Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Stroke ; 46(10): 2926-34, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26306753

RESUMO

BACKGROUND AND PURPOSE: Both pathogenic and regulatory immune processes are involved in the middle cerebral artery occlusion (MCAO) model of experimental stroke, including interactions involving the programmed death 1 (PD-1) receptor and its 2 ligands, PD-L1 and PD-L2. Although PD-1 reduced stroke severity, PD-L1 and PD-L2 appeared to play pathogenic roles, suggesting the use of anti-PD-L monoclonal antibody therapy for MCAO. METHODS: Male C57BL/6 mice were treated with a single dose of anti-PD-L1 monoclonal antibody 4 hours after MCAO and evaluated for clinical, histological and immunologic changes after 96 hours of reperfusion. RESULTS: Blockade of the PD-L1 checkpoint using a single injection of 200 µg anti-PD-L1 monoclonal antibody given intravenously 4 hours after occlusion significantly reduced MCAO infarct volumes and improved neurological outcomes after 96 hours of reperfusion. Treatment partially reversed splenic atrophy and decreased central nervous system infiltrating immune cells concomitant with enhanced appearance of CD8(+) regulatory T cells in the lesioned central nervous system hemisphere. CONCLUSIONS: This study demonstrates for the first time the beneficial therapeutic effects of PD-L1 checkpoint blockade on MCAO, thus validating proposed mechanisms obtained in our previous studies using PD-1- and PD-L-deficient mice. These results provide strong support for the use of available humanized anti-PD-L1 antibodies for treatment of human stroke subjects.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Encéfalo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/imunologia , Animais , Antígeno B7-H1/imunologia , Encéfalo/imunologia , Encéfalo/patologia , Sistema Nervoso Central/imunologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença , Linfócitos T Reguladores/imunologia
2.
Metab Brain Dis ; 30(5): 1117-27, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25776868

RESUMO

Clinical improvement during pregnancy in multiple sclerosis (MS) patients suggests that sex hormones exert potent regulatory effects on autoimmune function. Our previous studies demonstrated that estrogen- (17ß-estradiol; E2) mediated protection against experimental autoimmune encephalomyelitis (EAE), a mouse model for MS, hinges on the B cells, leading to elevated numbers of IL-10 secreting CD1d(hi)CD5(+) B regulatory cells (Bregs) in wild type mice. Our data show that co-administration of E2 and IL-10(+) B cells ameliorates EAE disease severity and limits CNS infiltrating leukocytes in B cell deficient mice. Additionally, treatment with E2 and Bregs reduces demyelination and dramatically decreases the proportion of CD11b(+)CD45(hi) activated microglia/macrophages found in the CNS of immunized animals compared to vehicle, E2 or Breg cells alone. Furthermore, mice given E2 and Bregs exhibit increased numbers of peripheral programmed death-1 positive CD4(+)Foxp3(+) regulatory T cells (Tregs) and up-regulation of programmed death receptor-ligand-1 and CD80 expression on monocytes. Our study suggests IL-10 producing Bregs have powerful therapeutic potential as an agent against EAE when augmented with E2 treatment.


Assuntos
Linfócitos B/transplante , Sistema Nervoso Central , Encefalomielite Autoimune Experimental/terapia , Estradiol/administração & dosagem , Interleucina-10/administração & dosagem , Índice de Gravidade de Doença , Animais , Linfócitos B/imunologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Inflamação/imunologia , Inflamação/patologia , Inflamação/terapia , Interleucina-10/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Resultado do Tratamento
3.
Metab Brain Dis ; 30(4): 911-924, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25537181

RESUMO

Clinical stroke induces inflammatory processes leading to cerebral and splenic injury and profound peripheral immunosuppression. IL-10 expression is elevated during major CNS diseases and limits inflammation in the brain. Recent evidence demonstrated that transfer of IL-10(+) B-cells reduced infarct volume in male C57BL/6J (wild-type, WT) recipient mice when given 24 h prior to or 4 h after middle cerebral artery occlusion (MCAO). The purpose of this study was to determine if passively transferred IL-10(+) B-cells can exert therapeutic and immunoregulatory effects when injected 24 h after MCAO induction in B-cell-sufficient male WT mice. The results demonstrated that IL-10(+) B-cell treated mice had significantly reduced infarct volumes in the ipsilateral cortex and hemisphere and improved neurological deficits vs. Vehicle-treated control mice after 60 min occlusion and 96 h of reperfusion. The MCAO-protected B-cell recipient mice had less splenic atrophy and reduced numbers of activated, inflammatory T-cells, decreased infiltration of T-cells and a less inflammatory milieu in the ischemic hemispheres compared with Vehicle-treated control mice. These immunoregulatory changes occurred in concert with the predominant appearance of IL-10-secreting CD8(+)CD122(+) Treg cells in both the spleen and the MCAO-affected brain hemisphere. This study for the first time demonstrates a major neuroprotective role for IL-10(+) B-cells in treating MCAO in male WT mice at a time point well beyond the ~4 h tPA treatment window, leading to the generation of a dominant IL-10(+)CD8(+)CD122(+) Treg population associated with spleen preservation and reduced CNS inflammation.


Assuntos
Linfócitos B/metabolismo , Linfócitos B/transplante , Linfócitos T CD8-Positivos/fisiologia , Interleucina-10/metabolismo , Subunidade beta de Receptor de Interleucina-2/fisiologia , Acidente Vascular Cerebral/terapia , Transferência Adotiva/métodos , Animais , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Interleucina-10/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/patologia , Resultado do Tratamento
4.
Metab Brain Dis ; 30(6): 1515-29, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26246072

RESUMO

Inflammation plays a critical role in the pathogenesis of ischemic stroke. This process depends, in part, upon proinflammatory factors released by activated resident central nervous system (CNS) microglia (MG). Previous studies demonstrated that transfer of IL-10(+) B-cells reduced infarct volumes in male C57BL/6 J recipient mice when given 24 h prior to or therapeutically at 4 or 24 h after experimental stroke induced by 60 min middle cerebral artery occlusion (MCAO). The present study assesses possible sex differences in immunoregulation by IL-10(+) B-cells on primary male vs. female MG cultured from naïve and ischemic stroke-induced mice. Thus, MG cultures were treated with recombinant (r)IL-10, rIL-4 or IL-10(+) B-cells after lipopolysaccharide (LPS) activation and evaluated by flow cytometry for production of proinflammatory and anti-inflammatory factors. We found that IL-10(+) B-cells significantly reduced MG production of TNF-α, IL-1ß and CCL3 post-MCAO and increased their expression of the anti-inflammatory M2 marker, CD206, by cell-cell interactions. Moreover, MG from female vs. male mice had higher expression of IL-4 and IL-10 receptors and increased production of IL-4, especially after treatment with IL-10(+) B-cells. These findings indicate that IL-10-producing B-cells play a crucial role in regulating MG activation, proinflammatory cytokine release and M2 phenotype induction, post-MCAO, with heightened sensitivity of female MG to IL-4 and IL-10. This study, coupled with our previous demonstration of increased numbers of transferred IL-10(+) B-cells in the ischemic hemisphere, provide a mechanistic basis for local regulation by secreted IL-10 and IL-4 as well as direct B-cell/MG interactions that promote M2-MG.


Assuntos
Isquemia Encefálica/imunologia , Isquemia Encefálica/metabolismo , Microglia/imunologia , Microglia/metabolismo , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/metabolismo , Animais , Linfócitos B/imunologia , Comunicação Celular , Técnicas de Cocultura , Feminino , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Lectinas Tipo C/biossíntese , Lectinas Tipo C/genética , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/biossíntese , Lectinas de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Caracteres Sexuais
5.
Metab Brain Dis ; 29(1): 59-73, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24374817

RESUMO

Clinical stroke induces inflammatory processes leading to cerebral and splenic injury and profound peripheral immunosuppression. IL-10 expression is elevated during major CNS diseases and limits inflammation in the brain. Recent evidence demonstrated that absence of B-cells led to larger infarct volumes and CNS damage after middle cerebral artery occlusion (MCAO) that could be prevented by transfer of IL-10(+) B-cells. The purpose of this study was to determine if the beneficial immunoregulatory effects on MCAO of the IL-10(+) B-cell subpopulation also extends to B-cell-sufficient mice that would better represent stroke subjects. CNS inflammation and infarct volumes were evaluated in male C57BL/6J (WT) mice that received either RPMI or IL-10(+) B-cells and underwent 60 min of middle cerebral artery occlusion (MCAO) followed by 96 h of reperfusion. Transfer of IL-10(+) B-cells markedly reduced infarct volume in WT recipient mice when given 24 h prior to or 4 h after MCAO. B-cell protected (24 h pre-MCAO) mice had increased regulatory subpopulations in the periphery, reduced numbers of activated, inflammatory T-cells, decreased infiltration of T-cells and a less inflammatory milieu in the ischemic hemispheres of the IL-10(+) B-cell-treated group. Moreover, transfer of IL-10(+) B-cells 24 h before MCAO led to a significant preservation of regulatory immune subsets in the IL-10(+) B-cell protected group presumably indicating their role in immunomodulatory mechanisms, post-stroke. Our studies are the first to demonstrate a major immunoregulatory role for IL-10(+) regulatory B-cells in preventing and treating MCAO in WT mice and also implicating their potential role in attenuating complications due to post-stroke immunosuppression.


Assuntos
Transferência Adotiva , Subpopulações de Linfócitos B/transplante , Infarto da Artéria Cerebral Média/terapia , Interleucina-10/metabolismo , Animais , Antígenos CD19/análise , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Encéfalo/imunologia , Encéfalo/patologia , Quimiotaxia de Leucócito , Genes Reporter , Imunocompetência , Separação Imunomagnética , Imunomodulação , Infarto da Artéria Cerebral Média/patologia , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/imunologia , Baço/imunologia , Baço/patologia , Subpopulações de Linfócitos T/imunologia
6.
J Neuroinflammation ; 10: 111, 2013 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-24015822

RESUMO

BACKGROUND: Stroke severity is worsened by recruitment of inflammatory immune cells into the brain. This process depends in part on T cell activation, in which the B7 family of co-stimulatory molecules plays a pivotal role. Previous studies demonstrated more severe infarcts in mice lacking programmed death-1 (PD-1), a member of the B7 family, thus implicating PD-1 as a key factor in limiting stroke severity. The purpose of this study was to determine if this protective effect of PD-1 involves either of its ligands, PD-L1 or PD-L2. METHODS: Central nervous system (CNS) inflammation and infarct volume were evaluated in male PD-L1 and PD-L2 knockout (-/-) mice undergoing 60 minutes of middle cerebral artery occlusion (MCAO) followed by 96 hours of reperfusion and compared to wild-type (WT) C57BL/6J mice. RESULTS: PD-L1-/- and PD-L2-/- mice had smaller total infarct volumes compared to WT mice. The PD-L1-/- and to a lesser extent PD-L2-/- mice had reduced levels of proinflammatory activated microglia and/or infiltrating monocytes and CD4+ T cells in the ischemic hemispheres. There was a reduction in ischemia-related splenic atrophy accompanied by lower activation status of splenic T cells and monocytes in the absence of PD-L1, suggesting a pathogenic rather than a regulatory role for both PD-1 ligands (PD-Ls). Suppressor T cells (IL-10-producing CD8+CD122+ T cells) trafficked to the brain in PD-L1-/- mice and there was decreased expression of CD80 on splenic antigen-presenting cells (APCs) as compared to the WT and PD-L2-/- mice. CONCLUSIONS: Our novel observations are the first to implicate PD-L1 involvement in worsening outcome of experimental stroke. The presence of suppressor T cells in the right MCAO-inflicted hemisphere in mice lacking PD-L1 implicates these cells as possible key contributors for controlling adverse effects of ischemia. Increased expression of CD80 on APCs in WT and PD-L2-/- mice suggests an overriding interaction leading to T cell activation. Conversely, low CD80 expression by APCs, along with increased PD-1 and PD-L2 expression in PD-L1-/- mice suggests alternative T cell signaling pathways, leading to a suppressor phenotype. These results suggest that agents (for example antibodies) that can target and neutralize PD-L1/2 may have therapeutic potential for treatment of human stroke.


Assuntos
Antígeno B7-H1/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia , Animais , Antígeno B7-H1/imunologia , Modelos Animais de Doenças , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
7.
Metab Brain Dis ; 28(3): 375-86, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23640015

RESUMO

Clinical stroke induces inflammatory processes leading to cerebral injury. IL-10 expression is elevated during major CNS diseases and limits inflammation in the brain. Recent evidence demonstrated that absence of B-cells led to larger infarct volumes and increased numbers of activated T-cells, monocytes and microglial cells in the brain, thus implicating a regulatory role of B-cell subpopulations in limiting CNS damage from stroke. The aim of this study was to determine whether the IL-10-producing regulatory B-cell subset can limit CNS inflammation and reduce infarct volume following ischemic stroke in B-cell deficient (µMT(-/-)) mice. Five million IL-10-producing B-cells were obtained from IL-10-GFP reporter mice and transferred i.v. to µMT(-/-)mice. After 24 h following this transfer, recipients were subjected to 60 min of middle cerebral artery occlusion (MCAO) followed by 48 h of reperfusion. Compared to vehicle-treated controls, the IL-10(+) B-cell-replenished µMT(-/-)mice had reduced infarct volume and fewer infiltrating activated T-cells and monocytes in the affected brain hemisphere. These effects in CNS were accompanied by significant increases in regulatory T-cells and expression of the co-inhibitory receptor, PD-1, with a significant reduction in the proinflammatory milieu in the periphery. These novel observations provide the first proof of both immunoregulatory and protective functions of IL-10-secreting B-cells in MCAO that potentially could impart significant benefit for stroke patients in the clinic.


Assuntos
Linfócitos B/metabolismo , Infarto Cerebral/metabolismo , Interleucina-10/sangue , Acidente Vascular Cerebral/sangue , Transferência Adotiva , Animais , Isquemia Encefálica/prevenção & controle , Éxons/genética , Citometria de Fluxo , Proteínas de Fluorescência Verde , Cadeias mu de Imunoglobulina/genética , Infarto da Artéria Cerebral Média/patologia , Inflamação/patologia , Camundongos , Camundongos Knockout , Monócitos/fisiologia , Baço/patologia
8.
Immunology ; 137(4): 282-93, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23039230

RESUMO

It is now well accepted that sex hormones have immunoregulatory activity and may prevent exacerbations in multiple sclerosis during pregnancy. Our previous studies demonstrated that oestrogen (17ß-oestradiol; E(2) ) protection against experimental autoimmune encephalomyelitis (EAE) is mediated mainly through oestrogen receptor-α (ERα) and the membrane receptor G-protein-coupled receptor 30 (GPR30) and is abrogated in the absence of B cells and the co-inhibitory receptor, Programmed Death-1 (PD-1). To critically evaluate the cell source of the E2 and PD-1 co-inhibitory pathways in EAE regulation, we assessed the requirement for ERs on transferred B cells and downstream effects on expression of PD-1/PD-ligand on CD4+ Foxp3+ regulatory T (Treg) cells in B-cell-replenished, E2-treated B-cell-deficient (µMT-/-) mice with EAE. The results clearly demonstrated involvement of ERα and GPR30 on transferred B cells that mediated the protective E2 treatment effect on EAE and further showed an E2-mediated B-cell-dependent up-regulation of PD-1 on CD4+ Foxp3+ Treg cells. These findings identify regulatory B-cell populations as key players in potentiating Treg-cell activity during E2-mediated protection against EAE.


Assuntos
Linfócitos B/metabolismo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Estradiol/uso terapêutico , Receptor de Morte Celular Programada 1/metabolismo , Receptores de Estradiol/metabolismo , Linfócitos T Reguladores/metabolismo , Regulação para Cima , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Encefalomielite Autoimune Experimental/imunologia , Estradiol/farmacologia , Feminino , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Estradiol/análise , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Regulação para Cima/efeitos dos fármacos
9.
Eur J Immunol ; 41(4): 1165-75, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21413005

RESUMO

Increased remissions in multiple sclerosis (MS) during pregnancy suggest that elevated levels of sex steroids exert immunoregulatory activity. Estrogen (E2=17ß-estradiol) protects against experimental autoimmune encephalomyelitis (EAE), but the cellular basis for E2-induced protection remains unclear. Studies demonstrate that depletion of B cells prior to induction of EAE exacerbates disease severity, implicating regulatory B cells. We thus evaluated pathogenic and E2-induced protective mechanisms in B-cell-deficient (µMT(-/-)) mice. EAE-protective effects of E2 were abrogated in µMT(-/-) mice, with no reduction in disease severity, cellular infiltration or pro-inflammatory factors in the central nervous system compared to untreated controls. E2 treatment of WT mice selectively upregulated expression of PD-L1 on B cells and increased the percentage of IL-10-producing CD1d(high) CD5(+) regulatory B cells. Upregulation of PD-L1 was critical for E2-mediated protection since E2 did not inhibit EAE in PD-L1(-/-) mice. Direct treatment of B cells with E2 significantly reduced proliferation of MOG(35-55)-specific T cells that required estrogen receptor-α (ERα). These results demonstrate, for the first time, a requirement for B cells in E2-mediated protection against EAE involving direct E2 effects on regulatory B cells mediated through ERα and the PD-1/PD-L1 negative co-stimulatory pathway. E2-primed B cells may represent an important regulatory mechanism in MS and have strong implications for women receiving current MS therapies that cause B-cell depletion.


Assuntos
Linfócitos B/imunologia , Encefalomielite Autoimune Experimental/imunologia , Estrogênios/imunologia , Animais , Antígenos CD1d/imunologia , Antígeno B7-1/imunologia , Antígeno B7-H1 , Antígenos CD5/imunologia , Células Cultivadas , Estradiol/imunologia , Feminino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/deficiência , Peptídeos/imunologia , Baço/imunologia
10.
Metab Brain Dis ; 27(4): 487-93, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22618587

RESUMO

Recent evidence emphasizes B-cells as a major regulatory cell type that plays an important role in limiting the pathogenic effects of ischemic stroke. The aim of the current study was to extend this initial observation to specifically examine the infiltration of regulatory B-cells and to determine if the effect of B-cells to limit the inflammatory response to cerebral ischemia is mediated by their action centrally or peripherally. Our data demonstrate the increased presence of a regulatory B-cell subset in the affected hemisphere of wild-type mice after middle cerebral artery occlusion (MCAO). We further explored the use of a novel method of stereotaxic cell delivery to bypass the blood brain barrier (BBB) and introduce CD19(+) B-cells directly into the striatum as compared to peripheral administration of B-cells. Infarct volumes after 60 minutes of MCAO and 48 hours of reperfusion were determined in B-cell deficient µMT( -/- ) mice with and without replacement of either B-cells or medium. Infarct size was significantly decreased in cerebral cortex after intrastriatal transfer of 100,000 B-cells to µMT(-/-) mice vs. controls, with a comparable effect on infarct size as obtained by 50 million B-cells transferred intraperitoneally. These findings support the hypothesis that B-cells play a protective role against ischemic brain injury, and suggest that B-cells may serve as a novel therapeutic agent for modulating the immune response in central nervous system inflammation after stroke.


Assuntos
Linfócitos B Reguladores/fisiologia , Transplante de Células , Infarto Cerebral/patologia , Infarto Cerebral/terapia , Acidente Vascular Cerebral/patologia , Acidente Vascular Cerebral/terapia , Animais , Antígenos CD19 , Separação Celular , Córtex Cerebelar/patologia , Imunofluorescência , Imuno-Histoquímica , Fluxometria por Laser-Doppler , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Artéria Cerebral Média/fisiologia , Neostriado/fisiologia , Técnicas Estereotáxicas
11.
J Immunol ; 183(4): 2622-31, 2009 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-19625649

RESUMO

The purpose of the present study was to determine the impact of NK cells on the development of protective adaptive immunity in response to nasal-pulmonary immunization against mycoplasma. Depletion of NK cells before nasal-pulmonary immunization enhanced resistance to mycoplasma respiratory infection. The effect of NK cells on the generation of protective immunity in lungs was dependent on lymphoid cells, as immunization of either SCID mice or immunocompetent mice depleted of CD4(+) T cells did not demonstrate any increased resistance in the presence or absence of NK cells. The presence of NK cells at the time of nasal-pulmonary immunization modulated mycoplasma-specific cytokine responses in lungs and lower respiratory nodes. In particular, NK cells skewed the mycoplasma-specific T cell cytokine responses in the draining lymph nodes to higher IL-4, IL-13, and IL-17 while lowering IFN-gamma responses. Adoptive transfer of total lung lymphocytes isolated from immunized mice into naive mice led to a significant reduction in the mycoplasma numbers in lungs, and the resistance was greater if cells were obtained from immunized mice that were depleted of NK cells. Similar results were obtained if purified B cells, T cells, or CD4(+) T cells were used. Interestingly, this is the first time that a favorable role of functional CD4(+) T cells in mediating protection in mycoplasma respiratory disease was demonstrated. Thus, NK cells can influence the responses of multiple lymphocyte populations capable of mediating resistance to mycoplasma infection.


Assuntos
Células Matadoras Naturais/imunologia , Infecções por Mycoplasma/prevenção & controle , Mycoplasma pulmonis/imunologia , Infecções Respiratórias/prevenção & controle , Administração por Inalação , Animais , Antígenos de Bactérias/administração & dosagem , Antígenos de Bactérias/imunologia , Células Cultivadas , Feminino , Gangliosídeo G(M1)/biossíntese , Imunidade Inata , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/microbiologia , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Camundongos Transgênicos , Infecções por Mycoplasma/imunologia , Infecções por Mycoplasma/microbiologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/microbiologia , Vacinação
12.
J Infect Dis ; 202(1): 39-51, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20504237

RESUMO

For vaccine development, it is critical to understand the regulatory mechanisms determining resistance and immunopathology against mycoplasma respiratory diseases. The present study evaluated the contribution of the polarizing cytokines interferon gamma (IFN-gamma) and interleukin 4 (IL-4) in the regulation of mycoplasma-specific immunity. The absence of a single cytokine (either IFN-gamma or IL-4) uniquely altered the expression of multiple chemokines and cytokines in the lungs of uninfected mice and influenced responses to mycoplasma infection. Most importantly, prior nasal-pulmonary immunization of IFN-gamma(-/-) mice led to exacerbated mycoplasma disease, whereas immunized IL-4(-/-) mice were dramatically more resistant than wild-type mice. Helper T cell type 2 responses in IFN-gamma(-/-) mice corresponded to immunopathologic reactions that developed after mycoplasma infection or immunization. Thus, adaptive immunity clearly can independently promote either protection or immunopathology against mycoplasma infection, and optimal vaccination appears to be dependent on promoting protective IFN-gamma-dependent networks (perhaps helper T cell type 1 responses) while minimizing the effect of IL-4-mediated responses, which dampen the generation of protective immunity.


Assuntos
Interferon gama/metabolismo , Interleucina-4/metabolismo , Infecções por Mycoplasma/imunologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/microbiologia , Imunidade Adaptativa , Animais , Feminino , Regulação da Expressão Gênica/imunologia , Interferon gama/genética , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-4/genética , Interleucina-5/genética , Interleucina-5/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Eur J Immunol ; 39(2): 426-38, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19180464

RESUMO

Immune responses to pathogens occur within the context of current and previous infections. Cross protection refers to the phenomena where infection with a particular pathogen provides enhanced resistance to a subsequent unrelated pathogen in an antigen-independent manner. Proposed mechanisms of antigen-independent cross protection have involved the secretion of IFN-gamma, which activates macrophages, thus providing enhanced innate immunity against the secondary viral or bacterial pathogen. Here we provide evidence that a primary infection with the chronic respiratory pathogen, Mycoplasma pulmonis, provides a novel form of cross protection against a secondary infection with Listeria monocytogenes that is not mediated by IFN-gamma, but instead relies upon IL-17 and mobilization of neutrophils. Mice infected with M. pulmonis have enhanced clearance of L. monocytogenes from the spleen and liver, which is associated with increased numbers of Gr-1(+)CD11b(+) cells and higher levels of IL-17. This enhanced clearance of L. monocytogenes was absent in mice depleted of Gr-1(+) cells or in mice deficient in the IL-17 receptor. Additionally, both the IL-17 receptor and neutrophils were essential for optimal clearance of M. pulmonis. Thus, a natural component of the immune response directed against M. pulmonis was able to enhance clearance of L. monocytogenes.


Assuntos
Interleucina-17/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Infecções por Mycoplasma/imunologia , Mycoplasma pulmonis/imunologia , Animais , Feminino , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Listeriose/microbiologia , Fígado/imunologia , Fígado/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Mycoplasma/microbiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Receptores de Interleucina-17/metabolismo , Baço/imunologia , Baço/microbiologia
14.
J Neuroimmunol ; 293: 45-53, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27049561

RESUMO

Sex hormones promote immunoregulatory effects on multiple sclerosis. The current study evaluated estrogen effects on regulatory B cells and resident CNS microglia during experimental autoimmune encephalomyelitis (EAE). Herein, we demonstrate an estrogen-dependent induction of multiple regulatory B cell markers indicative of IL-10 dependent as well as IFN-γ dependent pathways. Moreover, although estrogen pretreatment of EAE mice inhibited the infiltration of pro-inflammatory cells into the CNS, it enhanced the frequency of regulatory B cells and M2 microglia. Our study suggests that estrogen has a broad effect on the development of regulatory B cells during EAE, which in turn could promote neuroprotection.


Assuntos
Linfócitos B Reguladores/efeitos dos fármacos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Estradiol/uso terapêutico , Microglia/efeitos dos fármacos , Animais , Linfócitos B Reguladores/metabolismo , Encéfalo/patologia , Proliferação de Células/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Adjuvante de Freund/toxicidade , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Glicoproteína Mielina-Oligodendrócito/imunologia , Glicoproteína Mielina-Oligodendrócito/toxicidade , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/toxicidade , Toxina Pertussis/toxicidade , RNA Mensageiro/metabolismo , Medula Espinal/citologia , Baço/citologia , Estatísticas não Paramétricas , Fatores de Tempo
15.
J Neuroimmunol ; 285: 129-36, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26198929

RESUMO

Women with multiple sclerosis (MS) often experience clinical improvement during pregnancy, indicating that sex hormones might have therapeutic effects in MS. Our previous studies have demonstrated that B cells and PD-L1 are crucial for E2 (17ß-estradiol)-mediated protection against experimental autoimmune encephalomyelitis (EAE). We here demonstrate that the transfer of IL-10(+) B cells into E2-treated PD-L1(-/-) mice after EAE induction could partially restore E2-mediated protection and decrease the frequency of pro-inflammatory cells in the CNS compared to E2/saline treated PD-L1(-/-) mice. Hence, co-administration of IL-10(+) B cells and E2 might have a powerful therapeutic potential for treatment of EAE.


Assuntos
Linfócitos B/metabolismo , Antígeno B7-H1/deficiência , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/prevenção & controle , Estradiol/uso terapêutico , Interleucina-10/biossíntese , Animais , Linfócitos B/imunologia , Linfócitos B/transplante , Antígeno B7-H1/imunologia , Encefalomielite Autoimune Experimental/imunologia , Estradiol/farmacologia , Feminino , Interleucina-10/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico
16.
Transl Stroke Res ; 6(1): 60-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25270354

RESUMO

Recombinant T cell Receptor Ligand 1000 (RTL1000), a partial human major histocompatibility complex (MHC) molecule coupled to a human myelin peptide, reduces infarct size after experimental stroke in HLA-DRB1*1502 transgenic (DR2-Tg) mice. In this study, we characterized the therapeutic time window of opportunity for RTL1000; we explored the efficacy of a single dose of RTL1000 administration and determined if RTL1000 affords long-term neurobehavioral functional improvement after ischemic stroke. Male DR2-Tg mice underwent 60 min of intraluminal reversible middle cerebral artery occlusion (MCAO). RTL1000 or vehicle was injected 4, 6, or 8 h after MCAO, followed by three daily injections. In the single-dose study, one-time injection of RTL1000 was applied 4 h after MCAO. Cortical, striatal, and hemispheric infarct sizes were measured 24 or 96 h after stroke. Behavioral testing, including neuroscore evaluation, open field, paw preference, and novel object recognition, was performed up to 28 days after stroke. Our data showed that RTL1000 significantly reduced the infarct size 96 h after MCAO when the first injection was given at 4 and 6 h, but not 8 h, after the onset of stroke. A single dose of 400 or 100 µg RTL1000 also significantly reduced the infarct size 24 h after MCAO. Behavioral testing showed that RTL1000 treatment used 4 h after MCAO improved long-term cognitive outcome 28 days after stroke. Taken together, RTL1000 protects against acute injury if applied within a 6-h time window and improves long-term functional recovery after experimental stroke in DR2-Tg mice.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Isquemia Encefálica/patologia , Infarto da Artéria Cerebral Média , Ligantes , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Proteínas Recombinantes/administração & dosagem , Acidente Vascular Cerebral/patologia , Resultado do Tratamento
17.
Transl Stroke Res ; 5(5): 612-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24953050

RESUMO

RTL1000 is a partial human MHC molecule coupled to a human myelin peptide. We previously demonstrated that RTL1000 was protective against experimental ischemic stroke in HLA-DR2 transgenic (DR2-Tg) mice. Since thrombolysis with recombinant tissue plasminogen activator (t-PA) is a standard therapy for stroke, we determined if RTL1000 efficacy is altered when combined with t-PA in experimental stroke. Male DR2-Tg mice underwent 60 min of intraluminal middle cerebral artery occlusion (MCAO). t-PA or vehicle was infused intravenously followed by either a single or four daily subcutaneous injections of RTL1000 or vehicle. Infarct size was measured by 2, 3, 5-triphenyltetrazolium chloride staining at 24 or 96 h of reperfusion. Our data showed that t-PA alone reduced infarct size when measured at 24 h but not at 96 h after MCAO. RTL1000 alone reduced infarct size both at 24 and 96 h after MCAO. Combining RTL1000 with t-PA did not alter its ability to reduce infarct size at either 24 or 96 h after MCAO and provides additional protection in t-PA treated mice at 24 h after ischemic stroke. Taken together, RTL1000 treatment alone improves outcome and provides additional protection in t-PA-treated mice in experimental ischemic stroke.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Acidente Vascular Cerebral/tratamento farmacológico , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Modelos Animais de Doenças , Quimioterapia Combinada , Humanos , Infarto da Artéria Cerebral Média , Masculino , Camundongos , Resultado do Tratamento
18.
Front Cell Neurosci ; 8: 228, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25157219

RESUMO

Stroke outcome is worsened by the infiltration of inflammatory immune cells into ischemic brains. Our recent study demonstrated that PD-L1- and to a lesser extent PD-L2-deficient mice had smaller brain infarcts and fewer brain-infiltrating cells vs. wild-type (WT) mice, suggesting a pathogenic role for PD-ligands in experimental stroke. We sought to ascertain PD-L1 and PD-L2-expressing cell types that affect T-cell activation, post-stroke in the context of other known co-stimulatory molecules. Thus, cells from male WT and PD-L-deficient mice undergoing 60 min of middle cerebral artery occlusion (MCAO) followed by 96 h of reperfusion were treated with neutralizing antibodies to study co-stimulatory and co-inhibitory interactions between CD80, cytotoxic T-lymphocyte antigen-4 (CTLA-4), PD-1, and PD-Ls that regulate CD8(+) and CD4(+) T-cell activation. We found that antibody neutralization of PD-1 and CTLA-4 signaling post-MCAO resulted in higher proliferation in WT CD8(+) and CD4(+) T-cells, confirming an inhibitory role of PD-1 and CTLA-4 on T-cell activation. Also, CD80/CD28 interactions played a prominent regulatory role for the CD8(+) T-cells and the PD-1/PD-L2 interactions were dominant in controlling the CD4(+) T-cell responses in WT mice after stroke. A suppressive phenotype in PD-L1-deficient mice was attributed to CD80/CTLA-4 and PD-1/PD-L2 interactions. PD-L2 was crucial in modulating CD4(+) T-cell responses, whereas PD-L1 regulated both CD8(+) and CD4(+) T-cells. To establish the contribution of PD-L1 and PD-L2 on regulatory B-cells (Bregs), infarct volumes were evaluated in male PD-L1- and PD-L2-deficient mice receiving IL-10(+) B-cells 4h post-MCAO. PD-L2- but not PD-L1-deficient recipients of IL-10(+) B-cells had markedly reduced infarct volumes, indicating a regulatory role of PD-L2 on Bregs. These results imply that PD-L1 and PD-L2 differentially control induction of T- and Breg-cell responses after MCAO, thus suggesting that selective targeting of PD-L1 and PD-L2 might represent a valuable therapeutic strategy in stroke.

19.
J Clin Cell Immunol ; 4(3): 143, 2013 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24009988

RESUMO

Increased remissions in multiple sclerosis (MS) during late pregnancy may result from high levels of sex steroids such as estrogen and estriol. Estrogen (E2=17ß-estradiol) protects against experimental autoimmune encephalomyelitis (EAE), but the cellular basis for E2-induced protection remains unclear. Treatment with relatively low doses of E2 can protect against clinical and histological signs of MOG-35-55 induced EAE through mechanisms involving the PD-1 coinhibitory pathway and B-cells. The current study evaluated the contribution of PD-1 ligands, PD-L1 and PD-L2, on B-cells in E2-mediated protection against EAE in WT, PD-L1-/- and PD-L2-/- mice. Unlike PD-L2-/- mice that were fully protected against EAE after E2 treatment, E2-implanted PD-L1-/- mice were fully susceptible to EAE, with increased numbers of proliferating Th1/Th17 cells in the periphery and severe cellular infiltration and demyelination in the CNS. Moreover, transfer of B-cells from MOG-immunized PD-L1-/- or PD-L2-/- donors into E2-preconditioned B-cell deficient µMT-/- recipient mice revealed significantly reduced E2-mediated protection against EAE in recipients of PD-L1-/- B-cells, but near-complete protection in recipients of PD-L2-/- B-cells. We conclude that PD-1 interaction with PD-L1 but not PD-L2 on B-cells is crucial for E2-mediated protection in EAE and that strategies that enhance PD-1/PD-L1 interactions might potentiate E2 treatment effects in MS.

20.
PLoS One ; 8(2): e55984, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23390557

RESUMO

Mycoplasmas cause chronic respiratory diseases in animals and humans, and to date, development of vaccines have been problematic. Using a murine model of mycoplasma pneumonia, lymphocyte responses, specifically T cells, were shown to confer protection as well as promote immunopathology in mycoplasma disease. Because T cells play such a critical role, it is important to define the role of antigen presenting cells (APC) as these cells may influence either exacerbation of mycoplasma disease pathogenesis or enhancement of protective immunity. The roles of APC, such as dendritic cells and/or macrophages, and their ability to modulate adaptive immunity in mycoplasma disease are currently unknown. Therefore, the purpose of this study was to identify individual pulmonary APC populations that may contribute to the activation of T cell responses during mycoplasma disease pathogenesis. The present study indeed demonstrates increasing numbers of CD11c(-) F4/80(+) cells, which contain macrophages, and more mature/activated CD11c(+) F4/80(-) cells, containing DC, in the lungs after infection. CD11c(-) F4/80(+) macrophage-enriched cells and CD11c(+) F4/80(-) dendritic cell-enriched populations showed different patterns of cytokine mRNA expression, supporting the idea that these cells have different impacts on immunity in response to infection. In fact, DC containing CD11c(+) F4/80(-) cell populations from the lungs of infected mice were most capable of stimulating mycoplasma-specific CD4(+) Th cell responses in vitro. In vivo, these CD11c(+)F4/80(-) cells were co-localized with CD4(+) Th cells in inflammatory infiltrates in the lungs of mycoplasma-infected mice. Thus, CD11c(+)F4/80(-) dendritic cells appear to be the major APC population responsible for pulmonary T cell stimulation in mycoplasma-infected mice, and these dendritic cells likely contribute to responses impacting disease pathogenesis.


Assuntos
Células Dendríticas/imunologia , Pulmão/imunologia , Infecções por Mycoplasma/imunologia , Pneumonia por Mycoplasma/imunologia , RNA Mensageiro/imunologia , Linfócitos T/imunologia , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Antígeno CD11c/genética , Antígeno CD11c/imunologia , Citocinas/genética , Citocinas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/patologia , Feminino , Expressão Gênica , Humanos , Inflamação , Pulmão/microbiologia , Pulmão/patologia , Ativação Linfocitária , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/patologia , Camundongos , Mycoplasma/fisiologia , Infecções por Mycoplasma/microbiologia , Infecções por Mycoplasma/patologia , Pneumonia por Mycoplasma/microbiologia , Pneumonia por Mycoplasma/patologia , RNA Mensageiro/genética , Linfócitos T/microbiologia , Linfócitos T/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA