Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.290
Filter
1.
Virol J ; 21(1): 102, 2024 05 02.
Article in English | MEDLINE | ID: mdl-38698421

ABSTRACT

Human parechovirus, a member of the Picornaviridae family (PeVs), can lead to severe infections, including severe meningitis, meningoencephalitis, and sepsis-like syndrome. We report a case of human parechovirus-related encephalitis in a 52-year-old woman diagnosed with glioblastoma multiforme. She underwent surgical resection in June 2022. Unfortunately, her disease recurred, and she underwent a second resection in August 2022, followed by radiation therapy and Temozolomide therapy. She presented to the hospital with acute confusion followed by seizures, necessitating intubation for airway support. A cerebrospinal fluid (CSF) sample was obtained and processed using the Biofire FilmArray, which reported the detection of HSV-1. Despite being on Acyclovir, the patient did not show signs of improvement. Consequently, a second CSF sample was obtained and sent for next-generation sequencing (NGS), which returned a positive result for Parechovirus. In this presented case, the patient exhibited symptoms of an unknown infectious cause. The utilization of NGS and metagenomic analysis helped identify Parechovirus as the primary pathogen present, in addition to previously identified HSV. This comprehensive approach facilitated a thorough assessment of the underlying infection and guided targeted treatment. In conclusion, the application of NGS techniques and metagenomic analysis proved instrumental in identifying the root cause of the infection.


Subject(s)
Immunocompromised Host , Parechovirus , Picornaviridae Infections , Humans , Female , Middle Aged , Picornaviridae Infections/virology , Picornaviridae Infections/diagnosis , Parechovirus/genetics , Parechovirus/isolation & purification , Parechovirus/classification , Saudi Arabia , High-Throughput Nucleotide Sequencing , Glioblastoma/virology , Metagenomics , Encephalitis, Viral/virology , Encephalitis, Viral/diagnosis , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/isolation & purification , Hospitalization
2.
Emerg Microbes Infect ; 13(1): 2350168, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38687703

ABSTRACT

ABSTRACTBorna disease virus 1 (BoDV-1) was just recently shown to cause predominantly fatal encephalitis in humans. Despite its rarity, bornavirus encephalitis (BVE) can be considered a model disease for encephalitic infections caused by neurotropic viruses and understanding its pathomechanism is of utmost relevance. Aim of this study was to compare the extent and distribution pattern of cerebral inflammation with the clinical course of disease, and individual therapeutic procedures. For this, autoptic brain material from seven patients with fatal BVE was included in this study. Tissue was stained immunohistochemically for pan-lymphocytic marker CD45, the nucleoprotein of BoDV-1, as well as glial marker GFAP and microglial marker Iba1. Sections were digitalized and counted for CD45-positive and BoDV-1-positive cells. For GFAP and Iba1, a semiquantitative score was determined. Furthermore, detailed information about the individual clinical course and therapy were retrieved and summarized in a standardized way. Analysis of the distribution of lymphocytes shows interindividual patterns. In contrast, when looking at the BoDV-1-positive glial cells and neurons, a massive viral involvement in the brain stem was noticeable. Three of the seven patients received early high-dose steroids, which led to a significantly lower lymphocytic infiltration of the central nervous tissue and a longer survival compared to the patients who were treated with steroids later in the course of disease. This study highlights the potential importance of early high-dose immunosuppressive therapy in BVE. Our findings hint at a promising treatment option which should be corroborated in future observational or prospective therapy studies.ABBREVIATIONS: BoDV-1: Borna disease virus 1; BVE: bornavirus encephalitis; Cb: cerebellum; CNS: central nervous system; FL: frontal lobe; GFAP: glial fibrillary acid protein; Hc: hippocampus; Iba1: ionized calcium-binding adapter molecule 1; Iba1act: general activation of microglial cells; Iba1nod: formation of microglial nodules; IL: insula; Me: mesencephalon; Mo: medulla oblongata; OL: occipital lobe; pASS: per average of 10 screenshots; patearly: patients treated with early high dose steroid shot; patlate: patients treated with late or none high dose steroid shot; Po: pons; So: stria olfactoria; Str: striatum.


Subject(s)
Brain , Humans , Male , Female , Brain/virology , Brain/immunology , Borna Disease/drug therapy , Borna Disease/virology , Lymphocytes/immunology , Microfilament Proteins/metabolism , Leukocyte Common Antigens/metabolism , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Immunosuppression Therapy , Borna disease virus/physiology , Encephalitis, Viral/drug therapy , Encephalitis, Viral/virology , Encephalitis, Viral/immunology , Neuroglia/virology , Neuroglia/metabolism
3.
Eur J Clin Microbiol Infect Dis ; 43(5): 863-873, 2024 May.
Article in English | MEDLINE | ID: mdl-38438704

ABSTRACT

PURPOSE: Investigation of undiagnosed cases of infectious neurological diseases, especially in the paediatric population, remains a challenge. This study aimed to enhance understanding of viruses in CSF from children with clinically diagnosed meningitis and/or encephalitis (M/ME) of unknown aetiology using shotgun sequencing enhanced by hybrid capture (HCSS). METHODS: A single-centre prospective study was conducted at Sant Joan de Déu University Hospital, Barcelona, involving 40 M/ME episodes of unknown aetiology, recruited from May 2021 to July 2022. All participants had previously tested negative with the FilmArray Meningitis/Encephalitis Panel. HCSS was used to detect viral nucleic acid in the patients' CSF. Sequencing was performed on Illumina NovaSeq platform. Raw sequence data were analysed using CZ ID metagenomics and PikaVirus bioinformatics pipelines. RESULTS: Forty episodes of M/ME of unknown aetiology in 39 children were analysed by HCSS. A significant viral detection in 30 CSF samples was obtained, including six parechovirus A, three enterovirus ACD, four polyomavirus 5, three HHV-7, two BKV, one HSV-1, one VZV, two CMV, one EBV, one influenza A virus, one rhinovirus, and 13 HERV-K113 detections. Of these, one sample with BKV, three with HHV-7, one with EBV, and all HERV-K113 were confirmed by specific PCR. The requirement for Intensive Care Unit admission was associated with HCSS detections. CONCLUSION: This study highlights HCSS as a powerful tool for the investigation of undiagnosed cases of M/ME. Data generated must be carefully analysed and reasonable precautions must be taken before establishing association of clinical features with unexpected or novel virus findings.


Subject(s)
Metagenomics , Viruses , Humans , Child, Preschool , Prospective Studies , Female , Male , Child , Viruses/genetics , Viruses/isolation & purification , Viruses/classification , Infant , Metagenomics/methods , Encephalitis/virology , Encephalitis/cerebrospinal fluid , Encephalitis/diagnosis , Cerebrospinal Fluid/virology , Meningitis, Viral/virology , Meningitis, Viral/cerebrospinal fluid , Meningitis, Viral/diagnosis , Adolescent , High-Throughput Nucleotide Sequencing , Spain , Meningitis/virology , Meningitis/cerebrospinal fluid , Meningitis/diagnosis , Encephalitis, Viral/virology , Encephalitis, Viral/cerebrospinal fluid , Encephalitis, Viral/diagnosis
4.
J Vet Diagn Invest ; 36(3): 447-456, 2024 May.
Article in English | MEDLINE | ID: mdl-38500024

ABSTRACT

Astroviruses have been found in cattle and other species with encephalitis. Our objective was to determine the frequency of neurotropic bovine astrovirus (BoAstV) in cases of encephalitis in cattle ≥ 4-mo-old. Of 56 cases of idiopathic lymphocytic encephalitis examined retrospectively (1988-2019), fixed brain from 11 cases (19%) tested positive by semi-quantitative RT-PCR for BoAstV CH13/NeuroS1. None of the control cases tested positive, including 32 with other forms of encephalitis and 40 with no neurologic disease. Most astrovirus-positive cases were 1-2-y-old, with a range of 7 mo to 7 y, and affected both beef and dairy breeds with wide geographic distribution. BoAstV-positive cases had acute onset of neurologic signs of 12 h to 7 d before death or euthanasia. Affected cattle had lymphocytic inflammation throughout the brain including cerebrum, thalamus, midbrain, cerebellum, medulla oblongata, and spinal cord, and affecting gray and white matter. Further PCR testing identified a possible cause in 9 of the 45 (20%) remaining idiopathic cases of lymphocytic encephalitis, including eastern equine encephalitis virus, Listeria monocytogenes, bovine viral diarrhea virus, bovine alphaherpesvirus 1, and ovine gammaherpesvirus 2 (malignant catarrhal fever); we found no cases of infection by West Nile virus, rabies virus, or Chlamydia spp. No cause was identified in 36 of 56 (64%) cases of lymphocytic encephalitis. We frequently identified neurotropic BoAstV in cases of lymphocytic encephalitis that had no previously identified cause. Neurotropic BoAstV infections had gone undetected for decades, but the frequency of BoAstV infections has not increased among contemporary cases.


Subject(s)
Astroviridae Infections , Cattle Diseases , Animals , Cattle , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Astroviridae Infections/epidemiology , Cattle Diseases/virology , Cattle Diseases/epidemiology , Cattle Diseases/pathology , Retrospective Studies , Ontario/epidemiology , Female , Male , Encephalitis, Viral/veterinary , Encephalitis, Viral/virology , Encephalitis, Viral/epidemiology , Encephalitis, Viral/pathology , Astroviridae/isolation & purification , Astroviridae/genetics
5.
Jpn J Infect Dis ; 77(3): 155-160, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38296544

ABSTRACT

Human parainfluenza virus type 3 (HPIV-3, human respirovirus 3) is the second most frequently detected virus in lower respiratory tract infections in children after human respiratory syncytial virus (HRSV). HPIV-3, similar to related respiratory viruses such as HRSV and influenza virus, may cause encephalopathy; however, the relevance of HPIV-3 as a pathogenic factor in encephalopathy is unknown. We attempted to detect HPIV-1, HPIV-2, HPIV-3, HPIV-4, HRSV, and human metapneumovirus (HMPV) in 136 patients with encephalitis/encephalopathy or suspected encephalitis/encephalopathy during a 6-year period from 2014 to 2019. HPIV-3 was detected in 6 patients, followed by HRSV in 3 patients. The HPIV-3 strains detected were closely related to those detected in a patient with respiratory disease during the same period. Although HPIV-3 is less widely recognized than HRSV as a triggering virus of encephalopathy, our results suggest that HPIV-3 is as important as HRSV. Surveillance of the causative viruses of encephalopathy, including HPIV-3, would help clarify the causes of encephalopathy in Japan, as the cause is currently reported in less than half of cases in Japan.


Subject(s)
Parainfluenza Virus 3, Human , Respirovirus Infections , Humans , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/isolation & purification , Japan/epidemiology , Child, Preschool , Male , Female , Child , Infant , Respirovirus Infections/virology , Respirovirus Infections/epidemiology , Adolescent , Respiratory Tract Infections/virology , Respiratory Tract Infections/epidemiology , Phylogeny , Adult , Encephalitis, Viral/virology , Young Adult , Middle Aged , Brain Diseases/virology , Aged , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/isolation & purification
6.
J Virol ; 97(1): e0144222, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36541803

ABSTRACT

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.


Subject(s)
Encephalitis, Viral , Neurons , Reoviridae Infections , Reoviridae , Animals , Mice , Apoptosis/genetics , Apoptosis/immunology , Chemokines/immunology , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Neurons/immunology , NF-kappa B/genetics , NF-kappa B/metabolism , Reoviridae/immunology , Reoviridae/pathogenicity , Reoviridae Infections/immunology , Reoviridae Infections/virology , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology
7.
Emerg Microbes Infect ; 11(1): 6-13, 2022 Dec.
Article in English | MEDLINE | ID: mdl-34783638

ABSTRACT

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.


Subject(s)
Borna Disease/diagnosis , Borna disease virus/isolation & purification , Encephalitis, Viral/diagnosis , Aged , Animals , Borna Disease/epidemiology , Borna Disease/pathology , Borna Disease/virology , Borna disease virus/classification , Borna disease virus/genetics , Brain/pathology , Brain/virology , Encephalitis, Viral/epidemiology , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Endemic Diseases , Female , Germany/epidemiology , Humans , Male , Middle Aged , Phylogeny
8.
Front Immunol ; 12: 753683, 2021.
Article in English | MEDLINE | ID: mdl-34899705

ABSTRACT

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.


Subject(s)
Cytopathogenic Effect, Viral , Disease Models, Animal , Encephalitis, Viral/metabolism , Parechovirus/pathogenicity , Picornaviridae Infections/metabolism , Animals , Animals, Newborn , Apoptosis , Autophagy , Cell Line, Tumor , Cerebral Cortex/virology , Chlorocebus aethiops , Cytokines/biosynthesis , Cytokines/genetics , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Glioblastoma/pathology , Hippocampus/virology , Humans , Inflammation , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon Type I/pharmacology , Interferons/biosynthesis , Interferons/genetics , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Neuroblastoma/pathology , Parechovirus/drug effects , Parechovirus/physiology , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , Pyroptosis , Vero Cells , Virus Replication/drug effects , Interferon Lambda
9.
Front Immunol ; 12: 786953, 2021.
Article in English | MEDLINE | ID: mdl-34925368

ABSTRACT

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.


Subject(s)
Encephalitis, Viral/immunology , Rabies virus/immunology , Rabies/immunology , Viral Zoonoses/immunology , Africa/epidemiology , Animals , Dogs , Encephalitis, Viral/epidemiology , Encephalitis, Viral/transmission , Encephalitis, Viral/virology , Endemic Diseases , Humans , Immunity, Innate , Rabies/epidemiology , Rabies/transmission , Rabies/virology , Saliva/virology , Viral Zoonoses/epidemiology , Viral Zoonoses/transmission , Viral Zoonoses/virology , Virus Replication/immunology
10.
Viruses ; 13(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34960633

ABSTRACT

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.


Subject(s)
Encephalitis, Viral/immunology , Herpes Simplex/immunology , Immunity, Innate , Inflammation , Rabies virus/immunology , Rabies/immunology , Simplexvirus/immunology , Animals , Astrocytes/immunology , Astrocytes/virology , Blood-Brain Barrier/virology , Central Nervous System/immunology , Central Nervous System/virology , Encephalitis, Viral/virology , Herpes Simplex/virology , Humans , Microglia/immunology , Microglia/virology , Neuroglia/immunology , Neuroglia/virology , Rabies/virology , Signal Transduction
11.
Cells ; 10(11)2021 11 15.
Article in English | MEDLINE | ID: mdl-34831405

ABSTRACT

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.


Subject(s)
Cell Polarity , Dengue Virus/physiology , Encephalitis Virus, Japanese/physiology , Encephalitis, Viral/pathology , Encephalitis, Viral/virology , Macrophages/pathology , Severity of Illness Index , Animals , Animals, Suckling , Cell Line , Disease Models, Animal , Encephalitis, Japanese/immunology , Encephalitis, Japanese/pathology , Encephalitis, Japanese/virology , Encephalitis, Viral/immunology , Hippocampus/pathology , Inflammation/pathology , Mice, Inbred ICR , Neurotoxins/toxicity , Receptors, CCR2/metabolism , Tumor Necrosis Factor-alpha/metabolism
12.
Pak J Pharm Sci ; 34(5): 1809-1812, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34803019

ABSTRACT

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.


Subject(s)
Corpus Callosum/pathology , Encephalitis, Viral/pathology , Mumps/complications , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Child , China , Diuretics, Osmotic/therapeutic use , Encephalitis, Viral/virology , Humans , Male , Mannitol/therapeutic use , Methylprednisolone/therapeutic use , Ribavirin/therapeutic use
13.
Microbiol Spectr ; 9(3): e0148721, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34817277

ABSTRACT

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.


Subject(s)
Amantadine/pharmacology , Central Nervous System Viral Diseases/therapy , Central Nervous System Viral Diseases/veterinary , Drug Delivery Systems/methods , Nodaviridae/drug effects , Single-Domain Antibodies/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Central Nervous System/virology , Encephalitis, Viral/therapy , Encephalitis, Viral/virology , Fishes , Nanotubes, Carbon , Nodaviridae/immunology , Perciformes/virology , Single-Domain Antibodies/immunology
14.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769052

ABSTRACT

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.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Brain/virology , COVID-19/physiopathology , Encephalitis, Viral/virology , Pericytes/virology , Angiotensin-Converting Enzyme 2/genetics , Animals , Blood-Brain Barrier , Brain/pathology , COVID-19/etiology , Case-Control Studies , Encephalitis, Viral/pathology , Fibrinogen/metabolism , Humans , Immunohistochemistry/methods , Mice , Pericytes/metabolism , Pericytes/pathology , Receptor, Platelet-Derived Growth Factor beta/cerebrospinal fluid
15.
Trop Biomed ; 38(3): 435-445, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34608117

ABSTRACT

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.


Subject(s)
COVID-19/pathology , Central Nervous System/pathology , Nervous System Diseases/virology , Peripheral Nervous System/pathology , Angiotensin-Converting Enzyme 2/metabolism , Anosmia/virology , Central Nervous System/virology , Dysgeusia/virology , Encephalitis, Viral/virology , Humans , Meningitis, Viral/virology , Nervous System Diseases/pathology , Peripheral Nervous System/virology , SARS-CoV-2 , Seizures/virology
16.
Ocul Immunol Inflamm ; 29(6): 1225-1233, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34648417

ABSTRACT

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.


Subject(s)
COVID-19/diagnosis , Eye Infections, Viral/diagnosis , SARS-CoV-2 , Tears/virology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Nucleic Acid Testing , Conjunctivitis, Viral/diagnosis , Conjunctivitis, Viral/prevention & control , Conjunctivitis, Viral/virology , Encephalitis, Viral/diagnosis , Encephalitis, Viral/prevention & control , Encephalitis, Viral/virology , Eye Infections, Viral/prevention & control , Eye Infections, Viral/virology , Humans , Preventive Medicine/methods , Retinal Diseases/diagnosis , Retinal Diseases/prevention & control , Retinal Diseases/virology , SARS-CoV-2/pathogenicity
17.
J Virol ; 95(23): e0150621, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34495703

ABSTRACT

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.


Subject(s)
CD40 Antigens , CD40 Ligand , Encephalitis, Viral/prevention & control , Rift Valley Fever/immunology , Rift Valley fever virus/immunology , Rift Valley fever virus/physiology , T-Lymphocytes/immunology , Africa , Animals , Antibody Formation , B-Lymphocytes/immunology , Brain/virology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Epitopes , Female , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Sci Rep ; 11(1): 17751, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34493781

ABSTRACT

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.


Subject(s)
Cytokines/biosynthesis , Enterovirus A, Human/pathogenicity , Hand, Foot and Mouth Disease/virology , Neurons/virology , Acute Disease , Adult , Cell Line, Tumor , Child , Cytokines/genetics , Cytopathogenic Effect, Viral , Disease Outbreaks , Disease Susceptibility , Encephalitis, Viral/epidemiology , Encephalitis, Viral/virology , Enterovirus A, Human/classification , Enterovirus A, Human/genetics , Enterovirus A, Human/isolation & purification , Female , Gene Expression Regulation, Viral , Genotype , Hand, Foot and Mouth Disease/epidemiology , Humans , India/epidemiology , Male , Middle Aged , Neurons/metabolism , Paraplegia/epidemiology , Paraplegia/virology , Viral Tropism
19.
Viruses ; 13(9)2021 09 17.
Article in English | MEDLINE | ID: mdl-34578440

ABSTRACT

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.


Subject(s)
Astroviridae Infections/veterinary , Genome, Viral , Mamastrovirus/genetics , Open Reading Frames , Viral Proteins/genetics , Animals , Antigens, Viral , Astroviridae Infections/virology , Encephalitis, Viral/veterinary , Encephalitis, Viral/virology , Epitopes , Mamastrovirus/immunology , Mamastrovirus/metabolism , Nucleic Acid Conformation , Phylogeny , RNA, Viral/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Swine , Swine Diseases/virology , Untranslated Regions , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Proteins/metabolism
20.
Viruses ; 13(7)2021 06 23.
Article in English | MEDLINE | ID: mdl-34201545

ABSTRACT

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.


Subject(s)
Astroviridae Infections/veterinary , Astroviridae Infections/virology , Astroviridae/isolation & purification , Nervous System Diseases/veterinary , Nervous System Diseases/virology , Animals , Astroviridae/classification , Astroviridae/genetics , Astroviridae Infections/diagnosis , Astroviridae Infections/pathology , Brain/pathology , Brain/virology , Encephalitis, Viral/diagnosis , Encephalitis, Viral/pathology , Encephalitis, Viral/veterinary , Encephalitis, Viral/virology , Genome, Viral , Nervous System Diseases/diagnosis , Nervous System Diseases/pathology , Phylogeny
SELECTION OF CITATIONS
SEARCH DETAIL
...