Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Eur J Paediatr Neurol ; 29: 128-136, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32868196

RESUMO

PURPOSE: Herpes simplex virus encephalitis (HSE) is the most common cause of sporadic viral encephalitis in children and is responsible for epilepsy in approximately half of patients. In addition to medical treatment, epilepsy surgery may be offered to drug-resistant patients but carries a high risk of relapse of herpetic encephalitis. We are reporting our series of patients operated on between 2000 and 2019 with the systematic administration of acyclovir (ACV). RESULTS: Four pediatric patients aged 4.5-12.8 years with drug-resistant epilepsy post-HSE underwent a tailored focal resection following invasive recordings (three patients) and a complete callosotomy (one patient). The total number of the surgical procedures for the four patients was eight, and a systematic administration of ACV as a prophylactic treatment of herpetic encephalitis relapse was done at each step. No patients had a relapse and the ACV was well-tolerated in all the cases. Following surgery two patients are seizure free, the patient who underwent callosotomy is Engel 3 and the fourth patient, in whom a large epileptic zone has contraindicated a second surgery, is Engel 4. CONCLUSIONS: Our series demonstrated the dramatic efficacy of systematic ACV prophylaxis during all cranial surgeries. Moreover, our results on epilepsy, together with those of the literature, encourage more consideration regarding epilepsy surgery in this specific etiology. All types of surgical procedures (curative or palliative) can be offered to the patients, but in the case of focal surgery, due to the poor anatomical limits, invasive recordings are highly recommended.


Assuntos
Aciclovir/uso terapêutico , Antivirais/uso terapêutico , Encefalite por Herpes Simples/complicações , Epilepsia/terapia , Epilepsia/virologia , Adolescente , Criança , Pré-Escolar , Epilepsia Resistente a Medicamentos/cirurgia , Encefalite por Herpes Simples/prevenção & controle , Feminino , Humanos , Masculino , Prevenção Secundária/métodos
2.
Continuum (Minneap Minn) ; 24(5, Neuroinfectious Disease): 1284-1297, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30273240

RESUMO

PURPOSE OF REVIEW: This article discusses meningitis and encephalitis infections caused by viruses, excluding herpes family and human immunodeficiency virus (HIV). RECENT FINDINGS: The viral infections of the nervous system detailed in this article have no specific treatment other than supportive care. However, many of the viruses discussed are highly preventable by vaccination, proper skin protection against transmitting vectors, and postexposure prophylaxis. SUMMARY: While meningitis and encephalitis caused by viruses may have some clinical overlap, the management and outcomes can be highly disparate, making distinction between the two imperative. Furthermore, despite their relative rarity in terms of clinical disease, many of the viral infections discussed herein are highly preventable. Given the morbidity and mortality attached to such infections, provider and patient education are the best approach available to prevent these potentially devastating illnesses.


Assuntos
Gerenciamento Clínico , Encefalite por Herpes Simples/etiologia , Meningite Viral/etiologia , Adulto , Encefalite por Herpes Simples/diagnóstico por imagem , Encefalite por Herpes Simples/prevenção & controle , Encefalite por Herpes Simples/virologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Meningite Viral/diagnóstico por imagem , Meningite Viral/prevenção & controle , Meningite Viral/virologia
3.
Semin Perinatol ; 42(3): 168-175, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29544668

RESUMO

Neonatal herpes simplex virus (HSV) is an uncommon but devastating infection in the newborn, associated with significant morbidity and mortality. The use of PCR for identification of infected infants and acyclovir for treatment has significantly improved the prognosis for affected infants. The subsequent use of suppressive therapy with oral acyclovir following completion of parenteral treatment of acute disease has further enhanced the long-term prognosis for these infants. This review article will discuss the epidemiology, risk factors and routes of acquisition, clinical presentation, and evaluation of an infant suspected to have the infection, and treatment of proven neonatal HSV disease.


Assuntos
Herpes Simples/tratamento farmacológico , Transmissão Vertical de Doenças Infecciosas , Complicações Infecciosas na Gravidez/tratamento farmacológico , Aciclovir/uso terapêutico , Antivirais/uso terapêutico , Cesárea , Parto Obstétrico/métodos , Coagulação Intravascular Disseminada/etiologia , Encefalite por Herpes Simples/diagnóstico , Encefalite por Herpes Simples/tratamento farmacológico , Encefalite por Herpes Simples/prevenção & controle , Extração Obstétrica , Membranas Extraembrionárias , Feminino , Herpes Genital/diagnóstico , Herpes Genital/transmissão , Herpes Simples/complicações , Herpes Simples/diagnóstico , Herpes Simples/prevenção & controle , Herpes Simples/transmissão , Herpesvirus Humano 1 , Herpesvirus Humano 2 , Humanos , Recém-Nascido , Ceratite Herpética/diagnóstico , Ceratite Herpética/tratamento farmacológico , Ceratite Herpética/prevenção & controle , Trabalho de Parto , Falência Hepática/etiologia , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico , Complicações Infecciosas na Gravidez/prevenção & controle , Prognóstico , Insuficiência Respiratória/etiologia , Fatores de Risco , Dermatopatias Virais/diagnóstico , Dermatopatias Virais/tratamento farmacológico , Dermatopatias Virais/prevenção & controle , Fatores de Tempo
4.
J Pediatric Infect Dis Soc ; 6(4): e177-e179, 2017 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-28379476

RESUMO

A 10-day-old child was treated for neonatal herpes simplex virus (HSV) central nervous system (CNS) disease with 21 days of intravenous acyclovir and 6 months of oral acyclovir. She presented 7 years later with HSV CNS disease and new lesions in her brain, illustrating the non-benign nature of delayed recurrent HSV CNS disease.


Assuntos
Aciclovir/uso terapêutico , Antivirais/uso terapêutico , Encefalite por Herpes Simples/etiologia , Aciclovir/administração & dosagem , Antivirais/administração & dosagem , Encéfalo/patologia , Encéfalo/virologia , Criança , Encefalite por Herpes Simples/diagnóstico por imagem , Encefalite por Herpes Simples/prevenção & controle , Feminino , Humanos , Recém-Nascido , Doenças do Recém-Nascido/virologia , Imageamento por Ressonância Magnética , Neuroimagem , Recidiva , Simplexvirus
6.
Mol Neurobiol ; 52(3): 1547-1560, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25367881

RESUMO

In this study, we tried to explore the molecular mechanism that Corilagin protected against herpes simplex virus-1 encephalitis through inhibiting the TLR2 signaling pathways in vivo and in vitro. As a result, Corilagin significantly prevented increase in the levels of TLR2 and its downstream mediators following Malp2 or HSV-1 challenge. On the other hand, in spite of TLR2 knockdown, Corilagin could still significantly suppress the expression of P38 and NEMO, phosphor-P38, and nuclear factor kappa B. The mRNA and protein expression of TLR2 and its downstream mediators in the brain tissue were also significantly lowered in mice treated with Corilagin. In addition, Corilagin inhibited expression of tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 protein. In conclusion, Corilagin shows the potential to protect against HSV-1-induced encephalitis, and the beneficial effects may be mediated by inhibiting TLR2 signaling pathways.


Assuntos
Antivirais/farmacologia , Encefalite por Herpes Simples/prevenção & controle , Glucosídeos/farmacologia , Herpesvirus Humano 1 , Taninos Hidrolisáveis/farmacologia , RNA Mensageiro/biossíntese , Transdução de Sinais/efeitos dos fármacos , Receptor 2 Toll-Like/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Interleucina-6/biossíntese , Interleucina-6/genética , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipopeptídeos/toxicidade , Masculino , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Microglia/efeitos dos fármacos , Microglia/metabolismo , Fator 88 de Diferenciação Mieloide/biossíntese , Fator 88 de Diferenciação Mieloide/genética , NF-kappa B/biossíntese , NF-kappa B/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Interferência de RNA , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Receptores de Interleucina-1/biossíntese , Receptores de Interleucina-1/genética , Fator 6 Associado a Receptor de TNF/biossíntese , Fator 6 Associado a Receptor de TNF/genética , Receptor 2 Toll-Like/biossíntese , Receptor 2 Toll-Like/genética , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
Clin Perinatol ; 41(4): 945-55, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25459782

RESUMO

Genital herpes simplex virus (HSV) infections are very common worldwide. Approximately 22% of pregnant women are infected genitally with HSV, and most of them are unaware of this. The most devastating consequence of maternal genital herpes is HSV disease in the newborn. Although neonatal HSV infections remain uncommon, due to the significant morbidity and mortality associated with the infection, HSV infection in the newborn is often considered in the differential diagnosis of ill neonates. This review summarizes the epidemiology and management of neonatal HSV infections and discusses strategies to prevent HSV infection in the newborn.


Assuntos
Antivirais/uso terapêutico , DNA Viral/análise , Encefalite por Herpes Simples/prevenção & controle , Herpes Simples/tratamento farmacológico , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Complicações Infecciosas na Gravidez/tratamento farmacológico , Cesárea , Encefalite por Herpes Simples/líquido cefalorraquidiano , Feminino , Herpes Simples/diagnóstico , Herpes Simples/prevenção & controle , Herpes Simples/transmissão , Humanos , Recém-Nascido , Reação em Cadeia da Polimerase , Gravidez , Complicações Infecciosas na Gravidez/diagnóstico , Complicações Infecciosas na Gravidez/prevenção & controle , Simplexvirus/genética
8.
Antiviral Res ; 96(3): 414-21, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23043942

RESUMO

We evaluated the effects of agonists and antagonist of toll-like receptor (TLR) 9 in comparison with a TLR3 agonist in a mouse model of herpes simplex virus type 1 (HSV-1) encephalitis (HSE). BALB/c mice received a single intranasal dose of either a TLR3 agonist (polyinosinic:polycytidylic acid; PIC), TLR9 agonists (oligodeoxynucleotides (ODNs) 1585, 1826 or 2395) or a TLR9 antagonist (ODN 2088), 1 day before and, for selected groups, 3 days after infection with HSV-1. Mice that received the pre-treatment with vehicle, PIC, ODNs 1585, 1826, 2395 and 2088 before infection had survival rates of 25%, 65%, 55%, 40%, 55% and 30%, respectively (P<0.05 for PIC and ODNs 1585 and 2395 versus vehicle). Infected mice subsequently treated with vehicle, ODNs 2395 and 2088 had survival rates of 9%, 0% and 30%, respectively (P<0.05, ODN 2088 versus other groups). The pre-treatment of mice with ODN 2395 reduced both the viral load (P<0.05 at day 5) and the production of CCL2, IL-6 and CCL5 at days 3, 4 and 5 (P<0.05 for IL-6 at day 3 and P<0.05 for CCL2 and CCL5 at day 4). Treatment of infected mice with ODN 2088 reduced the production of the same cytokines (P=0.07 for CCL2 and P=0.09 for IL-6 at day 5). Pre-treatment of mice with TLR9 agonists before infection reduces brain viral load and cytokine levels resulting in increased HSE survival rates. On the other hand, TLR9 antagonists can be helpful to control the inflammatory response that could be detrimental after infection.


Assuntos
Encefalite por Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/antagonistas & inibidores , Animais , Antivirais/farmacologia , Encéfalo/virologia , Quimiocinas/imunologia , DNA Viral/genética , Encefalite por Herpes Simples/tratamento farmacológico , Encefalite por Herpes Simples/prevenção & controle , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/patogenicidade , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Oligodesoxirribonucleotídeos/farmacologia , Poli I-C/farmacologia , Receptor 3 Toll-Like/agonistas , Carga Viral
9.
J Virol ; 85(24): 12972-81, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21994441

RESUMO

Innate immune deficiencies result in a spectrum of severe clinical outcomes following infection. In particular, there is a strong association between loss of the signal transducer and activator of transcription (Stat) pathway, breach of the blood-brain barrier (BBB), and virus-induced neuropathology. The gene signatures that characterize resistance, disease, and mortality in the virus-infected nervous system have not been defined. Herpes simplex virus type 1 (HSV-1) is commonly associated with encephalitis in humans, and humans and mice lacking Stat1 display increased susceptibility to HSV central nervous system (CNS) infections. In this study, two HSV-1 strains were used, KOS (wild type [WT]), and Δvhs, an avirulent recombinant lacking the virion host shutoff (vhs) function. In addition, two mouse strains were used: strain 129 (control) and a Stat1-deficient (Stat1(-/-)) strain. Using combinations of these virus and mouse strains, we established a model of infection resulting in three different outcomes: viral clearance without neurological disease (Δvhs infection of control mice), neurological disease followed by viral clearance (Δvhs infection of Stat1(-/-) mice and WT infection of control mice), or neurological disease followed by death (WT infection of Stat1(-/-) mice). Through the use of functional genomics on the infected brain stems, we determined gene signatures that were representative of the three infection outcomes. We demonstrated a pathological signature in the brain stem of Stat1-deficient mice characterized by upregulation of transcripts encoding chemokine receptors, inflammatory markers, neutrophil chemoattractants, leukocyte adhesion proteins, and matrix metalloproteases. Additionally, there was a greater than 100-fold increase in the inflammatory markers interleukin 1ß (IL-1ß) and IL-6. Consistent with this gene signature, we demonstrated profound CNS inflammation with a concomitant lethal breach of the BBB. Taken together, our results indicated an essential role for normal Stat1-dependent signaling in mediating a nonpathological immune response to viral CNS infection.


Assuntos
Encefalite por Herpes Simples/imunologia , Encefalite por Herpes Simples/prevenção & controle , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/patogenicidade , Ceratite Herpética/imunologia , Ceratite Herpética/virologia , Fator de Transcrição STAT1/imunologia , Animais , Tronco Encefálico/patologia , Tronco Encefálico/virologia , Citocinas/biossíntese , Perfilação da Expressão Gênica , Histocitoquímica , Ceratite Herpética/complicações , Camundongos , Camundongos Knockout , Análise em Microsséries , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida
10.
PLoS Pathog ; 7(6): e1002071, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21655109

RESUMO

HSV-1 is the leading cause of sporadic encephalitis in humans. HSV infection of susceptible 129S6 mice results in fatal encephalitis (HSE) caused by massive inflammatory brainstem lesions comprising monocytes and neutrophils. During infection with pathogenic microorganisms or autoimmune disease, IgGs induce proinflammatory responses and recruit innate effector cells. In contrast, high dose intravenous immunoglobulins (IVIG) are an effective treatment for various autoimmune and inflammatory diseases because of potent anti-inflammatory effects stemming in part from sialylated IgGs (sIgG) present at 1-3% in IVIG. We investigated the ability of IVIG to prevent fatal HSE when given 24 h post infection. We discovered a novel anti-inflammatory pathway mediated by low-dose IVIG that protected 129S6 mice from fatal HSE by modulating CNS inflammation independently of HSV specific antibodies or sIgG. IVIG suppressed CNS infiltration by pathogenic CD11b(+) Ly6C(high) monocytes and inhibited their spontaneous degranulation in vitro. FcγRIIb expression was required for IVIG mediated suppression of CNS infiltration by CD45(+) Ly6C(low) monocytes but not for inhibiting development of Ly6C(high) monocytes. IVIG increased accumulation of T cells in the CNS, and the non-sIgG fraction induced a dramatic expansion of FoxP3(+) CD4(+) T regulatory cells (Tregs) and FoxP3(-) ICOS(+) CD4(+) T cells in peripheral lymphoid organs. Tregs purified from HSV infected IVIG treated, but not control, mice protected adoptively transferred mice from fatal HSE. IL-10, produced by the ICOS(+) CD4(+) T cells that accumulated in the CNS of IVIG treated, but not control mice, was essential for induction of protective anti-inflammatory responses. Our results significantly enhance understanding of IVIG's anti-inflammatory and immunomodulatory capabilities by revealing a novel sIgG independent anti-inflammatory pathway responsible for induction of regulatory T cells that secrete the immunosuppressive cytokine IL-10 and further reveal the therapeutic potential of IVIG for treating viral induced inflammatory diseases.


Assuntos
Encefalite por Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Interleucina-10/metabolismo , Animais , Antígenos Ly/metabolismo , Barreira Hematoencefálica/imunologia , Tronco Encefálico/patologia , Linfócitos T CD4-Positivos/imunologia , Encefalite por Herpes Simples/mortalidade , Encefalite por Herpes Simples/prevenção & controle , Encefalite por Herpes Simples/virologia , Citometria de Fluxo , Herpesvirus Humano 1/patogenicidade , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Imunoglobulinas Intravenosas/imunologia , Interleucina-10/administração & dosagem , Interleucina-10/imunologia , Leucócitos/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Monócitos/imunologia , Neutrófilos/imunologia , Linfócitos T Reguladores/imunologia , Fatores de Tempo
11.
J Virol ; 85(4): 1793-803, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21123390

RESUMO

Herpes simplex virus (HSV) glycoprotein B (gB) is an integral part of the multicomponent fusion system required for virus entry and cell-cell fusion. Here we investigated the mechanism of viral neutralization by the monoclonal antibody (MAb) 2c, which specifically recognizes the gB of HSV type 1 (HSV-1) and HSV-2. Binding of MAb 2c to a type-common discontinuous epitope of gB resulted in highly efficient neutralization of HSV at the postbinding/prefusion stage and completely abrogated the viral cell-to-cell spread in vitro. Mapping of the antigenic site recognized by MAb 2c to the recently solved crystal structure of the HSV-1 gB ectodomain revealed that its discontinuous epitope is only partially accessible within the observed multidomain trimer conformation of gB, likely representing its postfusion conformation. To investigate how MAb 2c may interact with gB during membrane fusion, we characterized the properties of monovalent (Fab and scFv) and bivalent [IgG and F(ab')(2)] derivatives of MAb 2c. Our data show that the neutralization capacity of MAb 2c is dependent on cross-linkage of gB trimers. As a result, only bivalent derivatives of MAb 2c exhibited high neutralizing activity in vitro. Notably, bivalent MAb 2c not only was capable of preventing mucocutaneous disease in severely immunodeficient NOD/SCID mice upon vaginal HSV-1 challenge but also protected animals even with neuronal HSV infection. We also report for the first time that an anti-gB specific monoclonal antibody prevents HSV-1-induced encephalitis entirely independently from complement activation, antibody-dependent cellular cytotoxicity, and cellular immunity. This indicates the potential for further development of MAb 2c as an anti-HSV drug.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 2/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/química , Especificidade de Anticorpos , Células COS , Chlorocebus aethiops , Encefalite por Herpes Simples/imunologia , Encefalite por Herpes Simples/prevenção & controle , Mapeamento de Epitopos , Epitopos/imunologia , Feminino , Herpes Genital/imunologia , Herpes Genital/prevenção & controle , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Moleculares , Testes de Neutralização , Mapeamento de Peptídeos , Células Vero , Proteínas do Envelope Viral/química
12.
J Immunol ; 181(12): 8604-12, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19050280

RESUMO

Viruses are recognized by the innate immune system through pattern recognition receptors (PRRs). For instance, HSV virions and genomic DNA are recognized by TLR2 and TLR9, respectively. Although several viruses and viral components have been shown to stimulate cells through TLRs, only very few studies have defined essential roles for single TLRs in innate immune defense in vivo. This could suggest that PRRs act in concert to mount the first line of defense against virus infections. To test this hypothesis we have examined the host response of C57BL/6, TLR2(-/-), TLR9(-/-), and TLR2/9(-/-) mice toward HSV-2 infection. After a systemic infection, the cytokine serum response was markedly reduced in the double knockout mice, but only partly affected in either strain of the single knockout mice. This was supported by in vitro data showing that HSV-induced cytokine expression relayed on TLR2 and TLR9 in a cytokine- and cell type-dependent manner. With respect to the cellular response to infection, we found that recruitment but not activation of NK cells was impaired in TLR2/9(-/-) mice. Importantly, the viral load in the brain, but not liver, was significantly higher in the brain of TLR2/9(-/-) mice whereas the viral loads in organs of single knockout mice were statistically indistinguishable from C57BL/6 mice. In the brain we found that TNF-alpha and the IFN-stimulated gene CXCL9 were expressed during infection and were dependent on either TLR2 or TLR9. Thus, TLR2 and TLR9 synergistically stimulate innate antiviral activities, thereby protecting against HSV infection in the brain.


Assuntos
Encefalite por Herpes Simples/imunologia , Encefalite por Herpes Simples/prevenção & controle , Herpesvirus Humano 2/imunologia , Receptor 2 Toll-Like/fisiologia , Receptor Toll-Like 9/fisiologia , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Células Cultivadas , Citocinas/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Encefalite por Herpes Simples/virologia , Feminino , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/fisiologia , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética
13.
J Infect Dis ; 195(6): 817-25, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17299711

RESUMO

BACKGROUND: Glucocorticoids (GCs) have not been established as an evidence-based therapy for herpes simplex virus (HSV) encephalitis. We sought to evaluate the effects of early versus delayed GC administration in a model of HSV-1 encephalitis. METHODS: Mice were inoculated intranasally with a clinical HSV-1 strain and had access to corticosterone (Cort; 0.2 mg/mL) in drinking water or received dexamethasone (DEX; 10 mg/kg) intraperitoneally. Cort was started immediately or 72 h after infection, whereas DEX was administrated 3 days after infection. RESULTS: Expression of the thymidine kinase transcript indicated widespread viral replication in the brain stem and many regions of the mediovestibular system. This neurovirulence provoked neuronal death and transcriptional activation of several immune genes. Notably, Toll-like receptors 2, 3, and 9 transcripts were strongly up-regulated. Mean life expectancy was higher in the group of mice that received delayed Cort (56%) than in the mice that received no Cort (44%) or early Cort treatments (13%) (P<.05, early vs. delayed or no Cort treatments). Delayed DEX treatment suppressed not only the expression of inflammatory genes as expected but also that of viral genes. CONCLUSIONS: Administration of GCs at a critical time during viral infection is associated with neuroprotection and survival in experimental HSV-1 encephalitis.


Assuntos
Corticosterona/uso terapêutico , Dexametasona/uso terapêutico , Encefalite por Herpes Simples/prevenção & controle , Glucocorticoides/uso terapêutico , Animais , Modelos Animais de Doenças , Esquema de Medicação , Feminino , Herpesvirus Humano 1/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Sobrevida , Replicação Viral
15.
Pediatrics ; 115(3): 804-9, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15741392

RESUMO

Cutaneous herpes simplex virus type 2 (HSV-2) infection was recognized at 19 days of age in a 1415-g female infant born at 31 weeks of gestation. Cerebrospinal fluid (CSF) HSV polymerase chain reaction (PCR) was negative, and MRI of the brain was normal. After a 14-day course of high-dose intravenous acyclovir, the infant developed a cutaneous recurrence at 38 days of age. CSF HSV PCR again was negative. She was subsequently begun on oral acyclovir to prevent cutaneous reactivation of HSV. At 3 months of age, the infant developed HSV encephalitis as manifested by fever, seizures, abnormal CSF indices, abnormal brain MRI, and positive CSF HSV PCR. No cutaneous disease was present. It is not known whether the HSV encephalitis in our patient represented reactivation of previously unrecognized central nervous system infection or new onset of central nervous system disease as a result of spread from other tissue or site to the brain. The failure of oral acyclovir to prevent such an occurrence, however, highlights gaps in our understanding of the pathogenesis of neonatal HSV disease and questions the use of acyclovir suppression to prevent neurologic sequelae.


Assuntos
Aciclovir/uso terapêutico , Antivirais/uso terapêutico , Encefalite por Herpes Simples/prevenção & controle , Herpes Simples/tratamento farmacológico , Herpesvirus Humano 2 , Doenças do Prematuro/tratamento farmacológico , Administração Oral , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Encefalite por Herpes Simples/complicações , Encefalite por Herpes Simples/patologia , Feminino , Herpesvirus Humano 2/isolamento & purificação , Herpesvirus Humano 2/fisiologia , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Injeções Intravenosas , Imageamento por Ressonância Magnética , Reação em Cadeia da Polimerase , Radiografia , Recidiva , Convulsões/etiologia , Ativação Viral/efeitos dos fármacos
16.
J Immunol ; 173(12): 7575-83, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15585885

RESUMO

Herpetic stromal keratitis (HSK), resulting from corneal HSV-1 infection, represents a T cell-mediated immunopathologic lesion. In T cell transgenic mice on a SCID or RAG knockout background, the T cells mediating lesions are unreactive to viral Ags. In these bystander models, animals develop ocular lesions but are unable to control infection. Transfer of HSV-immune cells into a CD8(+) T cell bystander model resulted in clearance of virus from eyes, animals survived, and lesions developed to greater severity. However, the adoptively transferred CD8(+) T cells were not evident in lesions, although they were readily detectable in the lymphoid tissues as well as in the peripheral and CNS. Our results indicate that viral-induced tissue damage can be caused by bystander cells, but these fail to control infection. Immune CD8(+) T cells trigger clearance of virus from the eye, but this appears to result by the T cells acting at sites distal to the cornea. A case is made that CD8(+) T cell control is expressed in the trigeminal ganglion, serving to curtail a source of virus to the cornea.


Assuntos
Efeito Espectador/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Substância Própria/imunologia , Substância Própria/patologia , Ceratite Herpética/patologia , Ceratite Herpética/prevenção & controle , Simplexvirus/imunologia , Transferência Adotiva , Animais , Efeito Espectador/genética , Linfócitos T CD8-Positivos/transplante , Doença Crônica , Substância Própria/virologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Encefalite por Herpes Simples/genética , Encefalite por Herpes Simples/imunologia , Encefalite por Herpes Simples/mortalidade , Encefalite por Herpes Simples/prevenção & controle , Feminino , Proteínas de Homeodomínio/genética , Ceratite Herpética/genética , Ceratite Herpética/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Carga Viral
17.
J Virol Methods ; 120(2): 161-5, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15288958

RESUMO

Ascorbate is an important antioxidant. However, in the presence of transition metals such as Cu(II) or Fe(III), it also has pro-oxidant capabilities. The effect of ascorbate-Cu(II) in the in vitro infection of herpes simplex virus type 2 (HSV-2) and its protecting effect in a murine model was investigated. HSV-2 was treated with different concentrations of ascorbate in the presence of Cu(II). A group of CF-1 mice were treated with the inactivated virus and other treated with maintenance medium containing only ascorbate-Cu(II). Weeks later, mice were challenged intranasally with infectious viruses. HSV-2 was completely inactivated by 2mM ascorbate plus 1mM Cu(II). Ascorbate or Cu(II) alone did not inactivate the virus. Compared with the control group, 60% of the immunized animals did not show any sign of encephalitis and survived the herpes virus infection, while a 7% survival rate was observed in the control group (P = 0.056). We concluded that the in vitro treatment of HSV-2 with ascorbate-Cu(II) is not only able to inactivate the virus, but also suggested that the viral particles induced a protective response against herpes encephalitis. This inactivation may provide an alternative method to develop new agents therapeutics.


Assuntos
Ácido Ascórbico/farmacologia , Cobre/farmacologia , Encefalite por Herpes Simples/prevenção & controle , Herpesvirus Humano 2/efeitos dos fármacos , Vacinas contra Herpesvirus/administração & dosagem , Inativação de Vírus , Animais , Ácido Ascórbico/administração & dosagem , Chlorocebus aethiops , Cobre/administração & dosagem , Encefalite por Herpes Simples/mortalidade , Encefalite por Herpes Simples/virologia , Herpes Simples/mortalidade , Herpes Simples/prevenção & controle , Herpes Simples/virologia , Herpesvirus Humano 2/crescimento & desenvolvimento , Vacinas contra Herpesvirus/imunologia , Imunização , Masculino , Camundongos , Células Vero
18.
J Neuroimmunol ; 152(1-2): 5-10, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15223232

RESUMO

We examined whether immunization with the nonpathogenic strain R-15 of herpes simplex virus-1 (HSV-1) may prevent the clinical and neuroendocrine changes induced by the pathogenic HSV-1 strain Syn17+. Inoculation of strain Syn17+ to control rats induced fever, marked motor hyperactivity and aggressive behavior, and increased serum ACTH, corticosterone (CS) and brain prostaglandin-E2 production. Mortality was 100%. Immunization with strain R-15 prior to challenge with Syn17+ induced the production of neutralizing antibodies to HSV-1 Syn17+, and abolished the above clinical and neuroendocrine changes. Mortality was completely prevented. These results indicate that immunization with HSV-1 strain R-15 protects rats from lethal HSV-1 encephalitis and prevents its clinical and neurochemical manifestations.


Assuntos
Encefalite por Herpes Simples/prevenção & controle , Herpesvirus Humano 1/imunologia , Sistemas Neurossecretores/fisiologia , Vacinação , Hormônio Adrenocorticotrópico/sangue , Animais , Comportamento Animal , Encéfalo/metabolismo , Corticosterona/sangue , Modelos Animais de Doenças , Encefalite por Herpes Simples/complicações , Masculino , Prostaglandinas/análise , Ratos
19.
J Neuroimmunol ; 140(1-2): 13-27, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12864968

RESUMO

Despite the generally restrictive nature of the blood-brain barrier (BBB), circulating lymphocytes can infiltrate into the central nervous system (CNS) during a variety of disease states. Although the contributions of these lymphocytes to CNS-associated disease have been identified in some viral models, the factors which govern this infiltration following herpes simplex virus (HSV) infection remain to be elucidated. We have developed a murine model of HSV encephalitis (HSE) to define the relationship among psychological stress, the recruitment of HSV-specific T cells into the CNS, and the development of HSE. Naive mice, as well as mice that had been vaccinated with a recombinant vaccinia virus (rVVESgB498-505) that elicits the generation of HSV-1 gB498-505-specific CD8(+) T cells, were infected intranasally (i.n.) with HSV-1 McIntyre. Beginning one day prior to HSV-1 infection and continuing for a total of 9 days, naive and vaccinated mice were exposed to a well-established stressor, restraint stress. Naive, stressed mice exhibited increased symptoms of HSE and HSE-associated mortality as compared to non-stressed controls. A concomitant increase in CD4(+) and CD8(+) T cells in the brain was observed throughout the infection, with CD8(+) T cells outnumbering CD4(+) T cells. The development of HSE in these naive, stressed mice was accompanied by a delayed infiltration of gB498-505-specific CD8(+) T cells after HSV spread into the brain. In contrast, both stressed and non-stressed rVVESgB498-505-vaccinated mice possessed gB498-505-specific CD8(+) T cells prior to HSV challenge and were protected against HSE despite having detectable HSV-1 DNA in the brain. Together, these findings suggest that a delayed infiltration of CD8(+) T cells into the brain may promote HSE in naive mice, while the presence of HSV-specific CD8(+) T cells in the brain prior to HSV challenge is protective, possibly by limiting HSV replication and spread within the CNS.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Encefalite por Herpes Simples/imunologia , Herpesvirus Humano 1 , Estresse Fisiológico/imunologia , Administração Intranasal , Animais , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Relação CD4-CD8 , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Movimento Celular/imunologia , Chlorocebus aethiops , Modelos Animais de Doenças , Encefalite por Herpes Simples/patologia , Encefalite por Herpes Simples/fisiopatologia , Encefalite por Herpes Simples/prevenção & controle , Epitopos de Linfócito T/imunologia , Herpesvirus Humano 1/crescimento & desenvolvimento , Herpesvirus Humano 1/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vacinas Sintéticas/administração & dosagem , Vírus Vaccinia/imunologia , Células Vero , Vacinas Virais/administração & dosagem
20.
J Infect Dis ; 185(11): 1550-60, 2002 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12023760

RESUMO

Herpes simplex virus (HSV) causes devastating infections in newborns. Maternal immunization is one potential strategy to reduce neonatal HSV disease. Female mice were immunized with an HSV type 2 (HSV-2) replication-defective mutant (HSV-2 dl5-29, which is defective for the genes UL5 and UL29) and then mated. Protection was evaluated in newborn mice after a virulent HSV-2 oral challenge. Heightened neonatal susceptibility was observed to a thymidine kinase-negative HSV-2 strain (HSV-2 186DeltaKpn) that is highly attenuated in adult mice. Maternal immunization with HSV-2 dl5-29 and HSV-2 186DeltaKpn reduced visceral spread of infectious challenge virus in pups after challenge at either 1 day or 1 week of age but did not prevent replication at the site of entry, spread to the central nervous system, or lethal encephalitis. No protection was seen in pups born to mock-immunized mothers or to mothers immunized with a UV-inactivated wild-type HSV-2 strain. Levels of protection correlated with levels of passively transferred maternal HSV-2-specific IgG antibody.


Assuntos
Vírus Defeituosos/imunologia , Encefalite por Herpes Simples/prevenção & controle , Herpes Genital/prevenção & controle , Herpesvirus Humano 2/imunologia , Complicações Infecciosas na Gravidez/prevenção & controle , Vacinas Virais/administração & dosagem , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Feminino , Herpesvirus Humano 2/genética , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Gravidez , Vacinas Virais/imunologia , Replicação Viral , Vísceras/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...