Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 15(8): e0237189, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32760124

RESUMO

Herpes simplex virus 1 (HSV) is a ubiquitous human virus resident in a majority of the global population as a latent infection. Acyclovir (ACV), is the standard of care drug used to treat primary and recurrent infections, supplemented in some patients with intravenous immunoglobulin (IVIG) treatment to suppress infection and deleterious inflammatory responses. As many diverse medications have recently been shown to change composition of the gut microbiome, we used Illumina 16S rRNA gene sequencing to determine the effects of ACV and IVIG on the gut bacterial community. We found that HSV, ACV and IVIG can all independently disrupt the gut bacterial community in a sex biased manner when given to uninfected C57BL/6 mice. Treatment of HSV infected mice with ACV or IVIG alone or together revealed complex interactions between these drugs and infection that caused pronounced sex biased dysbiosis. ACV reduced Bacteroidetes levels in male but not female mice, while levels of the Anti-inflammatory Clostridia (AIC) were reduced in female but not male mice, which is significant as these taxa are associated with protection against the development of graft versus host disease (GVHD) in hematopoietic stem cell transplant (HSCT) patients. Gut barrier dysfunction is associated with GVHD in HSCT patients and ACV also decreased Akkermansia muciniphila, which is important for maintaining gut barrier functionality. Cumulatively, our data suggest that long-term prophylactic ACV treatment of HSCT patients may contribute to GVHD and also potentially impact immune reconstitution. These data have important implications for other clinical settings, including HSV eye disease and genital infections, where ACV is given long-term.


Assuntos
Aciclovir/efeitos adversos , Antivirais/efeitos adversos , Disbiose/etiologia , Herpes Simples/microbiologia , Imunoglobulinas Intravenosas/efeitos adversos , Aciclovir/uso terapêutico , Animais , Antivirais/uso terapêutico , Bacteroidetes/patogenicidade , Clostridium/patogenicidade , Disbiose/microbiologia , Feminino , Microbioma Gastrointestinal , Herpes Simples/tratamento farmacológico , Imunoglobulinas Intravenosas/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores Sexuais
2.
Nat Commun ; 10(1): 2153, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-31089128

RESUMO

The gut commensal Bacteroides fragilis or its capsular polysaccharide A (PSA) can prevent various peripheral and CNS sterile inflammatory disorders. Fatal herpes simplex encephalitis (HSE) results from immune pathology caused by uncontrolled invasion of the brainstem by inflammatory monocytes and neutrophils. Here we assess the immunomodulatory potential of PSA in HSE by infecting PSA or PBS treated 129S6 mice with HSV1, followed by delayed Acyclovir (ACV) treatment as often occurs in the clinical setting. Only PSA-treated mice survived, with dramatically reduced brainstem inflammation and altered cytokine and chemokine profiles. Importantly, PSA binding by B cells is essential for induction of regulatory CD4+ and CD8+ T cells secreting IL-10 to control innate inflammatory responses, consistent with the lack of PSA mediated protection in Rag-/-, B cell- and IL-10-deficient mice. Our data reveal the translational potential of PSA as an immunomodulatory symbiosis factor to orchestrate robust protective anti-inflammatory responses during viral infections.


Assuntos
Bacteroides fragilis/imunologia , Encefalite por Herpes Simples/imunologia , Microbioma Gastrointestinal/imunologia , Herpesvirus Humano 1/imunologia , Polissacarídeos Bacterianos/imunologia , Aciclovir/uso terapêutico , Animais , Antivirais/uso terapêutico , Linfócitos B/imunologia , Linfócitos B/metabolismo , Bacteroides fragilis/metabolismo , Chlorocebus aethiops , Modelos Animais de Doenças , Encefalite por Herpes Simples/tratamento farmacológico , Encefalite por Herpes Simples/virologia , Feminino , Herpesvirus Humano 1/patogenicidade , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-10/metabolismo , Masculino , Camundongos , Camundongos Knockout , Polissacarídeos Bacterianos/metabolismo , Simbiose/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Células Vero
3.
Nat Immunol ; 20(2): 152-162, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30643259

RESUMO

Stimulator of interferon genes (STING) is an endoplasmic reticulum (ER) signaling adaptor that is essential for the type I interferon response to DNA pathogens. Aberrant activation of STING is linked to the pathology of autoimmune and autoinflammatory diseases. The rate-limiting step for the activation of STING is its translocation from the ER to the ER-Golgi intermediate compartment. Here, we found that deficiency in the Ca2+ sensor stromal interaction molecule 1 (STIM1) caused spontaneous activation of STING and enhanced expression of type I interferons under resting conditions in mice and a patient with combined immunodeficiency. Mechanistically, STIM1 associated with STING to retain it in the ER membrane, and coexpression of full-length STIM1 or a STING-interacting fragment of STIM1 suppressed the function of dominant STING mutants that cause autoinflammatory diseases. Furthermore, deficiency in STIM1 strongly enhanced the expression of type I interferons after viral infection and prevented the lethality of infection with a DNA virus in vivo. This work delineates a STIM1-STING circuit that maintains the resting state of the STING pathway.


Assuntos
Interferon Tipo I/imunologia , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Animais , Pré-Escolar , Chlorocebus aethiops , DNA Viral/imunologia , Modelos Animais de Doenças , Retículo Endoplasmático/metabolismo , Fibroblastos , Técnicas de Inativação de Genes , Células HEK293 , Herpes Simples/imunologia , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Humanos , Imunidade Inata , Células Jurkat , Macrófagos , Masculino , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Células NIH 3T3 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Cultura Primária de Células , Imunodeficiência Combinada Severa/sangue , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/imunologia , Células Vero
4.
Sci Rep ; 9(1): 20324, 2019 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-31889131

RESUMO

Chemotherapy induced peripheral neuropathy (CIPN), a toxic side effect of some cancer treatments, negatively impacts patient outcomes and drastically reduces survivor's quality of life (QOL). Uncovering the mechanisms driving chemotherapy-induced CIPN is urgently needed to facilitate the development of effective treatments, as currently there are none. Observing that C57BL/6 (B6) and 129SvEv (129) mice are respectively sensitive and resistant to Paclitaxel-induced pain, we investigated the involvement of the gut microbiota in this extreme phenotypic response. Reciprocal gut microbiota transfers between B6 and 129 mice as well as antibiotic depletion causally linked gut microbes to Paclitaxel-induced pain sensitivity and resistance. Microglia proliferated in the spinal cords of Paclitaxel treated mice harboring the pain-sensitive B6 microbiota but not the pain-resistant 129 microbiota, which exhibited a notable absence of infiltrating immune cells. Paclitaxel decreased the abundance of Akkermansia muciniphila, which could compromise barrier integrity resulting in systemic exposure to bacterial metabolites and products - that acting via the gut-immune-brain axis - could result in altered brain function. Other bacterial taxa that consistently associated with both bacteria and pain as well as microglia and pain were identified, lending support to our hypothesis that microglia are causally involved in CIPN, and that gut bacteria are drivers of this phenotype.


Assuntos
Antineoplásicos/efeitos adversos , Microbioma Gastrointestinal , Neuralgia/etiologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biodiversidade , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Masculino , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neuralgia/metabolismo , Paclitaxel/efeitos adversos , Paclitaxel/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia
5.
PLoS Pathog ; 14(1): e1006822, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29352287

RESUMO

Emergency hematopoiesis facilitates the rapid expansion of inflammatory immune cells in response to infections by pathogens, a process that must be carefully regulated to prevent potentially life threatening inflammatory responses. Here, we describe a novel regulatory role for the cytokine IFNγ that is critical for preventing fatal encephalitis after viral infection. HSV1 encephalitis (HSE) is triggered by the invasion of the brainstem by inflammatory monocytes and neutrophils. In mice lacking IFNγ (GKO), we observed unrestrained increases in G-CSF levels but not in GM-CSF or IL-17. This resulted in uncontrolled expansion and infiltration of apoptosis-resistant, degranulating neutrophils into the brainstem, causing fatal HSE in GKO but not WT mice. Excessive G-CSF in GKO mice also induced granulocyte derived suppressor cells, which inhibited T-cell proliferation and function, including production of the anti-inflammatory cytokine IL-10. Unexpectedly, we found that IFNγ suppressed G-CSF signaling by increasing SOCS3 expression in neutrophils, resulting in apoptosis. Depletion of G-CSF, but not GM-CSF, in GKO mice induced neutrophil apoptosis and reinstated IL-10 secretion by T cells, which restored their ability to limit innate inflammatory responses resulting in protection from HSE. Our studies reveals a novel, complex interplay among IFNγ, G-CSF and IL-10, which highlights the opposing roles of G-CSF and IFNγ in regulation of innate inflammatory responses in a murine viral encephalitis model and reveals G-CSF as a potential therapeutic target. Thus, the antagonistic G-CSF-IFNγ interactions emerge as a key regulatory node in control of CNS inflammatory responses to virus infection.


Assuntos
Proliferação de Células/genética , Sistema Nervoso Central/virologia , Quimiotaxia de Leucócito/genética , Encefalite Viral/prevenção & controle , Fator Estimulador de Colônias de Granulócitos/antagonistas & inibidores , Interferon gama/fisiologia , Neutrófilos/fisiologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Encefalite Viral/genética , Encefalite Viral/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fator Estimulador de Colônias de Granulócitos/fisiologia , Interferon gama/genética , Interferon gama/farmacologia , Masculino , Camundongos , Camundongos Knockout , Neutrófilos/efeitos dos fármacos
6.
Behav Neurol ; 2017: 5238402, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29358844

RESUMO

Herpes simplex virus 1 (HSV) encephalitis (HSE) has serious neurological complications, involving behavioral and cognitive impairments that cause significant morbidity and a reduced quality of life. We showed that HSE results from dysregulated central nervous system (CNS) inflammatory responses. We hypothesized that CNS inflammation is casually involved in behavioral abnormalities after HSE and that treatment with ACV and pooled human immunoglobulin (IVIG), an immunomodulatory drug, would improve outcomes compared to mice treated with phosphate buffered saline (PBS) or ACV alone. Anxiety levels were high in HSV-infected PBS and ACV-treated mice compared to mice treated with ACV + IVIG, consistent with reports implicating inflammation in anxiety induced by lipopolysaccharide (LPS) or stress. Female, but not male, PBS-treated mice were cognitively impaired, and unexpectedly, ACV was protective, while the inclusion of IVIG surprisingly antagonized ACV's beneficial effects. Distinct serum proteomic profiles were observed for male and female mice, and the antagonistic effects of ACV and IVIG on behavior were paralleled by similar changes in the serum proteome of ACV- and ACV + IVIG-treated mice. We conclude that inflammation and other factors mediate HSV-induced behavioral impairments and that the effects of ACV and IVIG on behavior involve novel mechanisms.


Assuntos
Aciclovir/farmacologia , Encefalite por Herpes Simples/tratamento farmacológico , Imunoglobulinas Intravenosas/farmacologia , Animais , Feminino , Herpesvirus Humano 1/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Blood ; 125(24): 3720-30, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-25931583

RESUMO

MicroRNAs (miRNAs) are a class of powerful posttranscriptional regulators implicated in the control of diverse biological processes, including regulation of hematopoiesis and the immune response. To define the biological functions of miR-142, which is preferentially and abundantly expressed in immune cells, we created a mouse line with a targeted deletion of this gene. Our analysis of miR-142(-/-) mice revealed a critical role for this miRNA in the development and homeostasis of lymphocytes. Marginal zone B cells expand in the knockout spleen, whereas the number of T and B1 B cells in the periphery is reduced. Abnormal development of hematopoietic lineages in miR-142(-/-) animals is accompanied by a profound immunodeficiency, manifested by hypoimmunoglobulinemia and failure to mount a productive immune response to soluble antigens and virus. miR-142(-/-) B cells express elevated levels of B-cell-activating factor (BAFF) receptor (BAFF-R) and as a result proliferate more robustly in response to BAFF stimulation. Lowering the BAFF-R gene dose in miR-142(-/-) mice rescues the B-cell expansion defect, suggesting that BAFF-R is a bona fide miR-142 target through which it controls B-cell homeostasis. Collectively, our results uncover miR-142 as an essential regulator of lymphopoiesis, and suggest that lesions in this miRNA gene may lead to primary immunodeficiency.


Assuntos
Linfócitos B/patologia , Deleção de Genes , Síndromes de Imunodeficiência/genética , Transtornos Imunoproliferativos/genética , Linfopoese , MicroRNAs/genética , Animais , Receptor do Fator Ativador de Células B/genética , Linfócitos B/imunologia , Linfócitos B/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Imunidade Celular , Imunidade Humoral , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/patologia , Transtornos Imunoproliferativos/imunologia , Transtornos Imunoproliferativos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/imunologia
8.
PLoS Pathog ; 11(3): e1004730, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25760441

RESUMO

The establishment of latent infections in sensory neurons is a remarkably effective immune evasion strategy that accounts for the widespread dissemination of life long Herpes Simplex Virus type 1 (HSV1) infections in humans. Periodic reactivation of latent virus results in asymptomatic shedding and transmission of HSV1 or recurrent disease that is usually mild but can be severe. An in-depth understanding of the mechanisms regulating the maintenance of latency and reactivation are essential for developing new approaches to block reactivation. However, the lack of a reliable mouse model that supports efficient in vivo reactivation (IVR) resulting in production of infectious HSV1 and/or disease has hampered progress. Since HSV1 reactivation is enhanced in immunosuppressed hosts, we exploited the antiviral and immunomodulatory activities of IVIG (intravenous immunoglobulins) to promote survival of latently infected immunodeficient Rag mice. Latently infected Rag mice derived by high dose (HD), but not low dose (LD), HSV1 inoculation exhibited spontaneous reactivation. Following hyperthermia stress (HS), the majority of HD inoculated mice developed HSV1 encephalitis (HSE) rapidly and synchronously, whereas for LD inoculated mice reactivated HSV1 persisted only transiently in trigeminal ganglia (Tg). T cells, but not B cells, were required to suppress spontaneous reactivation in HD inoculated latently infected mice. Transfer of HSV1 memory but not OVA specific or naïve T cells prior to HS blocked IVR, revealing the utility of this powerful Rag latency model for studying immune mechanisms involved in control of reactivation. Crossing Rag mice to various knockout strains and infecting them with wild type or mutant HSV1 strains is expected to provide novel insights into the role of specific cellular and viral genes in reactivation, thereby facilitating identification of new targets with the potential to block reactivation.


Assuntos
Modelos Animais de Doenças , Herpesvirus Humano 1/fisiologia , Síndromes de Imunodeficiência/virologia , Ativação Viral/fisiologia , Latência Viral/fisiologia , Transferência Adotiva , Animais , Herpes Simples/imunologia , Herpes Simples/virologia , Síndromes de Imunodeficiência/imunologia , Hibridização in Situ Fluorescente , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
J Gen Virol ; 96(Pt 6): 1347-1357, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25667322

RESUMO

West Nile virus (WNV), an important global human pathogen, targets neurons to cause lethal encephalitis, primarily in elderly and immunocompromised patients. Currently, there are no approved therapeutic agents or vaccines to treat WNV encephalitis. Recent studies have suggested that inflammation is a major contributor to WNV encephalitis morbidity. In this study we evaluated the use of IVIG (intravenous immunoglobulins - a clinical product comprising pooled human IgG) as an anti-inflammatory treatment in a model of lethal WNV infection. We report here that IVIG and pooled human WNV convalescent sera (WNV-IVIG) inhibited development of lethal WNV encephalitis by suppressing central nervous system (CNS) infiltration by CD45(high) leukocytes. Pathogenic Ly6C(high) CD11b(+) monocytes were the major infiltrating subset in the CNS of infected control mice, whereas IVIG profoundly reduced infiltration of these pathogenic Ly6C(high) monocytes into the CNS of infected mice. Interestingly, WNV-IVIG was more efficacious than IVIG in controlling CNS inflammation when mice were challenged with a high-dose inoculum (10(5) versus 10(4) p.f.u.) of WNV. Importantly, adsorption of WNV E-glycoprotein neutralizing antibodies did not abrogate IVIG protection, consistent with virus neutralization not being essential for IVIG protection. These findings confirmed the potent immunomodulatory activity of generic IVIG, and emphasized its potential as an effective immunotherapeutic drug for encephalitis and other virus induced inflammatory diseases.


Assuntos
Anticorpos Antivirais/administração & dosagem , Imunoglobulinas Intravenosas/administração & dosagem , Febre do Nilo Ocidental/prevenção & controle , Vírus do Nilo Ocidental/imunologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Humanos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Monócitos/imunologia , Análise de Sobrevida , Resultado do Tratamento
11.
Future Virol ; 8(3): 259-272, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23956785

RESUMO

HSV encephalitis (HSE) is the most prevalent sporadic viral encephalitis. Although safe and effective antiviral therapies and greatly improved noninvasive diagnostic procedures have significantly improved outcomes, mortality (~20%) and debilitating neurological sequelae in survivors remain unacceptably high. An encouraging new development is that the focus is now shifting away from the virus exclusively, to include consideration of the host immune response to infection in the pathology underlying development of HSE. In this article, the authors discuss results from recent studies in experimental mouse models, as well as clinical reports that demonstrate a role for exaggerated host inflammatory responses in the brain in the development of HSE that is motivating researchers and clinicians to consider new therapeutic approaches for treating HSE. The authors also discuss results from a few studies that have shown that immunomodulatory drugs can be highly protective against HSE, which supports a role for deleterious host inflammatory responses in HSE. The impressive outcomes of some immunomodulatory approaches in mouse models of HSE emphasize the urgent need for clinical trials to rigorously evaluate combination antiviral and immunomodulatory therapy in comparison with standard antiviral therapy for treatment of HSE, and support for such an initiative is gaining momentum.

12.
J Control Release ; 160(2): 194-9, 2012 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-22094102

RESUMO

Tumor necrosis factor-alpha (TNFα) is a classic proinflammatory cytokine implicated in the pathogenesis of several autoimmune and inflammatory diseases including viral encephalitis. Macrophages being major producers of TNFα are thus attractive targets for in vivo RNA interference (RNAi) mediated down regulation of TNFα. The application of RNAi technology to in vivo models however presents obstacles, including rapid degradation of RNA duplexes in plasma, insufficient delivery to the target cell population and toxicity associated with intravenous administration of synthetic RNAs and carrier compounds. We exploited the phagocytic ability of macrophages for delivery of Dicer-substrate small interfering RNAs (DsiRNAs) targeting TNFα (DsiTNFα) by intraperitoneal administration of lipid-DsiRNA complexes that were efficiently taken up by peritoneal macrophages and other phagocytic cells. We report that DsiTNFα-lipid complexes delivered intraperitoneally altered the disease outcome in an acute sepsis model. Down-regulation of TNFα in peritoneal CD11b+ monocytes reduced liver damage in C57BL/6 mice and significantly delayed acute mortality in mice treated with low dose LPS plus d-galactosamine (D-GalN).


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , RNA Helicases DEAD-box/metabolismo , Macrófagos/efeitos dos fármacos , RNA Interferente Pequeno/administração & dosagem , Ribonuclease III/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Antígeno CD11b/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/epidemiologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Modelos Animais de Doenças , Regulação para Baixo , Citometria de Fluxo , Galactosamina/farmacologia , Injeções Intraperitoneais , Lipopolissacarídeos/farmacologia , Lipossomos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/imunologia , RNA Interferente Pequeno/genética , Sepse/induzido quimicamente , Sepse/complicações , Sepse/imunologia , Transfecção , Fator de Necrose Tumoral alfa/genética
13.
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
14.
J Virol ; 82(14): 7078-88, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18480436

RESUMO

This study was undertaken to investigate possible immune mechanisms in fatal herpes simplex virus type 1 (HSV-1) encephalitis (HSE) after HSV-1 corneal inoculation. Susceptible 129S6 (129) but not resistant C57BL/6 (B6) mice developed intense focal inflammatory brain stem lesions of primarily F4/80(+) macrophages and Gr-1(+) neutrophils detectable by magnetic resonance imaging as early as day 6 postinfection (p.i.). Depletion of macrophages and neutrophils significantly enhanced the survival of infected 129 mice. Immunodeficient B6 (IL-7R(-/-) Kit(w41/w41)) mice lacking adaptive cells (B6-E mice) and transplanted with 129 bone marrow showed significantly accelerated fatal HSE compared to B6-E mice transplanted with B6 marrow or control nontransplanted B6-E mice. In contrast, there was no difference in ocular viral shedding in B6-E mice transplanted with 129 or B6 bone marrow. Acyclovir treatment of 129 mice beginning on day 4 p.i. (24 h after HSV-1 first reaches the brain stem) reduced nervous system viral titers to undetectable levels but did not alter brain stem inflammation or mortality. We conclude that fatal HSE in 129 mice results from widespread damage in the brain stem caused by destructive inflammatory responses initiated early in infection by massive infiltration of innate cells.


Assuntos
Tronco Encefálico/imunologia , Encefalite/imunologia , Encefalite/virologia , Herpes Simples/complicações , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Aciclovir/uso terapêutico , Animais , Antivirais/uso terapêutico , Transplante de Medula Óssea/imunologia , Tronco Encefálico/patologia , Tronco Encefálico/virologia , Encefalite/mortalidade , Olho/virologia , Citometria de Fluxo , Herpes Simples/virologia , Inflamação/imunologia , Inflamação/virologia , Procedimentos de Redução de Leucócitos , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Neutrófilos/imunologia , Receptores de Interleucina-7/deficiência , Fator de Células-Tronco/genética , Análise de Sobrevida , Subpopulações de Linfócitos T/imunologia , Eliminação de Partículas Virais
15.
Invest Ophthalmol Vis Sci ; 48(9): 4162-70, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17724202

RESUMO

PURPOSE: The host inflammatory response to ocular infection with herpes simplex virus (HSV) can be either protective, with disease-free survival, or it can promote diseases such as HSV corneal disease (or herpes stromal keratitis [HSK] in humans) and encephalitis (HSE), depending on mouse strain. The role of CXCR3 chemokine signaling in HSV-induced central nervous system (CNS) inflammation and corneal disease was evaluated, and responses in genetically susceptible and resistant strains of mice were contrasted. METHODS: Resistant C57BL/6J (B6) and susceptible 129S6 (129) mice were given monoclonal antibodies (mAbs) to neutralize the CXCR3 ligands monokine induced by interferon-gamma (MIG, CXCL9) and interferon inducible protein-10 (IP-10, CXCL10) during HSV infection. In addition, the development of HSV disease was monitored in CXCR3-null mutant mice derived from resistant (B6) and susceptible (BALB/c) strains. Inflammatory cells infiltrating the cornea and brain stem were isolated and stained for flow cytometric analysis. RESULTS: MIG and IP-10 were induced in nervous system tissue after HSV inoculation by the corneal route. HSV-infected 129 mice treated with MIG- or IP-10-neutralizing mAbs showed significantly enhanced survival compared with mice treated with control isotype antibody, whereas survival of the B6 mice was unaltered. Similarly, greater survival was observed for BALB.CXCR3(-/-) mice compared with control BALB/c mice. Reduced CNS inflammation was documented that extended to the cornea, such that HSV corneal disease severity was reduced in susceptible BALB.CXCR3(-/-). In contrast, although survival of B6 and B6.CXCR3(-/-) mice was indistinguishable, B6.CXCR3(-/-) mice developed more severe corneal and periocular skin disease. CONCLUSIONS: The effects of CXCR3 signaling in HSV infection are strongly dependent on mouse strain.


Assuntos
Encefalite por Herpes Simples/mortalidade , Ceratite Herpética/mortalidade , Receptores de Quimiocinas/fisiologia , Transdução de Sinais/fisiologia , Dermatopatias Virais/mortalidade , Animais , Tronco Encefálico/metabolismo , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/fisiologia , Córnea/virologia , Suscetibilidade a Doenças , Encefalite por Herpes Simples/genética , Encefalite por Herpes Simples/fisiopatologia , Citometria de Fluxo , Herpesvirus Humano 1/fisiologia , Imunidade Inata , Ceratite Herpética/genética , Ceratite Herpética/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores CXCR3 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dermatopatias Virais/genética , Dermatopatias Virais/fisiopatologia , Gânglio Trigeminal/metabolismo , Regulação para Cima
17.
J Virol ; 81(3): 1451-60, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17108044

RESUMO

Tumor necrosis factor (TNF) is a multifunctional cytokine that has a role in induction and regulation of host innate and adaptive immune responses. The importance of TNF antiviral mechanisms is reflected by the diverse strategies adopted by different viruses, particularly members of the herpesvirus family, to block TNF responses. TNF binds and signals through two receptors, Tnfrsf1a (TNF receptor 1 [TNFR1], or p55) and Tnfrsf1b (TNFR2, or p75). We report here that herpes simplex virus 1 (HSV-1) infection of TNF-/- mice on the resistant C57BL/6 genetic background results in significantly increased susceptibility (P < 0.0001, log rank test) to fatal HSV encephalitis (HSE) and prolonged persistence of elevated levels of virus in neural tissues. In contrast, although virus titers in neural tissues of p55-/- N13 mice were elevated to levels comparable to what was found for the TNF-/- mice, the p55-/- N13 mice were as resistant as control C57BL/6 mice (P > 0.05). The incidence of fatal HSE was significantly increased by in vivo neutralization of TNF using soluble TNFR1 (sTNFR1) or depletion of macrophages in C57BL/6 mice (P = 0.0038 and P = 0.0071, respectively). Strikingly, in vivo neutralization of TNF in HSV-1-infected p55-/- p75-/- mice by use of three independent approaches (treatment with soluble p55 receptor, anti-TNF monoclonal antibody, or in vivo small interfering RNA against TNF) resulted in significantly increased mortality rates (P = 0.005), comparable in magnitude to those for C57BL/6 mice treated with sTNFR1 (P = 0.0018). Overall, these results indicate that while TNF is required for resistance to fatal HSE, both p55 and p75 receptors are dispensable. Precisely how TNF mediates protection against HSV-1 mortality in p55-/- p75-/- mice remains to be determined.


Assuntos
Antivirais/farmacologia , Encefalite por Herpes Simples/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Transdução de Sinais/fisiologia , Simplexvirus/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Animais , Encefalite por Herpes Simples/virologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores do Fator de Necrose Tumoral/genética , Simplexvirus/genética , Simplexvirus/patogenicidade
18.
Nat Protoc ; 1(2): 508-17, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17406276

RESUMO

RNA interference is a powerful tool for target-specific knockdown of gene expression. The triggers for this process are duplex small interfering RNAs (siRNAs) of 21-25 nt with 2-bp 3' overhangs produced in cells by the RNase III family member Dicer. We have observed that short RNAs that are long enough to serve as Dicer substrates (D-siRNA) can often evoke more potent RNA interference than the corresponding 21-nt siRNAs; this is probably a consequence of the physical handoff of the Dicer-produced siRNAs to the RNA-induced silencing complex. Here we describe the design parameters for D-siRNAs and a protocol for in vitro and in vivo intraperitoneal delivery of D-siRNAs and siRNAs to macrophages. siRNA delivery and transfection and analysis of macrophages in vivo can be accomplished within 36 h.


Assuntos
Macrófagos/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ribonuclease III/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Camundongos , RNA Interferente Pequeno/química , Especificidade por Substrato , Fatores de Tempo , Transfecção
19.
Nat Biotechnol ; 22(3): 321-5, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14990954

RESUMO

Small interfering RNAs (siRNA) are potent reagents for directed post-transcriptional gene silencing and a major new genetic tool for investigating mammalian cells. When synthetic siRNAs are used for gene silencing, the costs can be substantial because of variations in siRNA efficacies. An alternative to chemically synthesized siRNAs are siRNAs produced by bacteriophage T7 RNA polymerase. We found that siRNAs synthesized from the T7 RNA polymerase system can trigger a potent induction of interferon alpha and beta in a variety of cell lines. Surprisingly, we also found very potent induction of interferon alpha and beta by short single-stranded RNAs (ssRNAs) transcribed with T3, T7 and Sp6 RNA polymerases. Analyses of the potential mediators of this response revealed that the initiating 5' triphosphate is required for interferon induction. We describe here an improved method for T7 siRNA synthesis that alleviates the interferon response while maintaining full efficacy of the siRNAs.


Assuntos
RNA Polimerases Dirigidas por DNA/metabolismo , Regulação da Expressão Gênica/fisiologia , Interferons/metabolismo , Rim/metabolismo , Interferência de RNA/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Artefatos , Bacteriófagos/enzimologia , Linhagem Celular , RNA Polimerases Dirigidas por DNA/genética , Engenharia Genética/métodos , Herpesvirus Humano 1/fisiologia , Humanos , Interferons/genética , Rim/crescimento & desenvolvimento , Rim/virologia , RNA/genética , RNA/metabolismo
20.
J Virol ; 77(21): 11661-73, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14557652

RESUMO

During studies to determine a role for tumor necrosis factor (TNF) in herpes simplex virus type 1 (HSV-1) infection using TNF receptor null mutant mice, we discovered a genetic locus, closely linked to the TNF p55 receptor (Tnfrsf1a) gene on mouse chromosome 6 (c6), that determines resistance or susceptibility to HSV-1. We named this locus the herpes resistance locus, Hrl, and showed that it also mediates resistance to HSV-2. Hrl has at least two alleles, Hrl(r), expressed by resistant strains like C57BL/6 (B6), and Hrl(s), expressed by susceptible strains like 129S6 (129) and BALB/c. Although Hrl is inherited as an autosomal dominant gene, resistance to HSV-1 is strongly sex biased such that female mice are significantly more resistant than male mice. Analysis of backcrosses between resistant B6 and susceptible 129 mice revealed that a second locus, tentatively named the sex modifier locus, Sml, functions to augment resistance of female mice. Besides determining resistance, Hrl is one of several genes involved in the control of HSV-1 replication in the eye and ganglion. Remarkably, Hrl also affects reactivation of HSV-1, possibly by interaction with some unknown gene(s). We showed that Hrl is distinct from Cmv1, the gene that determines resistance to murine cytomegalovirus, which is encoded in the major NK cell complex just distal of p55 on c6. Hrl has been mapped to a roughly 5-centimorgan interval on c6, and current efforts are focused on obtaining a high-resolution map for Hrl.


Assuntos
Mapeamento Cromossômico , Ligação Genética , Herpesvirus Humano 1/patogenicidade , Herpesvirus Humano 2/patogenicidade , Imunidade Inata/genética , Animais , Antígenos CD/genética , Cruzamentos Genéticos , Cistos Glanglionares/virologia , Herpes Genital/genética , Herpes Genital/mortalidade , Herpes Simples/genética , Herpes Simples/mortalidade , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/imunologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos BALB C/genética , Camundongos Endogâmicos C57BL/genética , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Fatores Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...