Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Pediatr Res ; 91(1): 204-208, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33674737

RESUMO

BACKGROUND: The developmental stages and function of immune cells in the central nervous system during infancy and childhood are poorly understood. We analyzed whether cytokine and chemokine profiles in children and adolescents with viral central nervous system infections were different depending on age. METHODS: The acute phase cerebrospinal fluid of 80 children (mean age 98 months, range 1-206 months) were analyzed for protein levels of interleukin-1ß (IL-1ß), IL-1-RA, IL-4, IL-6, IL-7, IL-8, IL-10, IL-12, IL-13, IL-15, IL-17, IL-18, monocyte chemoattractant protein-1 (MCP-1), interferon (IFN) gamma-induced protein 10 (IP-10), IFN-γ, and macrophage migration inhibitory factor (MIF). RESULTS: We found an age-dependent increased expression of IL-4, IL-6, IL-13, MIF, IP-10, and IFN-γ and a decreased expression of MCP-1 and IL-15 in response to a viral infection of the central nervous system. In contrast, all other cytokines and chemokine were unaffected by the age of the patient. CONCLUSION: These findings demonstrate that the immunological response to a viral infection matures during childhood and adolescence. This may in turn be of importance for the outcome of a viral infection and the risk for subsequent sequela. It also demonstrates that age is a factor that needs to be considered when using cytokines and chemokines as biomarkers for infections in the central nervous system. IMPACT: The immunological response to a viral infection matures during childhood and adolescence. This may be of importance for the outcome of a viral infection and the risk for subsequent sequela. It also demonstrates that age is a factor that needs to be considered when using cytokines and chemokines as biomarkers for infections in the central nervous system.


Assuntos
Envelhecimento/patologia , Viroses do Sistema Nervoso Central/patologia , Inflamação/patologia , Adolescente , Biomarcadores/metabolismo , Viroses do Sistema Nervoso Central/metabolismo , Quimiocinas/metabolismo , Criança , Pré-Escolar , Estudos de Coortes , Citocinas/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Inflamação/metabolismo , Masculino
2.
mBio ; 12(6): e0271221, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34781742

RESUMO

Poliomyelitis-like illness is a common clinical manifestation of neurotropic viral infections. Functional loss and death of motor neurons often lead to reduced muscle tone and paralysis, causing persistent motor sequelae among disease survivors. Despite several reports demonstrating the molecular basis of encephalopathy, the pathogenesis behind virus-induced flaccid paralysis remained largely unknown. The present study for the first time aims to elucidate the mechanism responsible for limb paralysis by studying clinical isolates of Japanese encephalitis virus (JEV) and Chandipura virus (CHPV) responsible for causing acute flaccid paralysis (AFP) in vast regions of Southeast Asia and the Indian subcontinent. An experimental model for studying virus-induced AFP was generated by intraperitoneal injection of 10-day-old BALB/c mice. Progressive decline in motor performance of infected animals was observed, with paralysis being correlated with death of motor neurons (MNs). Furthermore, we demonstrated that upon infection, MNs undergo an extrinsic apoptotic pathway in a RIG-I-dependent fashion via transcription factors pIRF-3 and pIRF-7. Both gene-silencing experiments using specific RIG-I-short interfering RNA and in vivo morpholino abrogated cellular apoptosis, validating the important role of pattern recognition receptor (PRR) RIG-I in MN death. Hence, from our experimental observations, we hypothesize that host innate response plays a significant role in deterioration of motor functioning upon neurotropic virus infections. IMPORTANCE Neurotropic viral infections are an increasingly common cause of immediate or delayed neuropsychiatric sequelae, cognitive impairment, and movement disorders or, in severe cases, death. Given the highest reported disability-adjusted life years and mortality rate worldwide, a better understanding of molecular mechanisms for underlying clinical manifestations like AFP will help in development of more effective tools for therapeutic solutions.


Assuntos
Viroses do Sistema Nervoso Central/metabolismo , Viroses do Sistema Nervoso Central/fisiopatologia , Proteína DEAD-box 58/metabolismo , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Neurônios Motores/citologia , Mielite/metabolismo , Mielite/fisiopatologia , Doenças Neuromusculares/metabolismo , Doenças Neuromusculares/fisiopatologia , Vesiculovirus/fisiologia , Animais , Morte Celular , Viroses do Sistema Nervoso Central/genética , Viroses do Sistema Nervoso Central/virologia , Proteína DEAD-box 58/genética , Vírus da Encefalite Japonesa (Espécie)/genética , Feminino , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Masculino , Camundongos , Atividade Motora , Neurônios Motores/metabolismo , Neurônios Motores/virologia , Mielite/genética , Mielite/virologia , Doenças Neuromusculares/genética , Doenças Neuromusculares/virologia , Vesiculovirus/genética
3.
Drug Discov Ther ; 14(6): 262-272, 2021 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-33390561

RESUMO

The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was identified in 2019 in Wuhan, China. Clinically, respiratory tract symptoms as well as other organs disorders are observed in patients positively diagnosed coronavirus disease 2019 (COVID-19). In addition, neurological symptoms, mainly anosmia, ageusia and headache were observed in many patients. Once in the central nervous system (CNS), the SARS-CoV-2 can reside either in a quiescent latent state, or eventually in actively state leading to severe acute encephalitis, characterized by neuroinflammation and prolonged neuroimmune activation. SRAS-CoV-2 requires angiotensin-converting enzyme 2 (ACE2) as a cell entry receptor. The expression of this receptor in endothelial cells of blood-brain barrier (BBB) shows that SRAS-CoV-2 may have higher neuroinvasive potential compared to known coronaviruses. This review summarizes available information regarding the impact of SRAS-CoV-2 in the brain and tended to identify its potential pathways of neuroinvasion. We offer also an understanding of the long-term impact of latently form of SARS-CoV-2 on the development of neurodegenerative disorders. As a conclusion, the persistent infection of SRAS-CoV-2 in the brain could be involved on human neurodegenerative diseases that evolve a gradual process, perhapes, over several decades.


Assuntos
COVID-19/virologia , Viroses do Sistema Nervoso Central/virologia , Doenças Neurodegenerativas/virologia , Neurônios/virologia , SARS-CoV-2/patogenicidade , Tropismo Viral , Animais , COVID-19/complicações , Viroses do Sistema Nervoso Central/metabolismo , Viroses do Sistema Nervoso Central/patologia , Interações Hospedeiro-Patógeno , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Neurônios/patologia , Latência Viral
4.
Front Immunol ; 11: 1977, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973802

RESUMO

Zika virus (ZIKV), a mosquito-borne flavivirus, came into the spotlight in 2016 when it was found to be associated with an increased rate of microcephalic newborns in Brazil. The virus has further been recognized to cause neurologic complications in children and adults in the form of myelitis, encephalitis, acute disseminated encephalomyelitis (ADEM) and Guillain Barre Syndrome in a fraction of infected individuals. With the ultimate goal of identifying correlates of protection to guide the design of an effective vaccine, the study of the immune response to ZIKV infection has become the focus of research worldwide. Both innate and adaptive immune responses seem to be essential for controlling the infection. Induction of sufficient levels of neutralizing antibodies has been strongly correlated with protection against reinfection in various models, while the role of CD8 T cells as antiviral effectors in the CNS has been controversial. In an attempt to improve our understanding regarding the role of ZIKV-induced CD8 T cells in protective immunity inside the CNS, we have expanded on previous studies in intracranially infected mice. In a recent study, we have demonstrated that, peripheral ZIKV infection in adult C57BL/6 mice induces a robust CD8 T cell response that peaks within a week. In the present study, we used B cell deficient as well as wild-type mice to show that there is a race between CXCR3-dependent recruitment of the effector CD8 T cells and local ZIKV replication, and that CD8 T cells are capable of local viral control if they arrive in the brain early after viral invasion, in appropriate numbers and differentiation state. Our data highlight the benefits of considering this subset when designing vaccines against Zika virus.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Viroses do Sistema Nervoso Central/imunologia , Viroses do Sistema Nervoso Central/virologia , Interações Hospedeiro-Patógeno/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Zika virus/imunologia , Animais , Biomarcadores , Encéfalo/diagnóstico por imagem , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Linfócitos T CD8-Positivos/metabolismo , Viroses do Sistema Nervoso Central/diagnóstico , Viroses do Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Feminino , Imunização , Imunofenotipagem , Contagem de Linfócitos , Depleção Linfocítica , Camundongos , Camundongos Knockout , Carga Viral , Infecção por Zika virus/diagnóstico , Infecção por Zika virus/metabolismo
6.
Methods Mol Biol ; 2142: 41-57, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32367357

RESUMO

Detection of Zika virus (ZIKV) in the central nervous system (CNS) is a critical step when studying the pathogenesis of the infection in animal models. Both viral load determination and immunohistochemistry (IHC) staining are useful methods to quantitatively and qualitatively characterize viral infections in target tissues. Here, we describe viral RNA load determination by droplet digital PCR as well as protein detection by polymer-based IHC as effective techniques to quantify and localize ZIKV in the CNS of mice.


Assuntos
Sistema Nervoso Central/virologia , Imuno-Histoquímica/métodos , Reação em Cadeia da Polimerase/métodos , Infecção por Zika virus/diagnóstico , Zika virus/isolamento & purificação , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/virologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Viroses do Sistema Nervoso Central/diagnóstico , Viroses do Sistema Nervoso Central/metabolismo , Viroses do Sistema Nervoso Central/patologia , Viroses do Sistema Nervoso Central/virologia , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , RNA Viral/análise , RNA Viral/metabolismo , Carga Viral/métodos , Zika virus/genética , Zika virus/metabolismo , Infecção por Zika virus/metabolismo , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
7.
J Neurosci ; 40(18): 3517-3532, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32245829

RESUMO

One of the first signs of viral infection is body-wide aches and pain. Although this type of pain usually subsides, at the extreme, viral infections can induce painful neuropathies that can last for decades. Neither of these types of pain sensitization is well understood. A key part of the response to viral infection is production of interferons (IFNs), which then activate their specific receptors (IFNRs) resulting in downstream activation of cellular signaling and a variety of physiological responses. We sought to understand how type I IFNs (IFN-α and IFN-ß) might act directly on nociceptors in the dorsal root ganglion (DRG) to cause pain sensitization. We demonstrate that type I IFNRs are expressed in small/medium DRG neurons and that their activation produces neuronal hyper-excitability and mechanical pain in mice. Type I IFNs stimulate JAK/STAT signaling in DRG neurons but this does not apparently result in PKR-eIF2α activation that normally induces an anti-viral response by limiting mRNA translation. Rather, type I IFNs stimulate MNK-mediated eIF4E phosphorylation in DRG neurons to promote pain hypersensitivity. Endogenous release of type I IFNs with the double-stranded RNA mimetic poly(I:C) likewise produces pain hypersensitivity that is blunted in mice lacking MNK-eIF4E signaling. Our findings reveal mechanisms through which type I IFNs cause nociceptor sensitization with implications for understanding how viral infections promote pain and can lead to neuropathies.SIGNIFICANCE STATEMENT It is increasingly understood that pathogens interact with nociceptors to alert organisms to infection as well as to mount early host defenses. Although specific mechanisms have been discovered for diverse bacterial and fungal pathogens, mechanisms engaged by viruses have remained elusive. Here we show that type I interferons, one of the first mediators produced by viral infection, act directly on nociceptors to produce pain sensitization. Type I interferons act via a specific signaling pathway (MNK-eIF4E signaling), which is known to produce nociceptor sensitization in inflammatory and neuropathic pain conditions. Our work reveals a mechanism through which viral infections cause heightened pain sensitivity.


Assuntos
Viroses do Sistema Nervoso Central/metabolismo , Interferon Tipo I/toxicidade , Nociceptores/metabolismo , Limiar da Dor/fisiologia , Dor/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Animais , Células Cultivadas , Viroses do Sistema Nervoso Central/induzido quimicamente , Viroses do Sistema Nervoso Central/patologia , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Nociceptores/efeitos dos fármacos , Nociceptores/patologia , Dor/induzido quimicamente , Dor/patologia , Limiar da Dor/efeitos dos fármacos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/patologia
8.
J Virol ; 93(16)2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31167912

RESUMO

Enterovirus D68 (EV-D68) is an emerging virus that has been identified as a cause of recent outbreaks of acute flaccid myelitis (AFM), a poliomyelitis-like spinal cord syndrome that can result in permanent paralysis and disability. In experimental mouse models, EV-D68 spreads to, infects, and kills spinal motor neurons following infection by various routes of inoculation. The topography of virus-induced motor neuron loss correlates with the pattern of paralysis. The mechanism(s) by which EV-D68 spreads to target motor neurons remains unclear. We sought to determine the capacity of EV-D68 to spread by the neuronal route and to determine the role of known EV-D68 receptors, sialic acid and intracellular adhesion molecule 5 (ICAM-5), in neuronal infection. To do this, we utilized a microfluidic chamber culture system in which human induced pluripotent stem cell (iPSC) motor neuron cell bodies and axons can be compartmentalized for independent experimental manipulation. We found that EV-D68 can infect motor neurons via their distal axons and spread by retrograde axonal transport to the neuronal cell bodies. Virus was not released from the axons via anterograde axonal transport after infection of the cell bodies. Prototypic strains of EV-D68 depended on sialic acid for axonal infection and transport, while contemporary circulating strains isolated during the 2014 EV-D68 outbreak did not. The pattern of infection did not correspond with the ICAM-5 distribution and expression in either human tissue, the mouse model, or the iPSC motor neurons.IMPORTANCE Enterovirus D68 (EV-D68) infections are on the rise worldwide. Since 2014, the United States has experienced biennial spikes in EV-D68-associated acute flaccid myelitis (AFM) that have left hundreds of children paralyzed. Much remains to be learned about the pathogenesis of EV-D68 in the central nervous system (CNS). Herein we investigated the mechanisms of EV-D68 CNS invasion through neuronal pathways. A better understanding of EV-D68 infection in experimental models may allow for better prevention and treatment strategies of EV-D68 CNS disease.


Assuntos
Transporte Axonal , Enterovirus Humano D/fisiologia , Infecções por Enterovirus/metabolismo , Infecções por Enterovirus/virologia , Interações Hospedeiro-Patógeno , Neurônios Motores/metabolismo , Neurônios Motores/virologia , Ácido N-Acetilneuramínico/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Viroses do Sistema Nervoso Central/metabolismo , Viroses do Sistema Nervoso Central/virologia , Modelos Animais de Doenças , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Neurônios Motores/citologia , Mielite/metabolismo , Mielite/virologia , Proteínas do Tecido Nervoso/metabolismo , Doenças Neuromusculares/metabolismo , Doenças Neuromusculares/virologia , Paralisia/etiologia
9.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(6): 704-710, 2018 Jun 20.
Artigo em Chinês | MEDLINE | ID: mdl-29997093

RESUMO

OBJECTIVE: To explore whether vimentin (VIM) mediates the activation of inflammasome in mice with EV71 infection in the central nervous system. METHODS: Forty VIM knockout mice (VIM-/-, 3 to 5 days old) were randomly divided into control group and infection group. The infection group was intraperitoneally injected with EV71 (108 TCID50), while the control group was injected with PBS (10 µL); another 40 wild-type mice (WT, 3 to 5 days old) were grouped in the same manner. The general conditions of mice were observed each day. Western blotting, ELISA, and RT-PCR were used to measure the levels of IL-1ß and casepase-1 in the brain or cerebrospinal fluid. The pathological changes in the cerebella and brain were observed using immunohistochemistry. RESULTS: Compared with the control group, the VIM-/- mice infected with EV71 showed no significant changes in NLRP3, IL-1ß or caspase-1 expression. The WT mice infected with EV71 showed obviously increased NLRP3, IL-1ß, and caspase-1 expressions in the central nervous system. The neurons of infected VIM-/- mice exhibited milder cell damage than the those in WT mice. CONCLUSION: VIM mediates the activation of inflammasome and promotes brain inflammation and neuronal damage in mice with EV71 infection in the central nervous system.


Assuntos
Encéfalo/efeitos dos fármacos , Viroses do Sistema Nervoso Central/metabolismo , Enterovirus Humano A , Infecções por Enterovirus/metabolismo , Inflamassomos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/efeitos dos fármacos , Vimentina/farmacologia , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Caspase 1/líquido cefalorraquidiano , Caspase 1/metabolismo , Viroses do Sistema Nervoso Central/líquido cefalorraquidiano , Viroses do Sistema Nervoso Central/virologia , Infecções por Enterovirus/líquido cefalorraquidiano , Infecções por Enterovirus/virologia , Interleucina-1beta/líquido cefalorraquidiano , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/líquido cefalorraquidiano , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Distribuição Aleatória , Vimentina/genética
10.
J Neuroinflammation ; 15(1): 20, 2018 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343258

RESUMO

BACKGROUND: Varicella zoster virus (VZV) reactivation spans the spectrum from uncomplicated segmental herpes zoster to life-threatening disseminated CNS infection. Moreover, in the absence of a small animal model for this human pathogen, studies of pathogenesis at the organismal level depend on analysis of human biosamples. Changes in cerebrospinal fluid (CSF) metabolites may reflect critical aspects of host responses and end-organ damage in neuroinfection and neuroinflammation. We therefore applied a targeted metabolomics screen of CSF to three clinically distinct forms of VZV reactivation and infectious and non-infectious disease controls in order to identify biomarkers for CNS involvement in VZV reactivation. METHODS: Metabolite profiles were determined by targeted liquid chromatography-mass spectrometry in CSF from patients with segmental zoster (shingles, n = 14), facial nerve zoster (n = 16), VZV meningitis/encephalitis (n = 15), enteroviral meningitis (n = 10), idiopathic Bell's palsy (n = 11), and normal pressure hydrocephalus (n = 15). RESULTS: Concentrations of 88 metabolites passing quality assessment clearly separated the three VZV reactivation forms from each other and from the non-infected samples. Internal cross-validation identified four metabolites (SM C16:1, glycine, lysoPC a C26:1, PC ae C34:0) that were particularly associated with VZV meningoencephalitis. SM(OH) C14:1 accurately distinguished facial nerve zoster from Bell's palsy. Random forest construction revealed even more accurate classifiers (signatures comprising 2-4 metabolites) for most comparisons. Some of the most accurate biomarkers correlated only weakly with CSF leukocyte count, indicating that they do not merely reflect recruitment of inflammatory cells but, rather, specific pathophysiological mechanisms. Across all samples, only the sum of hexoses and the amino acids arginine, serine, and tryptophan correlated negatively with leukocyte count. Increased expression of the metabolites associated with VZV meningoencephalitis could be linked to processes relating to neuroinflammation/immune activation, neuronal signaling, and cell stress, turnover, and death (e.g., autophagy and apoptosis), suggesting that these metabolites might sense processes relating to end-organ damage. CONCLUSIONS: The results provide proof-of-concept for the value of CSF metabolites as (1) disease-associated signatures suggesting pathophysiological mechanisms, (2) degree and nature of neuroinflammation, and (3) biomarkers for diagnosis and risk stratification of VZV reactivation and, likely, neuroinfections due to other pathogens. TRIAL REGISTRATION: Not applicable (non-interventional study).


Assuntos
Viroses do Sistema Nervoso Central/líquido cefalorraquidiano , Herpesvirus Humano 3/fisiologia , Metabolômica/métodos , Ativação Viral/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/líquido cefalorraquidiano , Biomarcadores/metabolismo , Viroses do Sistema Nervoso Central/diagnóstico , Viroses do Sistema Nervoso Central/metabolismo , Feminino , Humanos , Masculino , Espectrometria de Massas/métodos , Pessoa de Meia-Idade , Estudos Prospectivos , Adulto Jovem
11.
Hum Pathol ; 61: 140-147, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27993575

RESUMO

In this retrospective study from one institution, we performed a clinicopathological study of a cohort of patients with posttransplant lymphoproliferative disorder (PTLD) confined to the central nervous system. We also identified a comparison cohort of patients with de novo primary diffuse large B-cell lymphoma of the central nervous system. We performed a detailed morphologic review, evaluated Epstein-Barr virus (EBV) by in situ hybridization, and interpreted a panel of immunohistochemical stains in a subset of cases including Hans classification markers (CD10, BCL6, MUM1), p53, CD30, Myc, and BCL2. All 17 of the posttransplant and none of 11 de novo cases were EBV positive (P < .005). Morphologic patterns identified in the PTLD cases were monomorphic diffuse large B-cell lymphoma pattern (10 patients) and "T-cell-rich" pattern (7 patients). The monomorphic posttransplant cases were more likely to be Myc negative (P = .015) and CD30 positive (P < .005) than the de novo cases, and showed a similarly low rate of p53 positivity by immunohistochemistry. No prognostic factors for overall survival were identified. Central nervous system PTLD is EBV positive, typically lacks p53 and Myc expression by immunohistochemistry, and can present with numerous background T lymphocytes.


Assuntos
Linfócitos B/química , Neoplasias do Sistema Nervoso Central/química , Viroses do Sistema Nervoso Central/metabolismo , Infecções por Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/isolamento & purificação , Imuno-Histoquímica , Linfoma Difuso de Grandes Células B/química , Transplante de Órgãos/efeitos adversos , Proteína Supressora de Tumor p53/análise , Adolescente , Adulto , Idoso , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos B/virologia , Biomarcadores/análise , Biópsia , Neoplasias do Sistema Nervoso Central/imunologia , Neoplasias do Sistema Nervoso Central/patologia , Neoplasias do Sistema Nervoso Central/virologia , Viroses do Sistema Nervoso Central/imunologia , Viroses do Sistema Nervoso Central/patologia , Viroses do Sistema Nervoso Central/virologia , Criança , Infecções por Vírus Epstein-Barr/imunologia , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/genética , Humanos , Hibridização In Situ , Linfoma Difuso de Grandes Células B/imunologia , Linfoma Difuso de Grandes Células B/patologia , Linfoma Difuso de Grandes Células B/virologia , Masculino , Pessoa de Meia-Idade , Minnesota , Valor Preditivo dos Testes , Proteínas Proto-Oncogênicas c-myc/análise , RNA Viral/genética , Estudos Retrospectivos , Linfócitos T/química , Linfócitos T/imunologia , Linfócitos T/patologia , Linfócitos T/virologia , Adulto Jovem
12.
J Neurol Sci ; 372: 40-48, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-28017244

RESUMO

Members of the toll-like receptor (TLR) family are pathogen recognition receptors that recognize pathogen-associated molecular patterns. TLRs mediate the modulation of innate immune responses and influence the development of adaptive immunity. TLR3 is the first identified antiviral TLR that recognizes dsRNA. TLR3 plays a central role in the activation of host immune responses to viral infections and shows detrimental or protective effects against various viral infections. Several viruses are neurotropic and can infect the central nervous system, leading to neuropathologies such as encephalitis, encephalopathy, meningitis, and neuritis. Single nucleotide polymorphism (SNP) in the TLR3 gene is associated with the susceptibility to a spectrum of nervous system viral infections. In this review, we discussed the existing knowledge of TLR3 immune responses on the basis of the experimental evidence and coherent picture of the SNP in TLR3 that is associated with various neurotropic virus infections.


Assuntos
Viroses do Sistema Nervoso Central/metabolismo , Receptor 3 Toll-Like/metabolismo , Animais , Antivirais/uso terapêutico , Viroses do Sistema Nervoso Central/imunologia , Viroses do Sistema Nervoso Central/terapia , Bases de Dados Bibliográficas/estatística & dados numéricos , Humanos , Receptor 3 Toll-Like/genética
13.
Tissue Barriers ; 4(1): e1142492, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27141421

RESUMO

Brain endothelial cells form a unique cellular structure known as the tight junction to regulate the exchanges between the blood and the parenchyma by limiting the paracellular diffusion of blood-borne substance. Together with the restricted pathway of transcytosis, the tight junction in the brain endothelial cells provides the central nervous system (CNS) with effective protection against both the foreign pathogens and the host immune cells, which is also termed the "blood-brain barrier." The blood-brain barrier is particularly important for defending against neurotropic viral infections that have become a major source of diseases worldwide. Many neurotropic viruses are able to cross the BBB and infect the CNS through very poorly understood processes. This review focuses upon the structural and functional changes of the brain endothelial tight junction in response to viral infections in the CNS and how the tight junction changes may be studied with advanced imaging and recording approaches to reveal novel processes used by the viruses to cross the barrier system. Additional emphasis is placed upon new countermeasures that can act directly upon the tight junction to improve the pathogen clearance and minimize the inflammatory damage.


Assuntos
Barreira Hematoencefálica/virologia , Viroses do Sistema Nervoso Central/metabolismo , Interações Hospedeiro-Patógeno , Animais , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar , Humanos , Junções Íntimas/metabolismo , Junções Íntimas/virologia
14.
Sci Rep ; 6: 22629, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26936683

RESUMO

Restriction of HIV-1 in myeloid-lineage cells is attributed in part to the nucleotidase activity of the SAM-domain and HD-domain containing protein (SAMHD1), which depletes free nucleotides, blocking reverse transcription. In the same cells, the Vpx protein of HIV-2 and most SIVs counteracts SAMHD1. Both Type I and II interferons may stimulate SAMHD1 transcription. The contributions of SAMHD1 to retroviral restriction in the central nervous system (CNS) have been the subject of limited study. We hypothesized that SAMHD1 would respond to interferon in the SIV-infected CNS but would not control virus due to SIV Vpx. Accordingly, we investigated SAMHD1 transcript abundance and association with the Type I interferon response in an SIV model. SAMHD1 transcript levels were IFN responsive, increasing during acute phase infection and decreasing during a more quiescent phase, but generally remaining elevated at all post-infection time points. In vitro, SAMHD1 transcript was abundant in macaque astrocytes and further induced by Type I interferon, while IFN produced a weaker response in the more permissive environment of the macrophage. We cannot rule out a contribution of SAMHD1 to retroviral restriction in relatively non-permissive CNS cell types. We encourage additional research in this area, particularly in the context of HIV-1 infection.


Assuntos
Viroses do Sistema Nervoso Central/metabolismo , Macrófagos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/metabolismo , Regulação para Cima/imunologia , Carga Viral/imunologia , Animais , Viroses do Sistema Nervoso Central/imunologia , Viroses do Sistema Nervoso Central/virologia , Feminino , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Macaca nemestrina , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Proteínas Monoméricas de Ligação ao GTP/biossíntese , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Vírus da Imunodeficiência Símia/imunologia
15.
Curr Opin Immunol ; 36: 47-53, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26163762

RESUMO

The innate immune system mediates protection against neurotropic viruses that replicate in the central nervous system (CNS). Virus infection within specific cells of the CNS triggers activation of several families of pattern recognition receptors including Toll-like receptors, retinoic acid-inducible gene I like receptors, nucleotide-binding oligomerization domain-like receptors, and cytosolic DNA sensors. In this review, we highlight recent advances in our understanding of how cell-intrinsic host defenses within the CNS modulate infection of different DNA and RNA viruses.


Assuntos
Viroses do Sistema Nervoso Central/imunologia , Viroses do Sistema Nervoso Central/virologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Imunomodulação , Viroses/imunologia , Viroses/virologia , Animais , Viroses do Sistema Nervoso Central/genética , Viroses do Sistema Nervoso Central/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , MicroRNAs/genética , Receptores Imunológicos/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Viroses/genética , Viroses/metabolismo
16.
J Interferon Cytokine Res ; 35(5): 373-84, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25536401

RESUMO

Human immunodeficiency virus-1 (HIV-1)-infected monocytes/macrophages and microglia release increased levels of proinflammatory cytokines and chemokines, including ELR+ (containing glutamic acid-leucine-arginine motif) chemokines. To investigate the role of HIV-1 infection on chemokine regulation, monocyte-derived macrophages (MDMs) from normal donors were infected with HIV-1 and the expression of chemokines and their downstream biological functions were evaluated. Among the tested chemokines, CXCL5 was upregulated significantly both at the mRNA and protein level in the HIV-1-infected MDMs compared with mock-infected cultures. Upregulation of CXCL5 in the HIV-1-infected MDMs is, in part, regulated by increased interleukin-1ß (IL-1ß) production and phosphorylation of ERK1/2. Functional analyses indicate that HIV-1-induced overexpression of CXCL5 has enhanced the ability to attract neutrophils, as observed by chemotaxis assay. However, exposure of NT2, SH-SY5Y cells, and primary neurons to HIV-1-infected MDM supernatants resulted in cell death that was not rescued by anti-CXCL5 antibody suggesting that CXCL5 does not have direct effect on neuronal death. Together, these results suggest that the increased level of CXCL5 in tissue compartments, including the central nervous system of HIV-1-infected individuals might alter the inflammatory response through the infiltration of neutrophils into tissue compartment, thus causing secondary effects on resident cells.


Assuntos
Viroses do Sistema Nervoso Central/genética , Viroses do Sistema Nervoso Central/virologia , Quimiocina CXCL5/genética , Regulação da Expressão Gênica , HIV-1/fisiologia , Macrófagos/metabolismo , Macrófagos/virologia , Transcrição Gênica , Linhagem Celular , Sobrevivência Celular/genética , Viroses do Sistema Nervoso Central/imunologia , Viroses do Sistema Nervoso Central/metabolismo , Quimiocina CXCL5/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Perfilação da Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/imunologia , Neurônios/metabolismo , Infiltração de Neutrófilos/imunologia , Replicação Viral
17.
Neurochem Res ; 39(11): 2143-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25119165

RESUMO

We investigated changes in the brain-derived neurotrophic factor (BDNF) and interleukin (IL)-6 levels in pediatric patients with central nervous system (CNS) infections, particularly viral infection-induced encephalopathy. Over a 5-year study period, 24 children hospitalized with encephalopathy were grouped based on their acute encephalopathy type (the excitotoxicity, cytokine storm, and metabolic error types). Children without CNS infections served as controls. In serum and cerebrospinal fluid (CSF) samples, BDNF and IL-6 levels were increased in all encephalopathy groups, and significant increases were noted in the influenza-associated and cytokine storm encephalopathy groups. Children with sequelae showed higher BDNF and IL-6 levels than those without sequelae. In pediatric patients, changes in serum and CSF BDNF and IL-6 levels may serve as a prognostic index of CNS infections, particularly for the diagnosis of encephalopathy and differentiation of encephalopathy types.


Assuntos
Encefalopatias/virologia , Fator Neurotrófico Derivado do Encéfalo/sangue , Fator Neurotrófico Derivado do Encéfalo/líquido cefalorraquidiano , Viroses do Sistema Nervoso Central/metabolismo , Interleucina-6/sangue , Interleucina-6/líquido cefalorraquidiano , Encefalopatias/sangue , Encefalopatias/líquido cefalorraquidiano , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino
18.
Immunol Res ; 57(1-3): 292-302, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24203439

RESUMO

Human immunodeficiency virus (HIV) invades the brain early during infection and generates a chronic inflammatory microenvironment that can eventually result in neurological disease, even in the absence of significant viral replication. Thus, HIV-1 infection of the brain has been characterized both as a neuroimmunological and neurodegenerative disorder. While the brain and central nervous system (CNS) have historically been regarded as immune privileged or immunologically quiescent, newer concepts of CNS immunity suggest an important if not defining role for innate immune responses generated by glial cells. Innate immunity may be the first line of defense against HIV infection of the brain and CNS, with multiple cellular elements providing responses that can be anti-viral and neuroprotective, but also potentially neurotoxic, impairing neurogenesis and promoting neuronal apoptosis. To investigate the effects of HIV exposure on neurogenesis and neuronal survival, we have studied the responses of human neuroepithelial progenitor (NEP) cells, which undergo directed differentiation into astrocytes and neurons in vitro. We identified a group of genes that were differentially expressed in NEP-derived cells during virus exposure. This included genes that are strongly related to interferon-induced responses and antigen presentation. Moreover, we observed that the host factor apolipoprotein E influences the innate immune response expressed by these cells, with a more robust response in the apolipoprotein E3/E3 genotype cultures compared to the apolipoprotein E3/E4 counterparts. Thus, neuroepithelial progenitors and their differentiated progeny recognize HIV and respond to it by mounting an innate immune response with a vigor that is influenced by the host factor apolipoprotein E.


Assuntos
Viroses do Sistema Nervoso Central/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Imunidade Inata , Apolipoproteínas E/metabolismo , Viroses do Sistema Nervoso Central/metabolismo , Infecções por HIV/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos
19.
J Immunol ; 191(2): 875-83, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23749635

RESUMO

Four of the five members of the Toll/IL-1R domain-containing adaptor family are required for signaling downstream of TLRs, promoting innate immune responses against different pathogens. However, the role of the fifth member of this family, sterile α and Toll/IL-1R domain-containing 1 (SARM), is unclear. SARM is expressed primarily in the CNS where it is required for axonal death. Studies in Caenorhabditis elegans have also shown a role for SARM in innate immunity. To clarify the role of mammalian SARM in innate immunity, we infected SARM(-/-) mice with a number of bacterial and viral pathogens. SARM(-/-) mice show normal responses to Listeria monocytogenes, Mycobacterium tuberculosis, and influenza virus, but show dramatic protection from death after CNS infection with vesicular stomatitis virus. Protection correlates with reduced CNS injury and cytokine production by nonhematopoietic cells, suggesting that SARM is a positive regulator of cytokine production. Neurons and microglia are the predominant source of cytokines in vivo, supporting a role for SARM as a link between neuronal injury and innate immunity.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Viroses do Sistema Nervoso Central/imunologia , Proteínas do Citoesqueleto/metabolismo , Infecções por Rhabdoviridae/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Proteínas do Domínio Armadillo/genética , Proteínas do Domínio Armadillo/imunologia , Células da Medula Óssea , Células Cultivadas , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/microbiologia , Sistema Nervoso Central/virologia , Viroses do Sistema Nervoso Central/metabolismo , Citocinas/biossíntese , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/imunologia , Imunidade Inata , Vírus da Influenza A/imunologia , Listeria monocytogenes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Mycobacterium tuberculosis/imunologia , Neurônios/metabolismo , Infecções por Rhabdoviridae/metabolismo
20.
Artigo em Chinês | MEDLINE | ID: mdl-23710867

RESUMO

OBJECTIVE: To explore the changes in the threshold of auditory brainstem response (ABR) and [Ca(2+)]I and calmodulin (CaM) in cochlear nucleus of newborn mice infected by murine cytomegalovirus (MCMV) in the brain. METHODS: Sixty-nine newborn mice were randomized into model group and control group. The model group (54 mice) was established by intracranial injection with MCMV viral suspension 20 l and the same volume of 0.9% sodium chloride was injected in the control group (15 mice). After 1 month, the ABR was tested in a sound-electric screen environment and the threshold was recorded. Then intracellular free calcium [Ca(2+)]i and the mRNA level of CaM in the cochlear nucleus were assayed by flow cytometry and RT-PCR. RESULTS: Compare to the control group [(64.0 ± 1.3) dBSPL], the threshold of ABR in the model group [(84.5 ± 2.7) dBSPL] was increased (F = 2.789,P = 0.000). Moreover, in the model group the intracellular free calcium [Ca(2+)]i and the mRNA level of CaM in the cochlear nucleus were increased (F = 1.290, P = 0.000; F = 4.252, P = 0.023), and the differences were statistically significant. CONCLUSIONS: The intracranial injection of MCMV can lead to abnormal changes in the threshold of ABR in mice, and the change of [Ca(2+) ]I/CaM in cochlear nucleus may be the important pathological basis of sensorineural hearing loss induced by MCMV infection.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Viroses do Sistema Nervoso Central/metabolismo , Núcleo Coclear/metabolismo , Infecções por Citomegalovirus/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico , Células 3T3 , Animais , Limiar Auditivo , Viroses do Sistema Nervoso Central/virologia , Citomegalovirus , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...