Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
1.
J Neurovirol ; 30(1): 57-70, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38167982

RESUMO

In recent years, we have seen the widespread devastations and serious health complications manifested by COVID-19 globally. Although we have effectively controlled the pandemic, uncertainties persist regarding its potential long-term effects, including prolonged neurological issues. To gain comprehensive insights, we conducted a meta-analysis of mass spectrometry-based proteomics data retrieved from different studies with a total of 538 COVID-19 patients and 523 healthy controls. The meta-analysis revealed that top-enriched pathways were associated with neurological disorders, including Alzheimer's (AD) and Parkinson's disease (PD). Further analysis confirmed a direct correlation in the expression patterns of 24 proteins involved in Alzheimer's and 23 proteins in Parkinson's disease with COVID-19. Protein-protein interaction network and cluster analysis identified SNCA as a hub protein, a known biomarker for Parkinson's disease, in both AD and PD. To the best of our knowledge, this is the first meta-analysis study providing proteomic profiling evidence linking COVID-19 to neurological complications.


Assuntos
Doença de Alzheimer , Biomarcadores , COVID-19 , Doença de Parkinson , Mapas de Interação de Proteínas , Proteoma , SARS-CoV-2 , COVID-19/sangue , COVID-19/virologia , COVID-19/metabolismo , Humanos , Doença de Parkinson/virologia , Doença de Parkinson/sangue , Doença de Parkinson/metabolismo , Doença de Parkinson/genética , Doença de Alzheimer/sangue , Doença de Alzheimer/virologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Biomarcadores/sangue , Biomarcadores/metabolismo , alfa-Sinucleína/sangue , alfa-Sinucleína/metabolismo , Proteômica/métodos
2.
Transl Psychiatry ; 13(1): 396, 2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38104129

RESUMO

Although there are indications of a trend towards less severe acute respiratory symptoms and a decline in overall lethality from the novel Coronavirus Disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), more and more attention has been paid to the long COVID, including the increased risk of Alzheimer's disease (AD) in COVID-19 patients. In this study, we aim to investigate the involvement of N-terminal amyloid precursor protein (APP) in SARS-CoV-2-induced amyloid-ß (Aß) pathology. Utilizing both in vitro and in vivo methodologies, we first investigated the interaction between the spike protein of SARS-CoV-2 and N-terminal APP via LSPR and CoIP assays. The in vitro impacts of APP overexpression on virus infection were further evaluated in HEK293T/ACE2 cells, SH-SY5Y cells, and Vero cells. We also analyzed the pseudovirus infection in vivo in a mouse model overexpressing human wild-type APP. Finally, we evaluated the impact of APP on pseudovirus infection within human brain organoids and assessed the chronic effects of pseudovirus infection on Aß levels. We reported here for the first time that APP, the precursor of the Aß of AD, interacts with the Spike protein of SARS-CoV-2. Moreover, both in vivo and in vitro data further indicated that APP promotes the cellular entry of the virus, and exacerbates Aß-associated pathology in the APP/PS1 mouse model of AD, which can be ameliorated by N-terminal APP blockage. Our findings provide experimental evidence to interpret APP-related mechanisms underlying AD-like neuropathology in COVID-19 patients and may pave the way to help inform risk management and therapeutic strategies against diseases accordingly.


Assuntos
Doença de Alzheimer , COVID-19 , Internalização do Vírus , Animais , Humanos , Camundongos , Doença de Alzheimer/complicações , Doença de Alzheimer/virologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Chlorocebus aethiops , COVID-19/complicações , Modelos Animais de Doenças , Células HEK293 , Camundongos Transgênicos , Síndrome Pós-COVID-19 Aguda , Presenilina-1 , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus , Células Vero
3.
J Neurochem ; 163(6): 517-530, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36321194

RESUMO

Inflammation associated with viral infection of the nervous system has been involved in the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD) and multiple sclerosis. Polyinosinic:polycytidylic acid (poly[I:C]) is a Toll-like receptor 3 (TLR3) agonist that mimics the inflammatory response to systemic viral infections. Despite growing recognition of the role of glial cells in AD pathology, their involvement in the accumulation and clearance of amyloid ß (Aß) in the brain of patients with AD is poorly understood. Neprilysin (NEP) and insulin-degrading enzyme (IDE) are the main Aß-degrading enzymes in the brain. This study investigated whether poly(I:C) regulated Aß degradation and neurotoxicity by modulating NEP and IDE protein levels through TLR3 in astrocytes. To this aim, primary rat primary astrocyte cultures were treated with poly(I:C) and inhibitors of the TLR3 signaling. Protein levels were assessed by Western blot. Aß toxicity to primary neurons was measured by lactate dehydrogenase release. Poly(I:C) induced a significant decrease in NEP levels on the membrane of astrocytes as well as in the culture medium. The degradation of exogenous Aß was markedly delayed in poly(I:C)-treated astrocytes. This delay significantly increased the neurotoxicity of exogenous Aß1-42. Altogether, these results suggest that viral infections induce Aß neurotoxicity by decreasing NEP levels in astrocytes and consequently preventing Aß degradation.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Astrócitos , Insulisina , Neprilisina , Viroses , Animais , Ratos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/virologia , Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo , Astrócitos/virologia , Insulisina/metabolismo , Neprilisina/metabolismo , Receptor 3 Toll-Like/antagonistas & inibidores , Poli I-C/farmacologia , Viroses/complicações
4.
Physiol Int ; 109(2): 135-162, 2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35895572

RESUMO

Coronavirus disease 2019 (COVID-19) is a contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In addition to the pulmonary manifestations, COVID-19 patients may present a wide range of neurological disorders as extrapulmonary presentations. In this view, several studies have recently documented the worsening of neurological symptoms within COVID-19 morbidity in patients previously diagnosed with neurodegenerative diseases (NDs). Moreover, several cases have also been reported in which the patients presented parkinsonian features after initial COVID-19 symptoms. These data raise a major concern about the possibility of communication between SARS-CoV-2 infection and the initiation and/or worsening of NDs. In this review, we have collected compelling evidence suggesting SARS-CoV-2, as an environmental factor, may be capable of developing NDs. In this respect, the possible links between SARS-CoV-2 infection and molecular pathways related to most NDs and the pathophysiological mechanisms of the NDs such as Alzheimer's disease, vascular dementia, frontotemporal dementia, Parkinson's disease, and amyotrophic lateral sclerosis will be explained.


Assuntos
COVID-19/complicações , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/virologia , SARS-CoV-2 , Idoso , Doença de Alzheimer/genética , Doença de Alzheimer/virologia , Humanos , Doença de Parkinson/genética , Doença de Parkinson/virologia
5.
Cells ; 11(8)2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35455977

RESUMO

The novel coronavirus (2019-nCoVCOVID-19) belongs to the Beta coronavirus family, which contains MERS-CoV (Middle East respiratory syndrome coronavirus) and SARS-CoV (severe acute respiratory syndrome coronavirus). SARS-CoV-2 activates the innate immune system, thereby activating the inflammatory mechanism, causing the release of inflammatory cytokines. Moreover, it has been suggested that COVID-19 may penetrate the central nervous system, and release inflammatory cytokines in the brains, inducing neuroinflammation and neurodegeneration. Several links connect COVID-19 with Alzheimer's disease (AD), such as elevated oxidative stress, uncontrolled release of the inflammatory cytokines, and mitochondrial apoptosis. There are severe concerns that excessive immune cell activation in COVID-19 may aggravate the neurodegeneration and amyloid-beta pathology of AD. Here, we have collected the evidence, showing the links between the two diseases. The focus has been made to collect the information on the activation of the inflammation, its contributors, and shared therapeutic targets. Furthermore, we have given future perspectives, research gaps, and overlapping pathological bases of the two diseases. Lastly, we have given the short touch to the drugs that have equally shown rescuing effects against both diseases. Although there is limited information available regarding the exact links between COVID-19 and neuroinflammation, we have insight into the pathological contributors of the diseases. Based on the shared pathological features and therapeutic targets, we hypothesize that the activation of the immune system may induce neurological disorders by triggering oxidative stress and neuroinflammation.


Assuntos
COVID-19 , Doenças Neuroinflamatórias , Doença de Alzheimer/virologia , Antioxidantes/metabolismo , COVID-19/complicações , COVID-19/fisiopatologia , Citocinas , Humanos , Doenças Neuroinflamatórias/virologia , Estresse Oxidativo , SARS-CoV-2
6.
J Alzheimers Dis ; 85(3): 1053-1061, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34924389

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a fatal neurodegenerative disease, the etiology of which is unclear. Previous studies have suggested that some viruses are neurotropic and associated with AD. OBJECTIVE: By using bioinformatics analysis, we investigated the potential association between viral infection and AD. METHODS: A total of 5,066 differentially expressed genes (DEGs) in the temporal cortex between AD and control samples were identified. These DEGs were then examined via weighted gene co-expression network analysis (WGCNA) and clustered into modules of genes with similar expression patterns. Of identified modules, module turquoise had the highest correlation with AD. The module turquoise was further characterized using Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enrichment analysis. RESULTS: Our results showed that the KEGG pathways of the module turquoise were mainly associated with viral infection signaling, specifically Herpes simplex virus, Human papillomavirus, and Epstein-Barr virus infections. A total of 126 genes were enriched in viral infection signaling pathways. In addition, based on values of module membership and gene significance, a total of 508 genes within the module were selected for further analysis. By intersecting these 508 genes with those 126 genes enriched in viral infection pathways, we identified 4 hub genes that were associated with both viral infection and AD: TLR2, COL1A2, NOTCH3, and ZNF132. CONCLUSION: Through bioinformatics analysis, we demonstrated a potential link between viral infection and AD. These findings may provide a platform to further our understanding of AD pathogenesis.


Assuntos
Doença de Alzheimer , Biologia Computacional , Perfilação da Expressão Gênica , Viroses/genética , Doença de Alzheimer/genética , Doença de Alzheimer/virologia , Bases de Dados Genéticas , Infecções por Vírus Epstein-Barr/genética , Herpesvirus Humano 4/genética , Humanos , Transdução de Sinais/genética
7.
J Alzheimers Dis ; 85(2): 729-744, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34776447

RESUMO

BACKGROUND: COVID-19 pandemic is a global crisis which results in millions of deaths and causes long-term neurological sequelae, such as Alzheimer's disease (AD). OBJECTIVE: We aimed to explore the interaction between COVID-19 and AD by integrating bioinformatics to find the biomarkers which lead to AD occurrence and development with COVID-19 and provide early intervention. METHODS: The differential expressed genes (DEGs) were found by GSE147507 and GSE132903, respectively. The common genes between COVID-19 and AD were identified. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interactions (PPI) network analysis were carried out. Hub genes were found by cytoscape. A multivariate logistic regression model was constructed. NetworkAnalyst was used for the analysis of TF-gene interactions, TF-miRNA coregulatory network, and Protein-chemical Interactions. RESULTS: Forty common DEGs for AD and COVID-19 were found. GO and KEGG analysis indicated that the DEGs were enriched in the calcium signal pathway and other pathways. A PPI network was constructed, and 5 hub genes were identified (ITPR1, ITPR3, ITPKB, RAPGEF3, MFGE8). Four hub genes (ITPR1, ITPR3, ITPKB, RAPGEF3) which were considered as important factors in the development of AD that were affected by COVID-19 were shown by nomogram. Utilizing NetworkAnalyst, the interaction network of 4 hub genes and TF, miRNA, common AD risk genes, and known compounds is displayed, respectively. CONCLUSION: COVID-19 patients are at high risk of developing AD. Vaccination is required. Four hub genes can be considered as biomarkers for prediction and treatment of AD development caused by COVID-19. Compounds with neuroprotective effects can be used as adjuvant therapy for COVID-19 patients.


Assuntos
Doença de Alzheimer/genética , COVID-19/virologia , Mapas de Interação de Proteínas/genética , SARS-CoV-2/patogenicidade , Doença de Alzheimer/complicações , Doença de Alzheimer/metabolismo , Doença de Alzheimer/virologia , Biologia Computacional/métodos , Bases de Dados Genéticas , Perfilação da Expressão Gênica/métodos , Humanos , SARS-CoV-2/genética
8.
Int J Mol Sci ; 22(24)2021 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-34948400

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered the pandemic Coronavirus Disease 19 (COVID-19), causing millions of deaths. The elderly and those already living with comorbidity are likely to die after SARS-CoV-2 infection. People suffering from Alzheimer's disease (AD) have a higher risk of becoming infected, because they cannot easily follow health roles. Additionally, those suffering from dementia have a 40% higher risk of dying from COVID-19. Herein, we collected from Gene Expression Omnibus repository the brain samples of AD patients who died of COVID-19 (AD+COVID-19), AD without COVID-19 (AD), COVID-19 without AD (COVID-19) and control individuals. We inspected the transcriptomic and interactomic profiles by comparing the COVID-19 cohort against the control cohort and the AD cohort against the AD+COVID-19 cohort. SARS-CoV-2 in patients without AD mainly activated processes related to immune response and cell cycle. Conversely, 21 key nodes in the interactome are deregulated in AD. Interestingly, some of them are linked to beta-amyloid production and clearance. Thus, we inspected their role, along with their interactors, using the gene ontologies of the biological process that reveals their contribution in brain organization, immune response, oxidative stress and viral replication. We conclude that SARS-CoV-2 worsens the AD condition by increasing neurotoxicity, due to higher levels of beta-amyloid, inflammation and oxidative stress.


Assuntos
Doença de Alzheimer/genética , COVID-19/complicações , COVID-19/genética , Doença de Alzheimer/complicações , Doença de Alzheimer/virologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/virologia , COVID-19/fisiopatologia , Comorbidade/tendências , Bases de Dados Factuais , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Humanos , Inflamação/metabolismo , Síndromes Neurotóxicas/metabolismo , Estresse Oxidativo/fisiologia , Pandemias , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Transcriptoma/genética
9.
Int J Mol Sci ; 22(14)2021 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-34298881

RESUMO

Chronic neurodegenerative diseases are complex, and their pathogenesis is uncertain. Alzheimer's disease (AD) is a neurodegenerative brain alteration that is responsible for most dementia cases in the elderly. AD etiology is still uncertain; however, chronic neuroinflammation is a constant component of brain pathology. Infections have been associated with several neurological diseases and viruses of the Herpes family appear to be a probable cause of AD neurodegenerative alterations. Several different factors may contribute to the AD clinical progression. Exogeneous viruses or other microbes and environmental pollutants may directly induce neurodegeneration by activating brain inflammation. In this paper, we suggest that exogeneous brain insults may also activate retrotransposons and silent human endogenous retroviruses (HERVs). The initial inflammation of small brain areas induced by virus infections or other brain insults may activate HERV dis-regulation that contributes to neurodegenerative mechanisms. Chronic HERV activation in turn may cause progressive neurodegeneration that thereafter merges in cognitive impairment and dementia in genetically susceptible people. Specific treatment for exogenous end endogenous pathogens and decreasing pollutant exposure may show beneficial effect in early intervention protocol to prevent the progression of cognitive deterioration in the elderly.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/virologia , Encéfalo/patologia , Encéfalo/virologia , Retrovirus Endógenos/patogenicidade , Viroses/patologia , Viroses/virologia , Animais , Transtornos Cognitivos/patologia , Transtornos Cognitivos/virologia , Encefalite/patologia , Encefalite/virologia , Humanos
10.
Antiviral Res ; 192: 105116, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34107282

RESUMO

Growing evidence supports that chronic or latent infection of the central nervous system might be implicated in Alzheimer's disease (AD). Among them, Herpes simplex virus type 1 (HSV-1) has emerged as a major factor in the etiology of the disease. Our group is devoted to the study of the relationship among HSV-1, oxidative stress (OS) and neurodegeneration. We have found that HSV-1 induces the main neuropathological hallmarks of AD, including the accumulation of intracellular amyloid beta (Aß), hyperphosphorylated tau protein and autophagic vesicles, that OS exacerbates these effects, and that matrix metalloproteinase 14 (MMP-14) participates in the alterations induced by OS. In this work, we focused on the role of MMP-14 in the degenerative markers raised by HSV-1 infection. Interestingly, we found that MMP-14 blockage is a potent inhibitor of HSV-1 infection efficiency, that also reduces the degeneration markers, accumulation of Aß and hyperphosphorylated tau, induced by the virus. Our results point to MMP-14 as a potent antiviral target to control HSV-1 infection and its associated neurodegenerative effects.


Assuntos
Herpes Simples/metabolismo , Herpesvirus Humano 1/fisiologia , Metaloproteinase 14 da Matriz/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/virologia , Peptídeos beta-Amiloides/metabolismo , Animais , Antivirais/farmacologia , Autofagossomos/metabolismo , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Herpes Simples/virologia , Herpesvirus Humano 1/efeitos dos fármacos , Humanos , Metaloproteinase 14 da Matriz/deficiência , Inibidores de Metaloproteinases de Matriz/farmacologia , Camundongos , Neuroblastoma/patologia , Estresse Oxidativo , Fosforilação , Proteínas tau/metabolismo
11.
J Biol Chem ; 297(1): 100845, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34052228

RESUMO

Alzheimer's disease (AD) is a devastating fatal neurodegenerative disease. An alternative to the amyloid cascade hypothesis is that a viral infection is key to the etiology of late-onset AD, with ß-amyloid (Aß) peptides playing a protective role. In the current study, young 5XFAD mice that overexpress mutant human amyloid precursor protein with the Swedish, Florida, and London familial AD mutations were infected with one of two strains of herpes simplex virus 1 (HSV-1), 17syn+ and McKrae, at three different doses. Contrary to previous work, 5XFAD genotype failed to protect mice against HSV-1 infection. The region- and cell-specific tropisms of HSV-1 were not affected by the 5XFAD genotype, indicating that host-pathogen interactions were not altered. Seven- to ten-month-old 5XFAD animals in which extracellular Aß aggregates were abundant showed slightly better survival rate relative to their wild-type (WT) littermates, although the difference was not statistically significant. In these 5XFAD mice, HSV-1 replication centers were partially excluded from the brain areas with high densities of Aß aggregates. Aß aggregates were free of HSV-1 viral particles, and the limited viral invasion to areas with a high density of Aß aggregates was attributed to phagocytic activity of reactive microglia. In the oldest mice (12-15 months old), the survival rate did not differ between 5XFAD and WT littermates. While the current study questions the antiviral role of Aß, it neither supports nor refutes the viral etiology hypothesis of late-onset AD.


Assuntos
Peptídeos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Interações Hospedeiro-Patógeno/genética , Viroses/genética , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/virologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/patologia , Encéfalo/virologia , Modelos Animais de Doenças , Herpes Simples/genética , Herpes Simples/patologia , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Humanos , Camundongos , Camundongos Transgênicos , Microglia/patologia , Microglia/virologia , Presenilina-1/genética , Viroses/complicações , Viroses/patologia , Viroses/virologia , Replicação Viral/genética
12.
Neurochem Int ; 146: 105032, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33781848

RESUMO

Mounting evidence suggests a major role of infectious agents in the pathogenesis of sporadic Alzheimer's disease (AD). Among them, herpes simplex virus type 1 (HSV-1) infection has emerged as a major factor in the etiology of AD. HSV-1 is able to induce some of the main alterations of the disease such as hyperphosphorylation of tau protein and accumulation of amyloid-ß peptide. Functional genomic analysis of a cell model of HSV-1 infection and oxidative stress developed in our laboratory revealed lysosomal system to be the main pathway altered, and the lysosome-associated membrane protein 2 (LAMP2) gene one of the most strongly modulated genes. The aim of this work is to study LAMP2 as an AD candidate gene and to investigate its role in the neurodegeneration induced by HSV-1 using a LAMP2 knockdown cell model. LAMP2 deficiency led to a significant reduction of viral DNA replication and formation of infectious particles. In addition, tau hyperphosphorylation and inhibition of Aß secretion induced by the virus were attenuated by the absence of LAMP2. Finally, genetic association studies revealed LAMP2 genetic variants to be associated with AD risk. In summary, our data indicate that LAMP2 could be a suitable candidate to mediate the AD-like phenotype caused by HSV-1.


Assuntos
Doença de Alzheimer/metabolismo , Herpes Simples/metabolismo , Herpes Simples/prevenção & controle , Herpesvirus Humano 1/metabolismo , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/genética , Doença de Alzheimer/virologia , Animais , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes/métodos , Herpes Simples/genética , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/antagonistas & inibidores , Proteína 2 de Membrana Associada ao Lisossomo/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/virologia
13.
Arch Virol ; 166(3): 733-753, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33502593

RESUMO

The chronic dysfunction of neuronal cells, both central and peripheral, a characteristic of neurological disorders, may be caused by irreversible damage and cell death. In 2016, more than 276 million cases of neurological disorders were reported worldwide. Moreover, neurological disorders are the second leading cause of death. Generally, the etiology of neurological diseases is not fully understood. Recent studies have related the onset of neurological disorders to viral infections, which may cause neurological symptoms or lead to immune responses that trigger these pathological signs. Currently, this relationship is mostly based on epidemiological data on infections and seroprevalence of patients who present with neurological disorders. The number of studies aiming to elucidate the mechanism of action by which viral infections may directly or indirectly contribute to the development of neurological disorders has been increasing over the years but these studies are still scarce. Comprehending the pathogenesis of these diseases and exploring novel theories may favor the development of new strategies for diagnosis and therapy in the future. Therefore, the objective of the present study was to review the main pieces of evidence for the relationship between viral infection and neurological disorders such as Alzheimer's disease, Parkinson's disease, Guillain-Barré syndrome, multiple sclerosis, and epilepsy. Viruses belonging to the families Herpesviridae, Orthomyxoviridae, Flaviviridae, and Retroviridae have been reported to be involved in one or more of these conditions. Also, neurological symptoms and the future impact of infection with SARS-CoV-2, a member of the family Coronaviridae that is responsible for the COVID-19 pandemic that started in late 2019, are reported and discussed.


Assuntos
COVID-19/patologia , Doenças do Sistema Nervoso/virologia , Tropismo Viral/fisiologia , Doença de Alzheimer/virologia , COVID-19/virologia , Epilepsia/virologia , Flaviviridae/metabolismo , Síndrome de Guillain-Barré/virologia , Herpesviridae/metabolismo , Humanos , Esclerose Múltipla/virologia , Doenças do Sistema Nervoso/patologia , Orthomyxoviridae/metabolismo , Doença de Parkinson/virologia , Retroviridae/metabolismo , SARS-CoV-2/metabolismo
14.
Alzheimers Dement ; 17(6): 1056-1065, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33399270

RESUMO

INTRODUCTION: The increasing evidence of SARS-CoV-2 impact on the central nervous system (CNS) raises key questions on its impact for risk of later life cognitive decline, Alzheimer's disease (AD), and other dementia. METHODS: The Alzheimer's Association and representatives from more than 30 countries-with technical guidance from the World Health Organization-have formed an international consortium to study the short-and long-term consequences of SARS-CoV-2 on the CNS-including the underlying biology that may contribute to AD and other dementias. This consortium will link teams from around the world covering more than 22 million COVID-19 cases to enroll two groups of individuals including people with disease, to be evaluated for follow-up evaluations at 6, 9, and 18 months, and people who are already enrolled in existing international research studies to add additional measures and markers of their underlying biology. CONCLUSIONS: The increasing evidence and understanding of SARS-CoV-2's impact on the CNS raises key questions on the impact for risk of later life cognitive decline, AD, and other dementia. This program of studies aims to better understand the long-term consequences that may impact the brain, cognition, and functioning-including the underlying biology that may contribute to AD and other dementias.


Assuntos
Encéfalo/virologia , COVID-19/complicações , Doença de Alzheimer/virologia , Disfunção Cognitiva/virologia , Demência/virologia , Humanos , SARS-CoV-2
15.
Int J Mol Sci ; 23(1)2021 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-35008671

RESUMO

Current data strongly suggest herpes simplex virus type 1 (HSV-1) infection in the brain as a contributing factor to Alzheimer's disease (AD). The consequences of HSV-1 brain infection are multilateral, not only are neurons and glial cells damaged, but modifications also occur in their environment, preventing the transmission of signals and fulfillment of homeostatic and immune functions, which can greatly contribute to the development of disease. In this review, we discuss the pathological alterations in the central nervous system (CNS) cells that occur, following HSV-1 infection. We describe the changes in neurons, astrocytes, microglia, and oligodendrocytes related to the production of inflammatory factors, transition of glial cells into a reactive state, oxidative damage, Aß secretion, tau hyperphosphorylation, apoptosis, and autophagy. Further, HSV-1 infection can affect processes observed during brain aging, and advanced age favors HSV-1 reactivation as well as the entry of the virus into the brain. The host activates pattern recognition receptors (PRRs) for an effective antiviral response during HSV-1 brain infection, which primarily engages type I interferons (IFNs). Future studies regarding the influence of innate immune deficits on AD development, as well as supporting the neuroprotective properties of glial cells, would reveal valuable information on how to harness cytotoxic inflammatory milieu to counter AD initiation and progression.


Assuntos
Doença de Alzheimer/patologia , Doença de Alzheimer/virologia , Encéfalo/patologia , Herpesvirus Humano 1/fisiologia , Neuroglia/patologia , Neurônios/patologia , Doença de Alzheimer/genética , Animais , Herpesvirus Humano 1/imunologia , Humanos , Estresse Oxidativo
16.
Cell Mol Life Sci ; 78(6): 2485-2501, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33244624

RESUMO

The aggregation of specific proteins and their amyloid deposition in affected tissue in disease has been studied for decades assuming a sole pathogenic role of amyloids. It is now clear that amyloids can also encode important cellular functions, one of which involves the interaction potential of amyloids with microbial pathogens, including viruses. Human expressed amyloids have been shown to act both as innate restriction molecules against viruses as well as promoting agents for viral infectivity. The underlying molecular driving forces of such amyloid-virus interactions are not completely understood. Starting from the well-described molecular mechanisms underlying amyloid formation, we here summarize three non-mutually exclusive hypotheses that have been proposed to drive amyloid-virus interactions. Viruses can indirectly drive amyloid depositions by affecting upstream molecular pathways or induce amyloid formation by a direct interaction with the viral surface or specific viral proteins. Finally, we highlight the potential of therapeutic interventions using the sequence specificity of amyloid interactions to drive viral interference.


Assuntos
Amiloide/metabolismo , Vírus/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/virologia , Peptídeos beta-Amiloides/metabolismo , HIV/fisiologia , Herpesviridae/fisiologia , Humanos , Orthomyxoviridae/fisiologia , Proteínas tau/metabolismo
17.
Med Sci Monit ; 26: e930340, 2020 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-33323916

RESUMO

Alterations in complex behavioral patterns during the extended period of the COVID-19 pandemic are predicted to promote a variety of psychiatric disease symptoms due to enforced social isolation and self-quarantine. Accordingly, multifaceted mental health problems will continue to increase, thereby creating a challenge for society and the health care system in general. Recent studies show that COVID-19 can directly or indirectly influence the central nervous system, potentially causing neurological pathologies such as Alzheimer disease and Parkinson disease. Thus, chronic COVID-19-related disease processes have the potential to cause serious mental illnesses, including depression, anxiety, and sleep disorders. Importantly, mental health problems can foster systemic changes in functionally-linked neuroendocrine conditions that heighten a person's susceptibility to COVID-19 infection. These altered defense mechanisms may include compromised "self-control" and "self-care", as well as a "lack of insight" into the danger posed by the virus. These consequences may have serious social impacts on the future of COVID-19 survivors. Compounding the functionally related issues of altered mental health parameters and viral susceptibility are the potential effects of compromised immunity on the establishment of functional herd immunity. Within this context, mental health takes on added importance, particularly in terms of the need to increase support for mental health research and community-based initiatives. Thus, COVID-19 infections continue to reveal mental health targets, a process we must now be prepared to deal with.


Assuntos
COVID-19/complicações , Saúde Mental , SARS-CoV-2/patogenicidade , Sobreviventes/psicologia , Doença de Alzheimer/epidemiologia , Doença de Alzheimer/prevenção & controle , Doença de Alzheimer/virologia , Ansiedade/epidemiologia , Ansiedade/prevenção & controle , Ansiedade/psicologia , COVID-19/epidemiologia , COVID-19/psicologia , Depressão/epidemiologia , Depressão/prevenção & controle , Depressão/psicologia , Suscetibilidade a Doenças/psicologia , Humanos , Pandemias , Doença de Parkinson/epidemiologia , Doença de Parkinson/prevenção & controle , Doença de Parkinson/virologia , Distanciamento Físico , Autocuidado/psicologia , Autocontrole/psicologia , Isolamento Social/psicologia
18.
J Clin Neurosci ; 82(Pt A): 63-70, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33317741

RESUMO

There is growing evidence demonstrating the relationship between herpes simplex virus type 1 (HSV-1) infection and Alzheimer's disease (AD). We searched PubMed, Embase, and Cochrane databases for relevant articles. The Newcastle-Ottawa Scale (NOS) was used to evaluate the qualities of these studies. Pooled odds ratios (ORs) with 95% confidence intervals (CIs) were calculated using random-effects models. We also performed subgroup analyses stratified by apolipoprotein ε4 (APOE ε4), NOS score, and the method of confirming AD. A total of 21 studies between 1990 and 2020 were identified. The pooled OR suggested that HSV-1 infection is a risk factor of AD: pooled OR 1.40 (95% CI: 1.13-1.75; I2 = 3%, P = 0.42). In the subgroup analyses, the pooled ORs of HSV-1 infection associated with AD were 0.75 (95% CI: 0.24-2.37) among the APOE ε4-positive individuals; 0.85 (95% CI: 0.61-1.17) among the APOE ε4-negative individuals; 1.51 (95% CI: 1.10-2.06) in the high NOS score studies; 1.23 (95% CI: 0.85-1.76) in the moderate NOS score studies; 1.47 (95% CI: 1.16-1.87) in the clinical diagnosis group, and 1.20 (95% CI: 0.77-1.87) in the autopsy group. Our up-to-date systematic review and meta-analysis suggest that HSV-1 infection is a risk factor of AD.


Assuntos
Doença de Alzheimer/virologia , Herpes Simples/complicações , Herpesvirus Humano 1 , Humanos , Razão de Chances , Fatores de Risco
20.
J Alzheimers Dis ; 78(3): 855-869, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33074235

RESUMO

Across the fields of virology and neuroscience, the role of neurotropic viruses in Alzheimer's disease (AD) has received renewed enthusiasm, with a particular focus on human herpesviruses (HHVs). Recent genomic analyses of brain tissue collections and investigations of the antimicrobial responses of amyloid-ß do not exclude a role of HHVs in contributing to or accelerating AD pathogenesis. Due to continued expansion in our aging cohort and the lack of effective treatments for AD, this composition examines a potential neuroviral theory of AD in light of these recent data. Consideration reveals a possible viral "Hit-and-Run" scenario of AD, as well as neurobiological mechanisms (i.e., neuroinflammation, protein quality control, oxidative stress) that may increase risk for AD following neurotropic infection. Although limitations exist, this theoretical framework reveals several novel therapeutic targets that may prove efficacious in AD.


Assuntos
Doença de Alzheimer/virologia , Encéfalo/metabolismo , Genoma Viral , Infecções por Herpesviridae/metabolismo , Herpesviridae/genética , Inflamação/metabolismo , Estresse Oxidativo , Animais , Encéfalo/virologia , DNA Viral , Infecções por Herpesviridae/virologia , Interações entre Hospedeiro e Microrganismos , Humanos , Técnicas In Vitro , Inflamação/virologia , Infecção Latente , Tropismo Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...