Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci Res ; 95(1-2): 437-446, 2017 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-27870460

RESUMO

Ischemic stroke is a leading cause of death and disability in the United States. It is known that males and females respond differently to stroke. Depending on age, the incidence, prevalence, mortality rate, and disability outcome of stroke differ between the sexes. Females generally have strokes at older ages than males and, therefore, have a worse stroke outcome. There are also major differences in how the sexes respond to stroke at the cellular level. Immune response is a critical factor in determining the progress of neurodegeneration after stroke and is fundamentally different for males and females. Additionally, females respond to stroke therapies differently from males, yet they are often left out of the basic research that is focused on developing those therapies. With a resounding failure to translate stroke therapies from the bench to the bedside, it is clearer than ever that inclusion of both sexes in stroke studies is essential for future clinical success. This Mini-Review examines sex differences in the immune response to experimental stroke and its implications for therapy development. © 2016 Wiley Periodicals, Inc.


Assuntos
Sistema Imunitário , Caracteres Sexuais , Acidente Vascular Cerebral/imunologia , Pesquisa Translacional Biomédica , Envelhecimento , Animais , Modelos Animais de Doenças , Progressão da Doença , Hormônios Esteroides Gonadais/metabolismo , Humanos , Sistema Imunitário/patologia , Sistema Imunitário/fisiopatologia , Inflamação/etiologia , Acidente Vascular Cerebral/metabolismo
2.
Metab Brain Dis ; 31(3): 683-92, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26868919

RESUMO

Peroxisome proliferator-activated receptor alpha (PPARα) is a nuclear receptor transcription factor that plays a role in immune regulation. Because of its expression in cerebral tissue and immune cells, PPARα has been examined as an important regulator in immune-based neurological diseases. Many studies have indicated that pre-treatment of animals with PPARα agonists induces protection against stroke. However, our previous reports indicate that protection is only in males, not females, and can be attributed to different PPARα expression between the sexes. In the current study, we examine how loss of PPARα affects male and female mice in experimental stroke. Male and female PPARα knockout mice were subject to middle cerebral artery occlusion (MCAO) or sham surgery, and the ischemic (local) or spleen specific (peripheral) immune response was examined 96 h after reperfusion. We found that loss of PPARα perpetuated sex differences in stroke, and this was driven by the peripheral, not local, immune response. Specifically we observed an increase in peripheral pro-inflammatory and adhesion molecule gene expression in PPARα KO males after MCAO compared to females. Our data supports previous evidence that PPARα plays an important role in sex differences in the immune response to disease, including stroke.


Assuntos
Infarto da Artéria Cerebral Média/metabolismo , Leucócitos/metabolismo , PPAR alfa/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , PPAR alfa/genética , Caracteres Sexuais , Baço/metabolismo
3.
Metab Brain Dis ; 31(3): 539-47, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26581674

RESUMO

Males and females respond differently to stroke. Moreover, females often experience worse long-term stroke outcomes. Fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARα) agonist has been shown to improve stroke outcome and resolve neuroinflammation in male mice. The present study compares the effect of pretreatment with fenofibrate versus vehicle control in male and female mice during experimental stroke. Mice were treated with low-dose fenofibrate 30 min before and once a day for three additional days after stroke onset. We observed a reduction in infarct volume in male mice 96 h post-stroke with low-dose fenofibrate pretreatment that was due to increase of an M2 macrophage phenotype in the brain and an increase in regulatory cells in the periphery. These outcomes were not replicated in females, likely due to the lower PPARα expression in cells and tissues in females vs males. We conclude that PPARα agonist treatment prior to stroke is neuroprotective in males but not females. These findings indicate PPARα as a probable mechanism of sex difference in stroke outcome and support the need for representation of females in stroke therapy research.


Assuntos
Fenofibrato/uso terapêutico , Hipolipemiantes/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , PPAR alfa/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Hipolipemiantes/farmacologia , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Camundongos , PPAR alfa/agonistas , Fatores Sexuais , Acidente Vascular Cerebral/metabolismo
4.
Transl Stroke Res ; 7(1): 70-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26627498

RESUMO

Inflammation and thrombosis are tightly linked, with inflammation contributing to thromboembolism and to stroke outcome. Thromboembolism is a frequent cause of ischemic stroke; yet, the most used occlusion mouse models of experimental stroke do not effectively replicate thromboembolism. Our group recently described a novel thromboembolic mouse model of stroke that successfully occludes the middle cerebral artery with high reproducibility. In the current study, we characterize the peripheral and local immune outcomes as well as the ischemic response to immune therapy in a clinically relevant mouse model of thromboembolic stroke. Brain and spleen tissues were harvested 24 h after thromboembolic stroke and cells immunophenotyped by flow cytometry. We observed a significant increase in neutrophils and early activated T cells in the spleen and an increase in neutrophils and activated monocytes/microglia in the ischemic cortex after thromboembolic stroke. Moreover, as was shown previously for transient MCAO models, treatment of thromboembolic stroke with partial MHC constructs significantly reduced ischemic damage indicating an equivalent effect of this immune-based therapy in the thromboembolic model that better mimics the pathophysiology of human stroke.


Assuntos
Isquemia Encefálica , Imunoterapia/métodos , Proteínas Recombinantes de Fusão/farmacologia , Acidente Vascular Cerebral , Tromboembolia/complicações , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/etiologia , Isquemia Encefálica/imunologia , Modelos Animais de Doenças , Inflamação/tratamento farmacológico , Complexo Principal de Histocompatibilidade , Camundongos , Proteínas Recombinantes de Fusão/administração & dosagem , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/etiologia , Acidente Vascular Cerebral/imunologia
5.
J Syst Integr Neurosci ; 1(1): 20-28, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26634148

RESUMO

The peripheral immune response contributes to neurologic impairment after stroke and the extent of initial damage is greater in males than females. We have previously shown that spleen cells directly contribute to ischemic damage in males, as splenectomy prior to experimental stroke eliminates the sex differences in infarct volume. This study aims to determine which specific subset of immune cells exert pathogenic effects when injected 24 hours before MCAO induction into splenectomized male and female WT mice. The results demonstrate that CD4/CD8/CD11b treated mice had no significant effect on infarct volumes vs. vehicle-treated control mice after MCAO. However, there were significant alterations to the resident peripheral immune composition. These results suggest that there are regulatory factors resulting from splenectomy or other possible influences that inhibit peripheral immune cell contribution to neuroinflammation and thus contributing to differential effects of the spleen on stroke outcome in males and female mice.

6.
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
7.
J Neuroimmunol ; 278: 289-98, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25434281

RESUMO

The peripheral immune response contributes to neurodegeneration after stroke yet little is known about how this process differs between males and females. The current study demonstrates that splenectomy prior to experimental stroke eliminates sex differences in infarct volume and activated brain monocytes/microglia. In the periphery of both sexes, activated T cells correlate directly with stroke outcome while monocytes are reduced by splenectomy only in males. This study provides new information about the sex specific mechanisms of the peripheral immune response in neurodegeneration after stroke and demonstrates the need for representation of both sexes in basic and clinical stroke research.


Assuntos
Infarto Encefálico/prevenção & controle , Encefalite/prevenção & controle , Caracteres Sexuais , Esplenectomia , Animais , Encéfalo/patologia , Infarto Encefálico/etiologia , Morte Celular/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/etiologia , Feminino , Regulação da Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/complicações , Leucócitos/metabolismo , Leucócitos/patologia , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/patologia
8.
Front Cell Neurosci ; 8: 284, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25309326

RESUMO

Stroke is a leading cause of death and disability in the United States. The lack of clinical success in stroke therapies can be attributed, in part, to inadequate basic research on aging rodents. The current study demonstrates that recombinant TCR ligand therapy uses different immunological mechanisms to protect young and older mice from experimental stroke. In young mice, RTL1000 therapy inhibited splenocyte efflux while reducing frequency of T cells and macrophages in the spleen. Older mice treated with RTL1000 exhibited a significant reduction in inflammatory cells in the brain and inhibition of splenic atrophy. Our data suggest age specific differences in immune response to stroke that allow unique targeting of stroke immunotherapies.

9.
Clin Transl Allergy ; 3(1): 29, 2013 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-24004581

RESUMO

BACKGROUND: Omalizumab, is a humanized anti-IgE monoclonal antibody used to treat allergic asthma. Decreased serum IgE levels, lower eosinophil and B cell counts have been noted as a result of treatment. In vitro studies and animal models support the hypothesis that omalizumab inhibits IgE synthesis by B cells and causes elimination of IgE-expressing cells either by induction of apoptosis or induction of anergy or tolerance. METHODS: We examined the influence of omalizumab on human tonsillar B cell survival and on the genes involved in IgE synthesis. Tonsillar B cells were stimulated with IL-4 plus anti-CD40 antibody to induce class switch recombination to IgE production in the presence or absence of omalizumab. Cell viability was assessed and RNA extracted to examine specific genes involved in IgE synthesis. CONCLUSIONS: We found that omalizumab reduced viable cell numbers but this was not through induction of apoptosis. IL-4R and germline Cϵ mRNA levels were decreased as well as the number of membrane IgE+ cells in B cells treated with omalizumab. These data suggest that omalizumab may decrease IgE synthesis by human B cells by specifically targeting membrane IgE-bearing B cells and inducing a state of anergy.

10.
Clin Immunol ; 148(2): 149-61, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770626

RESUMO

In insulin dependent diabetes mellitus (T1D), self-reactive T cells infiltrate pancreatic islets and induce beta cell destruction and dysregulation of blood glucose. A goal is to control only the self-reactive T cells, leaving the remainder of the T cell population free to protect the host. One approach is blockade of the second signal for T cell activation while allowing the first (antigen-specific) signal to occur. This work proposes that small peptides that block interaction of second signals delivered through the counter receptors LFA-1:ICAM-1 will induce attacking T cells (receiving the antigen signal) to become anergic or undergo apoptosis. In NOD mice, the peptides eliminated T cell reactivity against pancreatic antigens and reduced cellular infiltration into islets, which retained stronger density of insulin staining at five weeks after cessation of therapy. In in vitro studies the peptides induced nonresponsiveness during activation of T cells from mice and from human peripheral blood.


Assuntos
Células Secretoras de Insulina/fisiologia , Molécula 1 de Adesão Intercelular/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Peptídeos/farmacologia , Linfócitos T/fisiologia , Envelhecimento , Animais , Glicemia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Células Secretoras de Insulina/imunologia , Molécula 1 de Adesão Intercelular/genética , Antígeno-1 Associado à Função Linfocitária/genética , Camundongos , Camundongos Endogâmicos NOD , Linfócitos T/imunologia
11.
J Diabetes Res ; 2013: 965832, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23762878

RESUMO

Type 1 diabetes (T1D) is characterized by hyperglycemia due to lost or damaged islet insulin-producing ß -cells. Rodent models of T1D result in hyperglycemia, but with different forms of islet deterioration. This study focused on 1 toxin-induced and 2 autoimmune rodent models of T1D: BioBreeding Diabetes Resistant rats, nonobese diabetic mice, and Dark Agouti rats treated with streptozotocin. Immunochemistry was used to evaluate the insulin levels in the ß -cells, cell composition, and insulitis. T1D caused complete or significant loss of ß -cells in all animal models, while increasing numbers of α -cells. Lymphocytic infiltration was noted in and around islets early in the progression of autoimmune diabetes. The loss of lymphocytic infiltration coincided with the absence of ß -cells. In all models, the remaining α - and δ -cells regrouped by relocating to the islet center. The resulting islets were smaller in size and irregularly shaped. Insulin injections subsequent to induction of toxin-induced diabetes significantly preserved ß -cells and islet morphology. Diabetes in animal models is anatomically heterogeneous and involves important changes in numbers and location of the remaining α - and δ -cells. Comparisons with human pancreatic sections from healthy and diabetic donors showed similar morphological changes to the diabetic BBDR rat model.


Assuntos
Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Hiperglicemia/patologia , Ilhotas Pancreáticas/patologia , Animais , Glicemia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Feminino , Humanos , Hiperglicemia/imunologia , Hiperglicemia/fisiopatologia , Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/fisiopatologia , Masculino , Camundongos , Ratos
12.
Cell Immunol ; 271(2): 418-27, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21943647

RESUMO

With antigen stimulation, naïve CD4+ T cells differentiate to several effector or memory cell populations, and cytokines contribute to differentiation outcome. Several proteins on these cells receive costimulatory signals, but a systematic comparison of their differential effects on naïve T cell differentiation has not been conducted. Two costimulatory proteins, CD28 and ICAM-1, resident on human naïve CD4+ T cells were compared for participation in differentiation. Under controlled conditions, and with no added cytokines, costimulation through either CD3+CD28 or CD3+CAM-1 induced differentiation to T effector and T memory cells. In contrast, costimulation through CD3+ICAM-1 induced differentiation to Treg cells whereas costimulation through CD3+CD28 did not.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Apresentação de Antígeno , Antígenos CD28/antagonistas & inibidores , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/metabolismo , Humanos , Técnicas In Vitro , Molécula 1 de Adesão Intercelular/metabolismo , Interleucina-10/antagonistas & inibidores , Interleucina-10/metabolismo , Interleucina-2/antagonistas & inibidores , Interleucina-2/metabolismo , Ativação Linfocitária , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...