Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Immunol ; 54(6): e2350761, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38566526

RESUMO

In multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), early pathological features include immune cell infiltration into the central nervous system (CNS) and blood-brain barrier (BBB) disruption. We investigated the role of junctional adhesion molecule-A (JAM-A), a tight junction protein, in active EAE (aEAE) pathogenesis. Our study confirms JAM-A expression at the blood-brain barrier and its luminal redistribution during aEAE. JAM-A deficient (JAM-A-/-) C57BL/6J mice exhibited milder aEAE, unrelated to myelin oligodendrocyte glycoprotein-specific CD4+ T-cell priming. While JAM-A absence influenced macrophage behavior on primary mouse brain microvascular endothelial cells (pMBMECs) under flow in vitro, it did not impact T-cell extravasation across primary mouse brain microvascular endothelial cells. At aEAE onset, we observed reduced lymphocyte and CCR2+ macrophage infiltration into the spinal cord of JAM-A-/- mice compared to control littermates. This correlated with increased CD3+ T-cell accumulation in spinal cord perivascular spaces and brain leptomeninges, suggesting JAM-A absence leads to T-cell trapping in central nervous system border compartments. In summary, JAM-A plays a role in immune cell infiltration and clinical disease progression in aEAE.


Assuntos
Barreira Hematoencefálica , Encefalomielite Autoimune Experimental , Células Endoteliais , Macrófagos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Animais , Encefalomielite Autoimune Experimental/imunologia , Camundongos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/imunologia , Medula Espinal/patologia , Medula Espinal/imunologia , Medula Espinal/metabolismo , Linfócitos T CD4-Positivos/imunologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Modelos Animais de Doenças
2.
Int J Mol Sci ; 21(18)2020 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-32899717

RESUMO

Multiple sclerosis (MS) is a chronic, inflammatory, autoimmune disease of the central nervous system (CNS) which is associated with lower life expectancy and disability. The experimental antigen-induced encephalomyelitis (EAE) in mice is a useful animal model of MS, which allows exploring the etiopathogenetic mechanisms and testing novel potential therapeutic drugs. A new therapeutic paradigm for the treatment of MS was introduced in 2010 through the sphingosine 1-phosphate (S1P) analogue fingolimod (FTY720, Gilenya®), which acts as a functional S1P1 antagonist on T lymphocytes to deplete these cells from the blood. In this study, we synthesized two novel structures, ST-1893 and ST-1894, which are derived from fingolimod and chemically feature a morpholine ring in the polar head group. These compounds showed a selective S1P1 activation profile and a sustained S1P1 internalization in cultures of S1P1-overexpressing Chinese hamster ovary (CHO)-K1 cells, consistent with a functional antagonism. In vivo, both compounds induced a profound lymphopenia in mice. Finally, these substances showed efficacy in the EAE model, where they reduced clinical symptoms of the disease, and, on the molecular level, they reduced the T-cell infiltration and several inflammatory mediators in the brain and spinal cord. In summary, these data suggest that S1P1-selective compounds may have an advantage over fingolimod and siponimod, not only in MS but also in other autoimmune diseases.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Cloridrato de Fingolimode/farmacologia , Morfolinos/farmacologia , Animais , Células CHO , Sistema Nervoso Central/efeitos dos fármacos , Cricetulus , Modelos Animais de Doenças , Encefalomielite/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Cloridrato de Fingolimode/análogos & derivados , Imunossupressores/uso terapêutico , Ligantes , Linfopenia/tratamento farmacológico , Lisofosfolipídeos/metabolismo , Camundongos , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato/metabolismo , Medula Espinal/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
3.
Int J Mol Sci ; 20(6)2019 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-30901861

RESUMO

Multiple sclerosis (MS) is the most common inflammatory disorder of the central nervous system (CNS) in young adults leading to severe disability. Besides genetic traits, environmental factors contribute to MS pathogenesis. Cigarette smoking increases the risk of MS in an HLA-dependent fashion, but the underlying mechanisms remain unknown. Here, we explored the effect of cigarette smoke exposure on spontaneous and induced models of experimental autoimmune encephalomyelitis (EAE) by evaluating clinical disease and, when relevant, blood leukocytes and histopathology. In the relapsing-remitting (RR) transgenic model in SJL/J mice, we observed very low incidence in both smoke-exposed and control groups. In the optico-spinal encephalomyelitis (OSE) double transgenic model in C57BL/6 mice, the early onset of EAE prevented a meaningful evaluation of the effects of cigarette smoke. In EAE models induced by immunization, daily exposure to cigarette smoke caused a delayed onset of EAE followed by a protracted disease course in SJL/J mice. In contrast, cigarette smoke exposure ameliorated the EAE clinical score in C57BL/6J mice. Our exploratory studies therefore show that genetic background influences the effects of cigarette smoke on autoimmune neuroinflammation. Importantly, our findings expose the challenge of identifying an animal model for studying the influence of cigarette smoke in MS.


Assuntos
Encefalomielite Autoimune Experimental/diagnóstico , Encefalomielite Autoimune Experimental/etiologia , Patrimônio Genético , Fumar/efeitos adversos , Idade de Início , Animais , Biópsia , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Encefalomielite Autoimune Experimental/metabolismo , Imuno-Histoquímica , Camundongos , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/etiologia , Esclerose Múltipla/metabolismo , Fenótipo , Medição de Risco , Fatores de Risco , Índice de Gravidade de Doença , Medula Espinal/metabolismo , Medula Espinal/patologia
4.
Brain Behav Immun ; 73: 3-20, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29920328

RESUMO

In multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) autoaggressive CD4+ T cells cross the blood-brain barrier (BBB) and cause neuroinflammation. Therapeutic targeting of CD4+ T-cell trafficking into the CNS by blocking α4-integrins has proven beneficial for the treatment of MS but comes with associated risks, probably due to blocking CD8+ T cell mediated CNS immune surveillance. Our recent observations show that CD8+ T cells also rely on α4ß1-integrins to cross the BBB. Besides vascular cell adhesion molecule-1 (VCAM-1), we identified junctional adhesion molecule-B (JAM-B) as a novel vascular α4ß1-integrin ligand involved in CD8+ T-cell migration across the BBB. This prompted us to investigate, if JAM-B also mediates CD4+ T-cell migration across the BBB. We first ensured that encephalitogenic T cells can bind to JAM-B in vitro and next compared EAE pathogenesis in JAM-B-/- C57BL/6J mice and their wild-type littermates. Following immunization with MOGaa35-55 peptide, JAM-B-/- mice developed ameliorated EAE compared to their wild-type littermates. At the same time, we isolated higher numbers of CD45+ infiltrating immune cells from the CNS of JAM-B-/- C57BL/6J mice suffering from EAE. Immunofluorescence staining revealed that the majority of CD45+ inflammatory cells accumulated in the leptomeningeal and perivascular spaces of the CNS behind the BBB but do not gain access to the CNS parenchyma. Trapping of CNS inflammatory cells was not due to increased inflammatory cell proliferation. Neither a loss of BBB integrity or BBB polarity potentially affecting local chemokine gradients nor a lack of focal gelatinase activation required for CNS parenchymal immune cell entry across the glia limitans could be detected in JAM-B-/- mice. Lack of a role for JAM-B in the effector phase of EAE was supported by the observation that we did not detect any role for JAM-B in EAE pathogenesis, when EAE was elicited by in vitro activated MOG aa35-55-specific CD4+ effector T cells. On the other hand, we also failed to demonstrate any role of JAM-B in in vivo priming, proliferation or polarization of MOGaa35-55-specific CD4+ T cells in peripheral immune organs. Finally, our study excludes expression of and thus a role for JAM-B on peripheral and CNS infiltrating myeloid cells. Taken together, although endothelial JAM-B is not required for immune cell trafficking across the BBB in EAE, in its absence accumulation of inflammatory cells mainly in CNS leptomeningeal spaces leads to amelioration of EAE.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Molécula B de Adesão Juncional/metabolismo , Molécula B de Adesão Juncional/fisiologia , Animais , Barreira Hematoencefálica/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Movimento Celular/fisiologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/fisiopatologia , Endotélio Vascular/metabolismo , Feminino , Integrina alfa4beta1/metabolismo , Molécula B de Adesão Juncional/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/metabolismo , Esclerose Múltipla/fisiopatologia , Glicoproteína Mielina-Oligodendrócito/farmacologia , Células Mieloides/metabolismo , Células Mieloides/fisiologia , Junções Íntimas/metabolismo
5.
Biochim Biophys Acta ; 1862(3): 461-71, 2016 03.
Artigo em Inglês | MEDLINE | ID: mdl-26527183

RESUMO

Each year about 650,000 Europeans die from stroke and a similar number lives with the sequelae of multiple sclerosis (MS). Stroke and MS differ in their etiology. Although cause and likewise clinical presentation set the two diseases apart, they share common downstream mechanisms that lead to damage and recovery. Demyelination and axonal injury are characteristics of MS but are also observed in stroke. Conversely, hallmarks of stroke, such as vascular impairment and neurodegeneration, are found in MS. However, the most conspicuous common feature is the marked neuroinflammatory response, marked by glia cell activation and immune cell influx. In MS and stroke the blood-brain barrier is disrupted allowing bone marrow-derived macrophages to invade the brain in support of the resident microglia. In addition, there is a massive invasion of auto-reactive T-cells into the brain of patients with MS. Though less pronounced a similar phenomenon is also found in ischemic lesions. Not surprisingly, the two diseases also resemble each other at the level of gene expression and the biosynthesis of other proinflammatory mediators. While MS has traditionally been considered to be an autoimmune neuroinflammatory disorder, the role of inflammation for cerebral ischemia has only been recognized later. In the case of MS the long track record as neuroinflammatory disease has paid off with respect to treatment options. There are now about a dozen of approved drugs for the treatment of MS that specifically target neuroinflammation by modulating the immune system. Interestingly, experimental work demonstrated that drugs that are in routine use to mitigate neuroinflammation in MS may also work in stroke models. Examples include Fingolimod, glatiramer acetate, and antibodies blocking the leukocyte integrin VLA-4. Moreover, therapeutic strategies that were discovered in experimental autoimmune encephalomyelitis (EAE), the animal model of MS, turned out to be also effective in experimental stroke models. This suggests that previous achievements in MS research may be relevant for stroke. Interestingly, the converse is equally true. Concepts on the neurovascular unit that were developed in a stroke context turned out to be applicable to neuroinflammatory research in MS. Examples include work on the important role of the vascular basement membrane and the BBB for the invasion of immune cells into the brain. Furthermore, tissue plasminogen activator (tPA), the only established drug treatment in acute stroke, modulates the pathogenesis of MS. Endogenous tPA is released from endothelium and astroglia and acts on the BBB, microglia and other neuroinflammatory cells. Thus, the vascular perspective of stroke research provides important input into the mechanisms on how endothelial cells and the BBB regulate inflammation in MS, particularly the invasion of immune cells into the CNS. In the current review we will first discuss pathogenesis of both diseases and current treatment regimens and will provide a detailed overview on pathways of immune cell migration across the barriers of the CNS and the role of activated astrocytes in this process. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.


Assuntos
Barreira Hematoencefálica/patologia , Sistema Nervoso Central/patologia , Inflamação/patologia , Esclerose Múltipla/patologia , Acidente Vascular Cerebral/patologia , Animais , Astrócitos/imunologia , Astrócitos/patologia , Barreira Hematoencefálica/imunologia , Encéfalo/imunologia , Encéfalo/patologia , Sistema Nervoso Central/imunologia , Humanos , Imunidade Celular , Inflamação/imunologia , Esclerose Múltipla/imunologia , Acidente Vascular Cerebral/imunologia
6.
Eur J Immunol ; 46(10): 2481-2483, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27469626

RESUMO

Considering the 3R rules of animal experimentation, we asked if refined scoring of experimental autoimmune encephalomyelitis (EAE) in mice improves documentation of clinical EAE allowing to perform a more powerful statistical analysis. Surprisingly, refined EAE scoring failed to improve statistical outcome comparing the overall disease courses between two groups of mice.


Assuntos
Encefalomielite Autoimune Experimental/diagnóstico , Esclerose Múltipla/diagnóstico , Projetos de Pesquisa , Animais , Modelos Animais de Doenças , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Variações Dependentes do Observador , Índice de Gravidade de Doença
7.
Acta Neuropathol ; 134(6): 851-868, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28762187

RESUMO

Neuroinflammation contributes substantially to stroke pathophysiology. Cerebral invasion of peripheral leukocytes-particularly T cells-has been shown to be a key event promoting inflammatory tissue damage after stroke. While previous research has focused on the vascular invasion of T cells into the ischemic brain, the choroid plexus (ChP) as an alternative cerebral T-cell invasion route after stroke has not been investigated. We here report specific accumulation of T cells in the peri-infarct cortex and detection of T cells as the predominant population in the ipsilateral ChP in mice as well as in human post-stroke autopsy samples. T-cell migration from the ChP to the peri-infarct cortex was confirmed by in vivo cell tracking of photoactivated T cells. In turn, significantly less T cells invaded the ischemic brain after photothrombotic lesion of the ipsilateral ChP and in a stroke model encompassing ChP ischemia. We detected a gradient of CCR2 ligands as the potential driving force and characterized the neuroanatomical pathway for the intracerebral migration. In summary, our study demonstrates that the ChP is a key invasion route for post-stroke cerebral T-cell invasion and describes a CCR2-ligand gradient between cortex and ChP as the potential driving mechanism for this invasion route.


Assuntos
Isquemia Encefálica/fisiopatologia , Movimento Celular/fisiologia , Plexo Corióideo/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia , Linfócitos T/fisiologia , Idoso , Idoso de 80 Anos ou mais , Animais , Lesões Encefálicas Traumáticas/patologia , Lesões Encefálicas Traumáticas/fisiopatologia , Isquemia Encefálica/patologia , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Quimiocina CCL2/metabolismo , Plexo Corióideo/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/patologia , Células Mieloides/fisiologia , Acidente Vascular Cerebral/patologia , Linfócitos T/patologia
8.
Curr Opin Hematol ; 22(1): 53-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25427141

RESUMO

PURPOSE OF REVIEW: Neutrophil extravasation from the blood into tissues is initiated by tethering and rolling of neutrophils on endothelial cells, followed by neutrophil integrin activation and shear resistant arrest, crawling, diapedesis and breaching the endothelial basement membrane harbouring pericytes. Endothelial intercellular cell adhesion molecule (ICAM)-1 and ICAM-2, in conjunction with ICAM-1 on pericytes, critically contribute to each step. In addition, epithelial ICAM-1 is involved in neutrophil migration to peri-epithelial sites. The most recent findings on the role of ICAM-1 and ICAM-2 for neutrophil migration into tissues will be reviewed here. RECENT FINDINGS: Signalling via endothelial ICAM-1 and ICAM-2 contributes to stiffness of the endothelial cells at sites of chronic inflammation and junctional maturation, respectively. Endothelial ICAM-2 contributes to neutrophil crawling and initiation of paracellular diapedesis, which then proceeds independent of ICAM-2. Substantial transcellular neutrophil diapedesis across the blood-brain barrier is strictly dependent on endothelial ICAM-1 and ICAM-2. Endothelial ICAM-1 or ICAM-2 is involved in neutrophil-mediated plasma leakage. ICAM-1 on pericytes assists the final step of neutrophil extravasation. Epithelial ICAM-1 rather indirectly promotes neutrophil migration to peri-epithelial sites. SUMMARY: ICAM-1 and ICAM-2 are involved in each step of neutrophil extravasation, and have redundant but also distinct functions. Analysis of the role of endothelial ICAM-1 requires simultaneous consideration of ICAM-2.


Assuntos
Antígenos CD/fisiologia , Moléculas de Adesão Celular/fisiologia , Molécula 1 de Adesão Intercelular/fisiologia , Neutrófilos/fisiologia , Migração Transendotelial e Transepitelial/fisiologia , Animais , Humanos , Neutrófilos/citologia
9.
Eur J Immunol ; 44(8): 2287-94, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24740164

RESUMO

T-cell migration across the blood-brain barrier is a crucial step in the pathogenesis of EAE, an animal model for MS. Live cell imaging studies demonstrated that P-selectin glycoprotein ligand-1 (PSGL-1) and its endothelial ligands E- and P-selectin mediate the initial rolling of T cells in brain vessels during EAE. As functional absence of PSGL-1 or E/P-selectins does not result in ameliorated EAE, we speculated that T-cell entry into the spinal cord is independent of PSGL-1 and E/P-selectin. Performing intravital microscopy, we observed the interaction of WT or PSGL-1(-/-) proteolipid protein-specific T cells in inflamed spinal cord microvessels of WT or E/P-selectin(-/-) SJL/J mice during EAE. T-cell rolling but not T-cell capture was completely abrogated in the absence of either PSGL-1 or E- and P-selectin, resulting in a significantly reduced number of T cells able to firmly adhere in the inflamed spinal cord microvessels, but did not lead to reduced T-cell invasion into the CNS parenchyma. Thus, PSGL-1 interaction with E/P-selectin is essential for T-cell rolling in inflamed spinal cord microvessels during EAE. Taken together with previous observations, our findings show that T-cell rolling is not required for successful T-cell entry into the CNS and initiation of EAE.


Assuntos
Selectina E/imunologia , Encefalomielite Autoimune Experimental/imunologia , Glicoproteínas de Membrana/imunologia , Microvasos/imunologia , Selectina-P/imunologia , Linfócitos T/imunologia , Animais , Barreira Hematoencefálica/imunologia , Adesão Celular/imunologia , Linhagem Celular , Movimento Celular/imunologia , Ligantes , Camundongos , Medula Espinal/imunologia
10.
Cell Tissue Res ; 358(2): 465-79, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25107608

RESUMO

Barrier characteristics of brain endothelial cells forming the blood-brain barrier (BBB) are tightly regulated by cellular and acellular components of the neurovascular unit. During embryogenesis, the accumulation of the heparan sulfate proteoglycan agrin in the basement membranes ensheathing brain vessels correlates with BBB maturation. In contrast, loss of agrin deposition in the vasculature of brain tumors is accompanied by the loss of endothelial junctional proteins. We therefore wondered whether agrin had a direct effect on the barrier characteristics of brain endothelial cells. Agrin increased junctional localization of vascular endothelial (VE)-cadherin, ß-catenin, and zonula occludens-1 (ZO-1) but not of claudin-5 and occludin in the brain endothelioma cell line bEnd5 without affecting the expression levels of these proteins. This was accompanied by an agrin-induced reduction of the paracellular permeability of bEnd5 monolayers. In vivo, the lack of agrin also led to reduced junctional localization of VE-cadherin in brain microvascular endothelial cells. Taken together, our data support the notion that agrin contributes to barrier characteristics of brain endothelium by stabilizing the adherens junction proteins VE-cadherin and ß-catenin and the junctional protein ZO-1 to brain endothelial junctions.


Assuntos
Junções Aderentes/metabolismo , Agrina/metabolismo , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Proteoglicanas de Heparan Sulfato/metabolismo , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Proliferação de Células , Galinhas , Células Endoteliais/citologia , Células HEK293 , Humanos , Camundongos , Microvasos/citologia , Microvasos/metabolismo , Permeabilidade , Estabilidade Proteica , Transporte Proteico , Coloração e Rotulagem , Proteína da Zônula de Oclusão-1/metabolismo , beta Catenina/metabolismo
11.
Acta Neuropathol ; 125(3): 395-412, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23269317

RESUMO

The migration of polymorphonuclear granulocytes (PMN) into the brain parenchyma and release of their abundant proteases are considered the main causes of neuronal cell death and reperfusion injury following ischemia. Yet, therapies targeting PMN egress have been largely ineffective. To address this discrepancy we investigated the temporo-spatial localization of PMNs early after transient ischemia in a murine transient middle cerebral artery occlusion (tMCAO) model and human stroke specimens. Using specific markers that distinguish PMN (Ly6G) from monocytes/macrophages (Ly6C) and that define the cellular and basement membrane boundaries of the neurovascular unit (NVU), histology and confocal microscopy revealed that virtually no PMNs entered the infarcted CNS parenchyma. Regardless of tMCAO duration, PMNs were mainly restricted to luminal surfaces or perivascular spaces of cerebral vessels. Vascular PMN accumulation showed no spatial correlation with increased vessel permeability, enhanced expression of endothelial cell adhesion molecules, platelet aggregation or release of neutrophil extracellular traps. Live cell imaging studies confirmed that oxygen and glucose deprivation followed by reoxygenation fail to induce PMN migration across a brain endothelial monolayer under flow conditions in vitro. The absence of PMN infiltration in infarcted brain tissues was corroborated in 25 human stroke specimens collected at early time points after infarction. Our observations identify the NVU rather than the brain parenchyma as the site of PMN action after CNS ischemia and suggest reappraisal of targets for therapies to reduce reperfusion injury after stroke.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Encéfalo/patologia , Granulócitos/patologia , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Animais , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , Barreira Hematoencefálica/patologia , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Endotélio/patologia , Lateralidade Funcional , Regulação da Expressão Gênica/fisiologia , Glucose/deficiência , Humanos , Hipóxia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Oxigênio/administração & dosagem
12.
Fluids Barriers CNS ; 20(1): 39, 2023 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-37264368

RESUMO

The choroid plexus (ChP) has been suggested as an alternative central nervous system (CNS) entry site for CCR6+ Th17 cells during the initiation of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis (MS). To advance our understanding of the importance of the ChP in orchestrating CNS immune cell entry during neuroinflammation, we here directly compared the accumulation of CD45+ immune cell subsets in the ChP, the brain and spinal cord at different stages of EAE by flow cytometry. We found that the ChP harbors high numbers of CD45int resident innate but also of CD45hi adaptive immune cell subsets including CCR6+ Th17 cells. With the exception to tissue-resident myeloid cells and B cells, numbers of CD45+ immune cells and specifically of CD4+ T cells increased in the ChP prior to EAE onset and remained elevated while declining in brain and spinal cord during chronic disease. Increased numbers of ChP immune cells preceded their increase in the cerebrospinal fluid (CSF). Th17 but also other CD4+ effector T-cell subsets could migrate from the basolateral to the apical side of the blood-cerebrospinal fluid barrier (BCSFB) in vitro, however, diapedesis of effector Th cells including that of Th17 cells did not require interaction of CCR6 with BCSFB derived CCL20. Our data underscore the important role of the ChP as CNS immune cell entry site in the context of autoimmune neuroinflammation.


Assuntos
Encefalomielite Autoimune Experimental , Animais , Camundongos , Plexo Corióideo/fisiologia , Doenças Neuroinflamatórias , Encéfalo , Sistema Nervoso Central , Camundongos Endogâmicos C57BL
13.
Glia ; 60(11): 1646-59, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22782669

RESUMO

Brain edema is the main cause of death from brain infarction. The polarized expression of the water channel protein aquaporin-4 (AQP4) on astroglial endfeet surrounding brain microvessels suggests a role in brain water balance. Loss of astrocyte foot process anchoring to the basement membrane (BM) accompanied by the loss of polarized localization of AQP4 to astrocytic endfeet has been shown to be associated with vasogenic/extracellular edema in neuroinflammation. Here, we asked if loss of astrocyte polarity is also observed in cytotoxic/intracellular edema following focal brain ischemia after transient middle cerebral artery occlusion (tMCAO). Upon mild focal brain ischemia, we observed diminished immunostaining for the BM components laminin α4, laminin α2, and the proteoglycan agrin, in the core of the lesion, but not in BMs in the surrounding penumbra. Staining for the astrocyte endfoot anchorage protein ß-dystroglycan (DG) was dramatically reduced in both the lesion core and the penumbra, and AQP4 and Kir4.1 showed a loss of polarized localization to astrocytic endfeet. Interestingly, we observed that mice deficient for agrin expression in the brain lack polarized localization of ß-DG and AQP4 at astrocytic endfeet and do not develop early cytotoxic/intracellular edema following tMCAO. Taken together, these data indicate that the binding of DG to agrin embedded in the subjacent BM promotes polarized localization of AQP4 to astrocyte endfeet. Reduced DG protein levels and redistribution of AQP4 as observed upon tMCAO might therefore counteract early edema formation and reflect a beneficial mechanism operating in the brain to minimize damage upon ischemia.


Assuntos
Aquaporina 4/metabolismo , Astrócitos/metabolismo , Isquemia Encefálica/metabolismo , Polaridade Celular , Edema/metabolismo , Animais , Astrócitos/patologia , Membrana Basal/metabolismo , Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/patologia , Edema/etiologia , Edema/patologia , Camundongos , Camundongos Transgênicos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo
14.
J Immunol ; 184(12): 7196-206, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20483748

RESUMO

Dendritic cells (DCs) within the CNS are recognized to play an important role in the effector phase and propagation of the immune response in experimental autoimmune encephalomyelitis (EAE), a mouse model for multiple sclerosis. However, the mechanisms regulating DC trafficking into the CNS still need to be characterized. In this study, we show by performing intravital fluorescence videomicroscopy of the inflamed spinal cord white-matter microvasculature in SJL mice with EAE that immature, and to a lesser extent, LPS-matured, bone marrow-derived DCs efficiently interact with the CNS endothelium by rolling, capturing, and firm adhesion. Immature but not LPS-matured DCs efficiently migrated across the wall of inflamed parenchymal microvessels into the CNS. Blocking alpha4 integrins interfered with the adhesion but not the rolling or capturing of immature and LPS-matured DCs to the CNS microvascular endothelium, inhibiting their migration across the vascular wall. Functional absence of beta1 integrins but not of beta7 integrins or alpha4beta7 integrin similarly reduced the adhesion of immature DCs to the CNS microvascular endothelium, demonstrating that alpha4beta1 but not alpha4beta7 integrin mediates this step of immature DCs interaction with the inflamed blood-brain barrier during EAE. Our study shows that during EAE, especially immature DCs migrate into the CNS, where they may be crucial for the perpetuation of the CNS-targeted autoimmune response. Thus therapeutic targeting of alpha4 integrins affects DC trafficking into the CNS and may therefore lead to the resolution of the CNS autoimmune inflammation by reducing the number of CNS professional APCs.


Assuntos
Barreira Hematoencefálica/metabolismo , Quimiotaxia de Leucócito/imunologia , Células Dendríticas/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Integrina alfa4beta1/metabolismo , Animais , Barreira Hematoencefálica/imunologia , Separação Celular , Células Dendríticas/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Citometria de Fluxo , Imunofluorescência , Integrina alfa4beta1/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Medula Espinal/imunologia , Medula Espinal/patologia
15.
Adv Healthc Mater ; 10(2): e2001375, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33241667

RESUMO

Delivery of therapeutics to the central nervous system (CNS) is challenging due to the presence of the blood-brain barrier (BBB). Amongst various approaches that have been explored to facilitate drug delivery to the CNS, the use of cells that have the intrinsic ability to cross the BBB is relatively unexplored, yet very attractive. This paper presents a first proof-of-concept that demonstrates the feasibility of activated effector/memory CD4+ helper T cells (CD4+ TEM cells) as carriers for the delivery of polymer nanoparticles across the BBB. This study shows that CD4+ TEM cells can be decorated with poly(ethylene glycol)-modified polystyrene nanoparticles using thiol-maleimide coupling chemistry, resulting in the immobilization of ≈105 nanoparticles per cell as determined by confocal microscopy. The ability of these cells to serve as carriers to transport nanoparticles across the BBB is established in vitro and in vivo. Using in vitro BBB models, CD4+ TEM cells are found to be able to transport nanoparticles across the BBB both under static conditions as well as under physiological flow. Finally, upon systemic administration, nanoparticle-modified T cells are shown to enter the brain parenchyma of mice, demonstrating the brain delivery potential of this T cell subset in allogeneic hosts.


Assuntos
Barreira Hematoencefálica , Nanopartículas , Animais , Transporte Biológico , Sistemas de Liberação de Medicamentos , Camundongos , Polímeros , Linfócitos T
16.
Neuropharmacology ; 186: 108464, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33460688

RESUMO

The sphingosine 1-phosphate (S1P) receptor 1 (S1P1) has emerged as a therapeutic target for the treatment of multiple sclerosis (MS). Fingolimod (FTY720) is the first functional antagonist of S1P1 that has been approved for oral treatment of MS. Previously, we have developed novel butterfly derivatives of FTY720 that acted similar to FTY720 in reducing disease symptoms in a mouse model of experimental autoimmune encephalomyelitis (EAE). In this study, we have synthesized a piperidine derivative of the oxazolo-oxazole compounds, denoted ST-1505, and its ring-opened analogue ST-1478, and characterised their in-vitro and in-vivo functions. Notably, the 3-piperidinopropyloxy moiety resembles a structural motif of pitolisant, a drug with histamine H3R antagonistic/inverse agonist activity approved for the treatment of narcolepsy. Both novel compounds exerted H3R affinities, and in addition, ST-1505 was characterised as a dual S1P1+3 agonist, whereas ST-1478 was a dual S1P1+5 agonist. Both multitargeting compounds were also active in mice and reduced the lymphocyte numbers as well as diminished disease symptoms in the mouse model of MS. The effect of ST-1478 was dependent on SK-2 activity suggesting that it is a prodrug like FTY720, but with a more selective S1P receptor activation profile, whereas ST-1505 is a fully active drug even in the absence of SK-2. In summary, these data suggest that the well soluble piperidine derivatives ST-1505 and ST-1478 hold promise as novel drugs for the treatment of MS and other autoimmune or inflammatory diseases, and by their H3R antagonist potency, they might additionally improve cognitive impairment during disease.


Assuntos
Encefalomielite Autoimune Experimental/prevenção & controle , Cloridrato de Fingolimode/administração & dosagem , Antagonistas dos Receptores Histamínicos H3/administração & dosagem , Esclerose Múltipla/prevenção & controle , Fármacos Neuroprotetores/administração & dosagem , Receptores de Esfingosina-1-Fosfato/agonistas , Animais , Células CHO , Cricetinae , Cricetulus , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Cloridrato de Fingolimode/análogos & derivados , Cloridrato de Fingolimode/química , Antagonistas dos Receptores Histamínicos H3/química , Antagonistas dos Receptores Histamínicos H3/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla/metabolismo , Fármacos Neuroprotetores/química , Estrutura Secundária de Proteína , Receptores de Esfingosina-1-Fosfato/metabolismo
17.
Cell Rep ; 35(9): 109201, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34077725

RESUMO

The chronic neuro-inflammatory character of multiple sclerosis (MS) suggests that the natural process to resolve inflammation is impaired. This protective process is orchestrated by specialized pro-resolving lipid mediators (SPMs), but to date, the role of SPMs in MS remains largely unknown. Here, we provide in vivo evidence that treatment with the SPM lipoxin A4 (LXA4) ameliorates clinical symptoms of experimental autoimmune encephalomyelitis (EAE) and inhibits CD4+ and CD8+ T cell infiltration into the central nervous system (CNS). Moreover, we show that LXA4 potently reduces encephalitogenic Th1 and Th17 effector functions, both in vivo and in isolated human T cells from healthy donors and patients with relapsing-remitting MS. Finally, we demonstrate that LXA4 affects the spinal cord lipidome by significantly reducing the levels of pro-inflammatory lipid mediators during EAE. Collectively, our findings provide mechanistic insight into LXA4-mediated amelioration of neuro-inflammation and highlight the potential clinical application of LXA4 for MS.


Assuntos
Encéfalo/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Lipidômica , Lipoxinas/farmacologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Linfócitos T/imunologia , Adulto , Animais , Encéfalo/patologia , Movimento Celular/efeitos dos fármacos , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Lipoxinas/química , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Medula Espinal/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th17/efeitos dos fármacos , Células Th17/imunologia
18.
J Neurosci Res ; 88(4): 712-21, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19795370

RESUMO

Monitoring pathology/regeneration in experimental models of de-/remyelination requires an accurate measure not only of functional changes but also of the amount of myelin. We tested whether X-ray diffraction (XRD), which measures periodicity in unfixed myelin, can assess the structural integrity of myelin in fixed tissue. From laboratories involved in spinal cord injury research and in studying the aging primate brain, we solicited "blind" samples and used an electronic detector to record rapidly the diffraction patterns (30 min each pattern) from them. We assessed myelin integrity by measuring its periodicity and relative amount. Fixation of tissue itself introduced +/-10% variation in periodicity and +/-40% variation in relative amount of myelin. For samples having the most native-like periods, the relative amounts of myelin detected allowed distinctions to be made between normal and demyelinating segments, between motor and sensory tracts within the spinal cord, and between aged and young primate CNS. Different periodicities also allowed distinctions to be made between samples from spinal cord and nerve roots and between well-fixed and poorly fixed samples. Our findings suggest that, in addition to evaluating the effectiveness of different fixatives, XRD could also be used as a robust and rapid technique for quantitating the relative amount of myelin among spinal cords and other CNS tissue samples from experimental models of de- and remyelination.


Assuntos
Sistema Nervoso Central/patologia , Doenças Desmielinizantes/patologia , Bainha de Mielina/patologia , Envelhecimento , Animais , Doenças Desmielinizantes/induzido quimicamente , Modelos Animais de Doenças , Etídio , Macaca mulatta , Camundongos , Bainha de Mielina/metabolismo , Ratos , Difração de Raios X/métodos
19.
Cell Host Microbe ; 28(1): 69-78.e6, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32497498

RESUMO

Clostridium perfringens ß-toxin (CPB) is a highly active ß-pore-forming toxin (ß-PFT) and the essential virulence factor for fatal, necro-hemorrhagic enteritis in animals and humans. The molecular mechanisms involved in CPB's action on its target, the endothelium of small intestinal vessels, are poorly understood. Here, we identify platelet endothelial cell adhesion molecule-1 (CD31 or PECAM-1) as the specific membrane receptor for CPB on endothelial cells. CD31 expression corresponds with the cell-type specificity of CPB, and it is essential for toxicity in cultured cells and mice. Ectopic CD31 expression renders resistant cells and liposomes susceptible to CPB-induced membrane damage. Moreover, the extracellular Ig6 domain of mouse, human, and porcine CD31 is essential for the interaction with CPB. Hence, our results explain the cell-type specificity of CPB in vitro and in the natural disease caused by C. perfringens type C.


Assuntos
Toxinas Bacterianas/metabolismo , Clostridium perfringens/patogenicidade , Células Endoteliais/metabolismo , Células Endoteliais/microbiologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Cultivadas , Infecções por Clostridium/microbiologia , Clostridium perfringens/fisiologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Domínios e Motivos de Interação entre Proteínas , Suínos , Fatores de Virulência/metabolismo
20.
Neurorehabil Neural Repair ; 23(6): 535-45, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19270266

RESUMO

BACKGROUND: The authors have shown that rats can be retrained to swim after a moderately severe thoracic spinal cord contusion. They also found that improvements in body position and hindlimb activity occurred rapidly over the first 2 weeks of training, reaching a plateau by week 4. Overground walking was not influenced by swim training, suggesting that swimming may be a task-specific model of locomotor retraining. OBJECTIVE: To provide a quantitative description of hindlimb movements of uninjured adult rats during swimming, and then after injury and retraining. METHODS: The authors used a novel and streamlined kinematic assessment of swimming in which each limb is described in 2 dimensions, as 3 segments and 2 angles. RESULTS: The kinematics of uninjured rats do not change over 4 weeks of daily swimming, suggesting that acclimatization does not involve refinements in hindlimb movement. After spinal cord injury, retraining involved increases in hindlimb excursion and improved limb position, but the velocity of the movements remained slow. CONCLUSION: These data suggest that the activity pattern of swimming is hardwired in the rat spinal cord. After spinal cord injury, repetition is sufficient to bring about significant improvements in the pattern of hindlimb movement but does not improve the forces generated, leaving the animals with persistent deficits. These data support the concept that force (load) and pattern generation (recruitment) are independent and may have to be managed together with respect to postinjury rehabilitation.


Assuntos
Membro Posterior/fisiopatologia , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/reabilitação , Natação , Animais , Fenômenos Biomecânicos , Modelos Animais de Doenças , Feminino , Testes Neuropsicológicos , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA