUMO

In 2021, three encephalitis cases due to the Borna disease virus 1 (BoDV-1) were diagnosed in the north and east of Germany. The patients were from the states of Thuringia, Saxony-Anhalt, and Lower Saxony. All were residents of known endemic areas for animal Borna disease but without prior diagnosed human cases. Except for one recently detected case in the state of Brandenburg, all >30 notified cases had occurred in, or were linked to, the southern state of Bavaria. Of the three detected cases described here, two infections were acute, while one infection was diagnosed retrospectively from archived brain autopsy tissue samples. One of the acute cases survived, but is permanently disabled. The cases were diagnosed by various techniques (serology, molecular assays, and immunohistology) following a validated testing scheme and adhering to a proposed case definition. Two cases were classified as confirmed BoDV-1 encephalitis, while one case was a probable infection with positive serology and typical brain magnetic resonance imaging, but without molecular confirmation. Of the three cases, one full virus genome sequence could be recovered. Our report highlights the need for awareness of a BoDV-1 etiology in cryptic encephalitis cases in all areas with known animal Borna disease endemicity in Europe, including virus-endemic regions in Austria, Liechtenstein, and Switzerland. BoDV-1 should be actively tested for in acute encephalitis cases with residence or rural exposure history in known Borna disease-endemic areas.


Assuntos
Doença de Borna/diagnóstico , Vírus da Doença de Borna/isolamento & purificação , Encefalite Viral/diagnóstico , Idoso , Animais , Doença de Borna/epidemiologia , Doença de Borna/patologia , Doença de Borna/virologia , Vírus da Doença de Borna/classificação , Vírus da Doença de Borna/genética , Encéfalo/patologia , Encéfalo/virologia , Encefalite Viral/epidemiologia , Encefalite Viral/patologia , Encefalite Viral/virologia , Doenças Endêmicas , Feminino , Alemanha/epidemiologia , Humanos , Masculino , Pessoa de Meia-Idade , Filogenia
3.
Front Immunol ; 12: 753683, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899705

RESUMO

Human parechovirus type 3 (PeV-A3) infection has been recognized as an emerging etiologic factor causing severe nerve disease or sepsis in infants and young children. But the neuropathogenic mechanisms of PeV-A3 remain unknown. To understand the pathogenesis of PeV-A3 infection in the neuronal system, PeV-A3-mediated cytopathic effects were analyzed in human glioblastoma cells and neuroblastoma cells. PeV-A3 induced interferons and inflammatory cytokine expression in these neuronal cells. The pronounced cytopathic effects accompanied with activation of death signaling pathways of apoptosis, autophagy, and pyroptosis were detected. A new experimental disease model of parechovirus encephalitis was established. In the disease model, intracranial inoculation with PeV-A3 in C57BL/6 neonatal mice showed body weight loss, hindlimb paralysis, and approximately 20% mortality. PeV-A3 infection in the hippocampus and cortex regions of the neonatal mouse brain was revealed. Mechanistic assay supported the in vitro results, indicating detection of PeV-A3 replication, inflammatory cytokine expression, and death signaling transduction in mouse brain tissues. These in vitro and in vivo studies revealed that the activation of death signaling and inflammation responses is involved in PeV-A3-mediated neurological disorders. The present results might account for some of the PeV-A3-associated clinical manifestations.


Assuntos
Efeito Citopatogênico Viral , Modelos Animais de Doenças , Encefalite Viral/metabolismo , Parechovirus/patogenicidade , Infecções por Picornaviridae/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Autofagia , Linhagem Celular Tumoral , Córtex Cerebral/virologia , Chlorocebus aethiops , Citocinas/biossíntese , Citocinas/genética , Encefalite Viral/patologia , Encefalite Viral/virologia , Glioblastoma/patologia , Hipocampo/virologia , Humanos , Inflamação , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Interferon Tipo I/farmacologia , Interferons/biossíntese , Interferons/genética , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Neuroblastoma/patologia , Parechovirus/efeitos dos fármacos , Parechovirus/fisiologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Piroptose , Células Vero , Replicação Viral/efeitos dos fármacos , Interferon lambda
4.
Front Immunol ; 12: 786953, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925368

RESUMO

Lyssaviruses cause the disease rabies, which is a fatal encephalitic disease resulting in approximately 59,000 human deaths annually. The prototype species, rabies lyssavirus, is the most prevalent of all lyssaviruses and poses the greatest public health threat. In Africa, six confirmed and one putative species of lyssavirus have been identified. Rabies lyssavirus remains endemic throughout mainland Africa, where the domestic dog is the primary reservoir - resulting in the highest per capita death rate from rabies globally. Rabies is typically transmitted through the injection of virus-laden saliva through a bite or scratch from an infected animal. Due to the inhibition of specific immune responses by multifunctional viral proteins, the virus usually replicates at low levels in the muscle tissue and subsequently enters the peripheral nervous system at the neuromuscular junction. Pathogenic rabies lyssavirus strains inhibit innate immune signaling and induce cellular apoptosis as the virus progresses to the central nervous system and brain using viral protein facilitated retrograde axonal transport. Rabies manifests in two different forms - the encephalitic and the paralytic form - with differing clinical manifestations and survival times. Disease symptoms are thought to be due mitochondrial dysfunction, rather than neuronal apoptosis. While much is known about rabies, there remain many gaps in knowledge about the neuropathology of the disease. It should be emphasized however, that rabies is vaccine preventable and dog-mediated human rabies has been eliminated in various countries. The global elimination of dog-mediated human rabies in the foreseeable future is therefore an entirely feasible goal.


Assuntos
Encefalite Viral/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Zoonoses Virais/imunologia , África/epidemiologia , Animais , Cães , Encefalite Viral/epidemiologia , Encefalite Viral/transmissão , Encefalite Viral/virologia , Doenças Endêmicas , Humanos , Imunidade Inata , Raiva/epidemiologia , Raiva/transmissão , Raiva/virologia , Saliva/virologia , Zoonoses Virais/epidemiologia , Zoonoses Virais/transmissão , Zoonoses Virais/virologia , Replicação Viral/imunologia
5.
Viruses ; 13(12)2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34960633

RESUMO

The environment of the central nervous system (CNS) represents a double-edged sword in the context of viral infections. On the one hand, the infectious route for viral pathogens is restricted via neuroprotective barriers; on the other hand, viruses benefit from the immunologically quiescent neural environment after CNS entry. Both the herpes simplex virus (HSV) and the rabies virus (RABV) bypass the neuroprotective blood-brain barrier (BBB) and successfully enter the CNS parenchyma via nerve endings. Despite the differences in the molecular nature of both viruses, each virus uses retrograde transport along peripheral nerves to reach the human CNS. Once inside the CNS parenchyma, HSV infection results in severe acute inflammation, necrosis, and hemorrhaging, while RABV preserves the intact neuronal network by inhibiting apoptosis and limiting inflammation. During RABV neuroinvasion, surveilling glial cells fail to generate a sufficient type I interferon (IFN) response, enabling RABV to replicate undetected, ultimately leading to its fatal outcome. To date, we do not fully understand the molecular mechanisms underlying the activation or suppression of the host inflammatory responses of surveilling glial cells, which present important pathways shaping viral pathogenesis and clinical outcome in viral encephalitis. Here, we compare the innate immune responses of glial cells in RABV- and HSV-infected CNS, highlighting different viral strategies of neuroprotection or Neuroinflamm. in the context of viral encephalitis.


Assuntos
Encefalite Viral/imunologia , Herpes Simples/imunologia , Imunidade Inata , Inflamação , Vírus da Raiva/imunologia , Raiva/imunologia , Simplexvirus/imunologia , Animais , Astrócitos/imunologia , Astrócitos/virologia , Barreira Hematoencefálica/virologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Encefalite Viral/virologia , Herpes Simples/virologia , Humanos , Microglia/imunologia , Microglia/virologia , Neuroglia/imunologia , Neuroglia/virologia , Raiva/virologia , Transdução de Sinais
6.
Microbiol Spectr ; 9(3): e0148721, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34817277

RESUMO

Viral diseases of the central nervous system (CNS) represent a major global health concern. Difficulties in treating these diseases are caused mainly by the biological tissues and barriers, which hinder the transport of drugs into the CNS. To counter this, a nanobody-mediated virus-targeting drug delivery platform (SWCNTs-P-A-Nb) is constructed for CNS viral disease therapy. Viral encephalopathy and retinopathy (VER), caused by nervous necrosis virus (NNV), is employed as a disease model. SWCNTs-P-A-Nb is successfully constructed by employing single-walled carbon nanotubes, amantadine, and NNV-specific nanobody (NNV-Nb) as the nanocarrier, anti-NNV drug, and targeting ligand, respectively. Results showed that SWCNTs-P-A-Nb has a good NNV-targeting ability in vitro and in vivo, improving the specific distribution of amantadine in NNV-infected sites under the guidance of NNV-Nb. SWCNTs-P-F-A-Nb can pass through the muscle and gill and be excreted by the kidney. SWCNTs-P-A-Nb can transport amantadine in a fast manner and prolong the action time, improving the anti-NNV activity of amantadine. Results so far have indicated that the nanobody-mediated NNV-targeting drug delivery platform is an effective method for VER therapy, providing new ideas and technologies for control of the CNS viral diseases. IMPORTANCE CNS viral diseases have resulted in many deadly epidemics throughout history and continue to pose one of the greatest threats to public health. Drug therapy remains challenging due to the complex structure and relative impermeability of the biological tissues and barriers. Therefore, development in the intelligent drug delivery platform is highly desired for CNS viral disease therapy. In the study, a nanobody-mediated virus-targeting drug delivery platform is constructed to explore the potential application of targeted therapy in CNS viral diseases. Our findings hold great promise for the application of targeted drug delivery in CNS viral disease therapy.


Assuntos
Amantadina/farmacologia , Viroses do Sistema Nervoso Central/terapia , Viroses do Sistema Nervoso Central/veterinária , Sistemas de Liberação de Medicamentos/métodos , Nodaviridae/efeitos dos fármacos , Anticorpos de Domínio Único/farmacologia , Animais , Antivirais/farmacologia , Linhagem Celular , Sistema Nervoso Central/virologia , Encefalite Viral/terapia , Encefalite Viral/virologia , Peixes , Nanotubos de Carbono , Nodaviridae/imunologia , Perciformes/virologia , Anticorpos de Domínio Único/imunologia
7.
Pak J Pharm Sci ; 34(5): 1809-1812, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34803019

RESUMO

We describe for the first time an child who demonstrated Mild encephalitis/encephalopathy with a reversible splenial lesion (MERS) after mumps infection in China. In this report, a 12-year-old boy came to Children's Hospital Affiliated to Zhengzhou University due to fever, swelling and pain under the earlobe for 4 days, and headache and vomiting for half of a day. Laboratory examinations showed a blood sodium level of 125mmol/L, both the Immunoglobulin M and Polymerase Chain Reaction results for the serum mumps virus were positive. Brain Magnetic Resonance Imaging (MRI) showed slight hypointense on T1 weighted images, hyperintense on T2-weighted images, fluid attenuated inversion recovery, diffusion-weighted images in the splenium of the corpus callosum indicative of MERS. On the 8th day, the patient no longer had swelling and pain around the parotid salivary glands, the sodium levels returned to normal. Onset of 14th d, follow-up brain MRI did not reveal any abnormalities. The case given to us indicates that MERS should be considered when patients after mumps infection presents with neurological symptoms and MRI should be performed to evaluate the splenium of the corpus callosum.


Assuntos
Corpo Caloso/patologia , Encefalite Viral/patologia , Caxumba/complicações , Anti-Inflamatórios/uso terapêutico , Antivirais/uso terapêutico , Criança , China , Diuréticos Osmóticos/uso terapêutico , Encefalite Viral/virologia , Humanos , Masculino , Manitol/uso terapêutico , Metilprednisolona/uso terapêutico , Ribavirina/uso terapêutico
8.
Cells ; 10(11)2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34831405

RESUMO

Infection with flaviviruses causes mild to severe diseases, including viral hemorrhagic fever, vascular shock syndrome, and viral encephalitis. Several animal models explore the pathogenesis of viral encephalitis, as shown by neuron destruction due to neurotoxicity after viral infection. While neuronal cells are injuries caused by inflammatory cytokine production following microglial/macrophage activation, the blockade of inflammatory cytokines can reduce neurotoxicity to improve the survival rate. This study investigated the involvement of macrophage phenotypes in facilitating CNS inflammation and neurotoxicity during flavivirus infection, including the Japanese encephalitis virus, dengue virus (DENV), and Zika virus. Mice infected with different flaviviruses presented encephalitis-like symptoms, including limbic seizure and paralysis. Histology indicated that brain lesions were identified in the hippocampus and surrounded by mononuclear cells. In those regions, both the infiltrated macrophages and resident microglia were significantly increased. RNA-seq analysis showed the gene profile shifting toward type 1 macrophage (M1) polarization, while M1 markers validated this phenomenon. Pharmacologically blocking C-C chemokine receptor 2 and tumor necrosis factor-α partly retarded DENV-induced M1 polarization. In summary, flavivirus infection, such as JEV and DENV, promoted type 1 macrophage polarization in the brain associated with encephalitic severity.


Assuntos
Polaridade Celular , Vírus da Dengue/fisiologia , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Viral/patologia , Encefalite Viral/virologia , Macrófagos/patologia , Índice de Gravidade de Doença , Animais , Animais Lactentes , Linhagem Celular , Modelos Animais de Doenças , Encefalite Japonesa/imunologia , Encefalite Japonesa/patologia , Encefalite Japonesa/virologia , Encefalite Viral/imunologia , Hipocampo/patologia , Inflamação/patologia , Camundongos Endogâmicos ICR , Neurotoxinas/toxicidade , Receptores CCR2/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
9.
Int J Mol Sci ; 22(21)2021 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-34769052

RESUMO

A wide range of neurological manifestations have been associated with the development of COVID-19 following SARS-CoV-2 infection. However, the etiology of the neurological symptomatology is still largely unexplored. Here, we used state-of-the-art multiplexed immunostaining of human brains (n = 6 COVID-19, median age = 69.5 years; n = 7 control, median age = 68 years) and demonstrated that expression of the SARS-CoV-2 receptor ACE2 is restricted to a subset of neurovascular pericytes. Strikingly, neurological symptoms were exclusive to, and ubiquitous in, patients that exhibited moderate to high ACE2 expression in perivascular cells. Viral dsRNA was identified in the vascular wall and paralleled by perivascular inflammation, as signified by T cell and macrophage infiltration. Furthermore, fibrinogen leakage indicated compromised integrity of the blood-brain barrier. Notably, cerebrospinal fluid from additional 16 individuals (n = 8 COVID-19, median age = 67 years; n = 8 control, median age = 69.5 years) exhibited significantly lower levels of the pericyte marker PDGFRß in SARS-CoV-2-infected cases, indicative of disrupted pericyte homeostasis. We conclude that pericyte infection by SARS-CoV-2 underlies virus entry into the privileged central nervous system space, as well as neurological symptomatology due to perivascular inflammation and a locally compromised blood-brain barrier.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Encéfalo/virologia , COVID-19/fisiopatologia , Encefalite Viral/virologia , Pericitos/virologia , Enzima de Conversão de Angiotensina 2/genética , Animais , Barreira Hematoencefálica , Encéfalo/patologia , COVID-19/etiologia , Estudos de Casos e Controles , Encefalite Viral/patologia , Fibrinogênio/metabolismo , Humanos , Imuno-Histoquímica/métodos , Camundongos , Pericitos/metabolismo , Pericitos/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/líquido cefalorraquidiano
10.
Trop Biomed ; 38(3): 435-445, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34608117

RESUMO

Ever since the first reported case series on SARS-CoV-2-induced neurological manifestation in Wuhan, China in April 2020, various studies reporting similar as well as diverse symptoms of COVID-19 infection relating to the nervous system were published. Since then, scientists started to uncover the mechanism as well as pathophysiological impacts it has on the current understanding of the disease. SARS-CoV-2 binds to the ACE2 receptor which is present in certain parts of the body which are responsible for regulating blood pressure and inflammation in a healthy system. Presence of the receptor in the nasal and oral cavity, brain, and blood allows entry of the virus into the body and cause neurological complications. The peripheral and central nervous system could also be invaded directly in the neurogenic or hematogenous pathways, or indirectly through overstimulation of the immune system by cytokines which may lead to autoimmune diseases. Other neurological implications such as hypoxia, anosmia, dysgeusia, meningitis, encephalitis, and seizures are important symptoms presented clinically in COVID-19 patients with or without the common symptoms of the disease. Further, patients with higher severity of the SARS-CoV-2 infection are also at risk of retaining some neurological complications in the long-run. Treatment of such severe hyperinflammatory conditions will also be discussed, as well as the risks they may pose to the progression of the disease. For this review, articles pertaining information on the neurological manifestation of SARS-CoV-2 infection were gathered from PubMed and Google Scholar using the search keywords "SARS-CoV-2", "COVID-19", and "neurological dysfunction". The findings of the search were filtered, and relevant information were included.


Assuntos
COVID-19/patologia , Sistema Nervoso Central/patologia , Doenças do Sistema Nervoso/virologia , Sistema Nervoso Periférico/patologia , Enzima de Conversão de Angiotensina 2/metabolismo , Anosmia/virologia , Sistema Nervoso Central/virologia , Disgeusia/virologia , Encefalite Viral/virologia , Humanos , Meningite Viral/virologia , Doenças do Sistema Nervoso/patologia , Sistema Nervoso Periférico/virologia , SARS-CoV-2 , Convulsões/virologia
11.
Ocul Immunol Inflamm ; 29(6): 1225-1233, 2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34648417

RESUMO

The novel pandemic coronavirus disease 2019 (COVID-19) leading to health and economic problems worldwide is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although COVID-19 mainly occurs as a lower respiratory tract infection, there is multiorgan involvement in infected patients. The disease is transmitted from person to person through air droplets or contact with contaminated surfaces. SARS-CoV-2 leads to this systemic involvement by attaching to angiotensin-converting enzyme 2 (ACE2) receptors located on several human cells. Since SARS-CoV-2 RNA has been found in tears of infected patients, ocular surface may allow the virus to transmit to nasopharynx via the nasolacrimal duct. This narrative review aims to sum up all segmental ocular complications, ocular adverse effects of COVID-19 treatment, and preventive measures suggested to minimize the SARS-CoV-2 transmission between patients and ophthalmologists by reviewing currently available literature.


Assuntos
COVID-19/diagnóstico , Infecções Oculares Virais/diagnóstico , SARS-CoV-2 , Lágrimas/virologia , COVID-19/prevenção & controle , COVID-19/virologia , Teste de Ácido Nucleico para COVID-19 , Conjuntivite Viral/diagnóstico , Conjuntivite Viral/prevenção & controle , Conjuntivite Viral/virologia , Encefalite Viral/diagnóstico , Encefalite Viral/prevenção & controle , Encefalite Viral/virologia , Infecções Oculares Virais/prevenção & controle , Infecções Oculares Virais/virologia , Humanos , Medicina Preventiva/métodos , Doenças Retinianas/diagnóstico , Doenças Retinianas/prevenção & controle , Doenças Retinianas/virologia , SARS-CoV-2/patogenicidade
12.
Viruses ; 13(9)2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34578440

RESUMO

Porcine astrovirus type 3 (PoAstV3) has been previously identified as a cause of polioencephalomyelitis in swine and continues to cause disease in the US swine industry. Herein, we describe the characterization of both untranslated regions, frameshifting signal, putative genome-linked virus protein (VPg) and conserved antigenic epitopes of several novel PoAstV3 genomes. Twenty complete coding sequences (CDS) were obtained from 32 diagnostic cases originating from 11 individual farms/systems sharing a nucleotide (amino acid) percent identity of 89.74-100% (94.79-100%), 91.9-100% (96.3-100%) and 90.71-100% (93.51-100%) for ORF1a, ORF1ab and ORF2, respectively. Our results indicate that the 5'UTR of PoAstV3 is highly conserved highlighting the importance of this region in translation initiation while their 3'UTR is moderately conserved among strains, presenting alternative configurations including multiple putative protein binding sites and pseudoknots. Moreover, two predicted conserved antigenic epitopes were identified matching the 3' termini of VP27 of PoAstV3 USA strains. These epitopes may aid in the design and development of vaccine components and diagnostic assays useful to control outbreaks of PoAstV3-associated CNS disease. In conclusion, this is the first analysis predicting the structure of important regulatory motifs of neurotropic mamastroviruses, which differ from those previously described in human astroviruses.


Assuntos
Infecções por Astroviridae/veterinária , Genoma Viral , Mamastrovirus/genética , Fases de Leitura Aberta , Proteínas Virais/genética , Animais , Antígenos Virais , Infecções por Astroviridae/virologia , Encefalite Viral/veterinária , Encefalite Viral/virologia , Epitopos , Mamastrovirus/imunologia , Mamastrovirus/metabolismo , Conformação de Ácido Nucleico , Filogenia , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Suínos , Doenças dos Suínos/virologia , Regiões não Traduzidas , Proteínas Virais/química , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
13.
J Virol ; 95(23): e0150621, 2021 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-34495703

RESUMO

Rift Valley fever virus (RVFV) is an arbovirus found throughout Africa. It causes disease that is typically mild and self-limiting; however, some infected individuals experience severe manifestations, including hepatitis, encephalitis, or even death. Reports of RVFV encephalitis are notable among immunosuppressed individuals, suggesting a role for adaptive immunity in preventing this severe complication. This phenomenon has been modeled in C57BL/6 mice depleted of CD4 T cells prior to infection with DelNSs RVFV (RVFV containing a deletion of nonstructural protein NSs), resulting in late-onset encephalitis accompanied by high levels of viral RNA in the brain in 30% of animals. In this study, we sought to define the specific type(s) of CD4 T cells that mediate protection from RVFV encephalitis. The viral epitopes targeted by CD4 and CD8 T cells were defined in C57BL/6 mice, and tetramers for both CD4 and CD8 T cells were generated. RVFV-specific CD8 T cells were expanded and of a cytotoxic and proliferating phenotype in the liver following infection. RVFV-specific CD4 T cells were identified in the liver and spleen following infection and phenotyped as largely Th1 or Tfh subtypes. Knockout mice lacking various aspects of pathways important in Th1 and Tfh development and function were used to demonstrate that T-bet, CD40, CD40L, and major histocompatibility complex class II (MHC-II) mediated protection from RVFV encephalitis, while gamma interferon (IFN-γ) and interleukin-12 (IL-12) were dispensable. Virus-specific antibody responses correlated with protection from encephalitis in all mouse strains, suggesting that Tfh/B cell interactions modulate clinical outcome in this model. IMPORTANCE The prevention of RVFV encephalitis requires intact adaptive immunity. In this study, we developed reagents to detect RVFV-specific T cells and provide evidence for Tfh cells and CD40/CD40L interactions as critical mediators of this protection.


Assuntos
Antígenos CD40 , Ligante de CD40 , Encefalite Viral/prevenção & controle , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/imunologia , Vírus da Febre do Vale do Rift/fisiologia , Linfócitos T/imunologia , África , Animais , Formação de Anticorpos , Linfócitos B/imunologia , Encéfalo/virologia , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Encefalite Viral/imunologia , Encefalite Viral/virologia , Epitopos , Feminino , Fígado/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
14.
Sci Rep ; 11(1): 17751, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493781

RESUMO

Enterovirus-A71 (EV-A71) associated Hand, foot and mouth disease (HFMD) is a highly contagious viral infection affecting children in Asia-Pacific region and has become a major threat to public health. Although several EV-A71 genotypes (C, D, and G) were isolated in India in recent years, no recognizable outbreak of EV-A71 caused HFMD, Acute Flaccid paralysis (AFP) or encephalitis have been reported so far. It is essential to study the pathogenicity or cell tropism of these Indian isolates in order to understand their tendency to cause disease. We investigated the susceptibility and cytokine responses of indigenous EV-A71 genotypes (D and G) isolated from cases of AFP and genotype C viruses isolated from cases of HFMD and encephalitis, in human cells in-vitro. Although all three EV-A71 genotypes could infect and replicate in human muscle and neuronal cells, the genotype D virus showed a delayed response in human neuronal cells. Quantification of cytokine secretion in response to these isolates followed by confirmation with gene expression assays in human neuronal cells revealed significantly higher secretion of pro-inflammatory cytokines TNF-α IL-8, IL-6, IP-10 (p < 0.001) in G genotype infected cells as compared to pathogenic C genotypes whereas the genotype D virus could not induce any of the inflammatory cytokines. These findings will help to better understand the host response to indigenous EV-A71 genotypes for management of future EV-A71 outbreaks in India, if any.


Assuntos
Citocinas/biossíntese , Enterovirus Humano A/patogenicidade , Doença de Mão, Pé e Boca/virologia , Neurônios/virologia , Doença Aguda , Adulto , Linhagem Celular Tumoral , Criança , Citocinas/genética , Efeito Citopatogênico Viral , Surtos de Doenças , Suscetibilidade a Doenças , Encefalite Viral/epidemiologia , Encefalite Viral/virologia , Enterovirus Humano A/classificação , Enterovirus Humano A/genética , Enterovirus Humano A/isolamento & purificação , Feminino , Regulação Viral da Expressão Gênica , Genótipo , Doença de Mão, Pé e Boca/epidemiologia , Humanos , Índia/epidemiologia , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Paraplegia/epidemiologia , Paraplegia/virologia , Tropismo Viral
16.
J Stroke Cerebrovasc Dis ; 30(9): 105915, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34217071

RESUMO

We report the case of a 35-year-old male with COVID-19 encephalitis presenting as a stroke mimic with sudden-onset expressive and receptive dysphasia, mild confusion and right arm incoordination. The patient received thrombolysis for a suspected ischaemic stroke, but later became febrile and SARS-CoV-2 was detected in cerebrospinal fluid. Electroencephalography demonstrated excess in slow waves, but neuroimaging was reported as normal. Respiratory symptoms were absent throughout and nasopharyngeal swab was negative for SARS-CoV-2. At the most recent follow-up, the patient had made a full neurological recovery. Clinicians should therefore consider testing for SARS-CoV-2 in CSF in patients who present with acute focal neurology, confusion and fever during the pandemic, even when there is no evidence of respiratory infection.


Assuntos
Teste de Ácido Nucleico para COVID-19 , COVID-19/diagnóstico , Encefalite Viral/diagnóstico , AVC Isquêmico/diagnóstico , RNA Viral/líquido cefalorraquidiano , SARS-CoV-2/genética , Adulto , COVID-19/líquido cefalorraquidiano , COVID-19/virologia , Diagnóstico Diferencial , Eletroencefalografia , Encefalite Viral/líquido cefalorraquidiano , Encefalite Viral/virologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Valor Preditivo dos Testes , Tomografia Computadorizada por Raios X
17.
Viruses ; 13(7)2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34201545

RESUMO

Astrovirus infections are among the main causes of diarrhea in children, but their significance for animal health has remained underestimated and largely unknown. This is changing due to the increasing amount of newly identified neurotropic astroviruses in cases of nonsuppurative encephalitis and neurological disease in humans, pigs, ruminant species and minks. Neurological cases in ruminants and humans usually occur sporadically and as isolated cases. This contrasts with the situation in pigs and minks, in which diseases associated with neurotropic astroviruses are endemic and occur on the herd level. Affected animals show neurological signs such as mild ataxia to tetraplegia, loss of orientation or trembling, and the outcome is often fatal. Non-suppurative inflammation with perivascular cuffing, gliosis and neuronal necrosis are typical histological lesions of astrovirus encephalitis. Since astroviruses primarily target the gastrointestinal tract, it is assumed that they infect the brain through the circulatory system or retrograde following the nerves. The phylogenetic analysis of neurotropic astroviruses has revealed that they are genetically closely related, suggesting the presence of viral determinants for tissue tropism and neuroinvasion. In this review, we summarize the current knowledge on neurotropic astrovirus infections in animals and propose future research activities.


Assuntos
Infecções por Astroviridae/veterinária , Infecções por Astroviridae/virologia , Astroviridae/isolamento & purificação , Doenças do Sistema Nervoso/veterinária , Doenças do Sistema Nervoso/virologia , Animais , Astroviridae/classificação , Astroviridae/genética , Infecções por Astroviridae/diagnóstico , Infecções por Astroviridae/patologia , Encéfalo/patologia , Encéfalo/virologia , Encefalite Viral/diagnóstico , Encefalite Viral/patologia , Encefalite Viral/veterinária , Encefalite Viral/virologia , Genoma Viral , Doenças do Sistema Nervoso/diagnóstico , Doenças do Sistema Nervoso/patologia , Filogenia
18.
Sci Immunol ; 6(60)2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34172587

RESUMO

Viral encephalitis initiates a series of immunological events in the brain that can lead to brain damage and death. Astrocytes express IFN-ß in response to neurotropic infection, whereas activated microglia produce proinflammatory cytokines and accumulate at sites of infection. Here, we observed that neurotropic vesicular stomatitis virus (VSV) infection causes recruitment of leukocytes into the central nervous system (CNS), which requires MyD88, an adaptor of Toll-like receptor and interleukin-1 receptor signaling. Infiltrating leukocytes, and in particular CD8+ T cells, protected against lethal VSV infection of the CNS. Reconstitution of MyD88, specifically in neurons, restored chemokine production in the olfactory bulb as well as leukocyte recruitment into the infected CNS and enhanced survival. Comparative analysis of the translatome of neurons and astrocytes verified neurons as the critical source of chemokines, which regulated leukocyte infiltration of the infected brain and affected survival.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Quimiocinas/metabolismo , Encefalite Viral/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Infecções por Rhabdoviridae/imunologia , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Modelos Animais de Doenças , Encefalite Viral/patologia , Encefalite Viral/virologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Neurônios/metabolismo , Bulbo Olfatório/citologia , Bulbo Olfatório/imunologia , Bulbo Olfatório/patologia , Bulbo Olfatório/virologia , Infecções por Rhabdoviridae/patologia , Infecções por Rhabdoviridae/virologia , Transdução de Sinais/imunologia , Vesiculovirus/imunologia
19.
Methods Mol Biol ; 2311: 109-130, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34033080

RESUMO

The complexity of the central nervous system (CNS) is not recapitulated in cell culture models. Thin slicing and subsequent culture of CNS tissue has become a valued means to study neuronal and glial biology within the context of the physiologically relevant tissue milieu. Modern membrane-interface slice culturing methodology allows for straightforward access to both CNS tissue and feeding medium, enabling experimental manipulations and analyses that would otherwise be impossible in vivo. CNS slices can be successfully maintained in culture for up to several weeks for investigation of evolving pathology and long-term intervention in models of chronic neurologic disease.Herein, membrane-interface slice culture models for studying viral encephalitis and myelitis are detailed, with emphasis on the use of these models for investigation of pathogenesis and evaluation of novel treatment strategies. We describe techniques to (1) generate brain and spinal cord slices from rodent donors, (2) virally infect slices, (3) monitor viral replication, (4) assess virally induced injury/apoptosis, (5) characterize "CNS-specific" cytokine production, and, (6) treat slices with cytokines/pharmaceuticals. Although our focus is on CNS viral infection, we anticipate that the described methods can be adapted to address a wide range of investigations within the fields of neuropathology, neuroimmunology, and neuropharmacology.


Assuntos
Encéfalo/virologia , Encefalite Viral/virologia , Mielite/virologia , Medula Espinal/virologia , Animais , Animais Recém-Nascidos , Antivirais/farmacologia , Apoptose , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Citocinas/metabolismo , Encefalite Viral/tratamento farmacológico , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Interações Hospedeiro-Patógeno , Mediadores da Inflamação/metabolismo , Camundongos , Mielite/tratamento farmacológico , Mielite/metabolismo , Mielite/patologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Técnicas de Cultura de Tecidos , Replicação Viral
20.
Front Immunol ; 12: 667478, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34025669

RESUMO

Viral encephalitis is the most common cause of encephalitis. It is responsible for high morbidity rates, permanent neurological sequelae, and even high mortality rates. The host immune response plays a critical role in preventing or clearing invading pathogens, especially when effective antiviral treatment is lacking. However, due to blockade of the blood-brain barrier, it remains unclear how peripheral immune cells contribute to the fight against intracerebral viruses. Here, we report that peripheral injection of an antibody against human Tim-3, an immune checkpoint inhibitor widely expressed on immune cells, markedly attenuated vesicular stomatitis virus (VSV) encephalitis, marked by decreased mortality and improved neuroethology in mice. Peripheral injection of Tim-3 antibody enhanced the recruitment of immune cells to the brain, increased the expression of major histocompatibility complex-I (MHC-I) on macrophages, and as a result, promoted the activation of VSV-specific CD8+ T cells. Depletion of macrophages abolished the peripheral injection-mediated protection against VSV encephalitis. Notably, for the first time, we found a novel post-translational modification of MHC-I by Tim-3, wherein, by enhancing the expression of MARCH9, Tim-3 promoted the proteasome-dependent degradation of MHC-I via K48-linked ubiquitination in macrophages. These results provide insights into the immune response against intracranial infections; thus, manipulating the peripheral immune cells with Tim-3 antibody to fight viruses in the brain may have potential applications for combating viral encephalitis.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Neutralizantes/administração & dosagem , Células Apresentadoras de Antígenos/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encefalite Viral/prevenção & controle , Receptor Celular 2 do Vírus da Hepatite A/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Infecções por Rhabdoviridae/prevenção & controle , Vesiculovirus/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/virologia , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Chlorocebus aethiops , Modelos Animais de Doenças , Encefalite Viral/imunologia , Encefalite Viral/metabolismo , Encefalite Viral/virologia , Células HEK293 , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Injeções Intraperitoneais , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Células RAW 264.7 , Infecções por Rhabdoviridae/imunologia , Infecções por Rhabdoviridae/metabolismo , Infecções por Rhabdoviridae/virologia , Ubiquitinação , Células Vero , Vesiculovirus/patogenicidade , Carga Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...