Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(1): 271-276, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30559188

RESUMO

Blood-brain barrier (BBB) disruption and transendothelial trafficking of immune cells into the central nervous system (CNS) are pathophysiological hallmarks of neuroinflammatory disorders like multiple sclerosis (MS). Recent evidence suggests that the kallikrein-kinin and coagulation system might participate in this process. Here, we identify plasma kallikrein (KK) as a specific direct modulator of BBB integrity. Levels of plasma prekallikrein (PK), the precursor of KK, were markedly enhanced in active CNS lesions of MS patients. Deficiency or pharmacologic blockade of PK renders mice less susceptible to experimental autoimmune encephalomyelitis (a model of MS) and is accompanied by a remarkable reduction of BBB disruption and CNS inflammation. In vitro analysis revealed that KK modulates endothelial cell function in a protease-activated receptor-2-dependent manner, leading to an up-regulation of the cellular adhesion molecules Intercellular Adhesion Molecule 1 and Vascular Cell Adhesion Molecule 1, thereby amplifying leukocyte trafficking. Our study demonstrates that PK is an important direct regulator of BBB integrity as a result of its protease function. Therefore, KK inhibition can decrease BBB damage and cell invasion during neuroinflammation and may offer a strategy for the treatment of MS.


Assuntos
Bradicinina/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Calicreínas/metabolismo , Receptor PAR-2/metabolismo , Animais , Barreira Hematoencefálica , Western Blotting , Bradicinina/fisiologia , Encefalomielite Autoimune Experimental/fisiopatologia , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Calicreínas/antagonistas & inibidores , Calicreínas/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla/metabolismo , Receptor PAR-2/fisiologia
2.
Int J Mol Sci ; 22(18)2021 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-34576080

RESUMO

Aside from the established immune-mediated etiology of multiple sclerosis (MS), compelling evidence implicates platelets as important players in disease pathogenesis. Specifically, numerous studies have highlighted that activated platelets promote the central nervous system (CNS)-directed adaptive immune response early in the disease course. Platelets, therefore, present a novel opportunity for modulating the neuroinflammatory process that characterizes MS. We hypothesized that the well-known antiplatelet agent acetylsalicylic acid (ASA) could inhibit neuroinflammation by affecting platelets if applied at low-dose and investigated its effect during experimental autoimmune encephalomyelitis (EAE) as a model to study MS. We found that oral administration of low-dose ASA alleviates symptoms of EAE accompanied by reduced inflammatory infiltrates and less extensive demyelination. Remarkably, the percentage of CNS-infiltrated CD4+ T cells, the major drivers of neuroinflammation, was decreased to 40.98 ± 3.28% in ASA-treated mice compared to 56.11 ± 1.46% in control animals at the disease maximum as revealed by flow cytometry. More interestingly, plasma levels of thromboxane A2 were decreased, while concentrations of platelet factor 4 and glycoprotein VI were not affected by low-dose ASA treatment. Overall, we demonstrate that low-dose ASA could ameliorate the platelet-dependent neuroinflammatory response in vivo, thus indicating a potential treatment approach for MS.


Assuntos
Aspirina/farmacologia , Plaquetas/patologia , Encéfalo/patologia , Inflamação/patologia , Esclerose Múltipla/sangue , Esclerose Múltipla/patologia , Inibidores da Agregação Plaquetária/farmacologia , Animais , Aspirina/administração & dosagem , Aspirina/uso terapêutico , Plaquetas/efeitos dos fármacos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Inflamação/imunologia , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Tromboxano A2/biossíntese
3.
New Dir Child Adolesc Dev ; 2021(177): 77-99, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33942957

RESUMO

Guided by risk and resilience and attachment perspectives, the present study examined whether teacher-student relationship quality and school climate can buffer against the deleterious effects of perceived personal and group ethnic discrimination on psychological and academic domains. We conducted multilevel analyses of seventh graders (40 classrooms; N = 456; 47% female) with different cultural self-identifications in Germany. Partially confirming pre-registered hypotheses, results indicated that high levels of perceived personal discrimination were negatively associated with global self-esteem and emotional school engagement. Contrary to our expectations, neither perceived personal nor group discrimination negatively predicted academic self-concept. In addition, teacher-student relationship quality but not school climate buffered the relationship between both personal and group discrimination and global self-esteem and emotional school engagement such that the association was less negative when relationship quality was high. Taken together, our results underscore the importance of considering the different targets of discrimination (i.e., personal self and own group), and that positive teacher-student relationship can be especially beneficial and empowering for youth who are exposed to ethnic discrimination.


Assuntos
Pessoal de Educação , Estudantes , Adolescente , Emoções , Feminino , Humanos , Masculino , Instituições Acadêmicas , Autoimagem
4.
Neurosignals ; 26(1): 77-93, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30481775

RESUMO

BACKGROUND/AIMS: Multiple sclerosis (MS) is a prototypical autoimmune central nervous system (CNS) disease. Particularly progressive forms of MS (PMS) show significant neuroaxonal damage as consequence of demyelination and neuronal hyperexcitation. Immuno-modulatory treatment strategies are beneficial in relapsing MS (RMS), but mostly fail in PMS. Pregabalin (Lyrica®) is prescribed to MS patients to treat neuropathic pain. Mechanistically, it targets voltage-dependent Ca2+ channels and reduces harmful neuronal hyperexcitation in mouse epilepsy models. Studies suggest that GABA analogues like pregabalin exert neuroprotective effects in animal models of ischemia and trauma. METHODS: We tested the impact of pregabalin in a mouse model of MS (experimental autoimmune encephalomyelitis, EAE) and performed histological and immunological evaluations as well as intravital two-photon-microscopy of brainstem EAE lesions. RESULTS: Both prophylactic and therapeutic treatments ameliorated the clinical symptoms of EAE and reduced immune cell infiltration into the CNS. On neuronal level, pregabalin reduced long-term potentiation in hippocampal brain slices indicating an impact on mechanisms of learning and memory. In contrast, T cells, microglia and brain endothelial cells were unaffected by pregabalin. However, we found a direct impact of pregabalin on neurons during CNS inflammation as it reversed the pathological elevation of neuronal intracellular Ca2+ levels in EAE lesions. CONCLUSION: The presented data suggest that pregabalin primarily acts on neuronal Ca2+ channel trafficking thereby reducing Ca2+-mediated cytotoxicity and neuronal damage in an animal model of MS. Future clinical trials need to assess the benefit for neuronal survival by expanding the indication for pregabalin administration to MS patients in further disease phases.

5.
Mult Scler ; 24(4): 432-439, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29512406

RESUMO

Multiple sclerosis (MS) is a common neurological disorder of putative autoimmune origin. Clinical studies delineate abnormal expression of specific cytokines over the course of disease. Preclinical studies using animal models of MS have yielded promising results in manipulating the activity of certain cytokines to improve the clinical outcome. However, the translation of these findings into the clinic is often disappointing. The reason for this might be the complex nature of cytokine networks and the pathogenesis of neuroinflammation, as well as an oversimplified interpretation of preclinical observations. This review presents an overview on cytokines that potentially contribute to the development of MS and provides examples of success and failure in translating basic science into clinical benefit for people with MS.


Assuntos
Doenças Autoimunes/imunologia , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/metabolismo , Animais , Doenças Autoimunes/metabolismo , Citocinas/imunologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Humanos , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Pesquisa
7.
Ann Neurol ; 80(6): 946-951, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27774643

RESUMO

Animal models have implicated an integral role for coagulation factors in neuroinflammatory diseases such as multiple sclerosis (MS) beyond their role in hemostasis. However, their relevance in humans requires further elucidation. This study aimed to determine whether levels of coagulation factors differ between patients with neuroimmunological disorders and respective controls. Individuals suffering from relapsing-remitting and secondary progressive MS had significantly higher prothrombin and factor X levels than healthy donors, whereas levels were unchanged in primary progressive MS and neuromyelitis optica patients. Our study demonstrates that coagulation factors may be key mediators in neuroinflammation and may therefore provide future targets for therapeutic strategies. Ann Neurol 2016;80:946-951.


Assuntos
Fator X/metabolismo , Esclerose Múltipla Crônica Progressiva/sangue , Esclerose Múltipla Recidivante-Remitente/sangue , Protrombina/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Fatores de Coagulação Sanguínea/metabolismo , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neuromielite Óptica/sangue , Adulto Jovem
8.
Blood ; 126(12): 1483-93, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26232171

RESUMO

After tissue injury, both wound sealing and apoptosis contribute to restoration of tissue integrity and functionality. Although the role of platelets (PLTs) for wound closure and induction of regenerative processes is well established, the knowledge about their contribution to apoptosis is incomplete. Here, we show that PLTs present the death receptor Fas ligand (FasL) on their surface after activation. Activated PLTs as well as the isolated membrane fraction of activated PLTs but not of resting PLTs induced apoptosis in a dose-dependent manner in primary murine neuronal cells, human neuroblastoma cells, and mouse embryonic fibroblasts. Membrane protein from PLTs lacking membrane-bound FasL (FasL(△m/△m)) failed to induce apoptosis. Bax/Bak-mediated mitochondrial apoptosis signaling in target cells was not required for PLT-induced cell death, but increased the apoptotic response to PLT-induced Fas signaling. In vivo, PLT depletion significantly reduced apoptosis in a stroke model and an inflammation-independent model of N-methyl-d-aspartic acid-induced retinal apoptosis. Furthermore, experiments using PLT-specific PF4Cre(+) FasL(fl/fl) mice demonstrated a role of PLT-derived FasL for tissue apoptosis. Because apoptosis secondary to injury prevents inflammation, our findings describe a novel mechanism on how PLTs contribute to tissue homeostasis.


Assuntos
Apoptose , Plaquetas/imunologia , Proteína Ligante Fas/imunologia , Neurônios/citologia , Ativação Plaquetária , Animais , Plaquetas/citologia , Plaquetas/patologia , Células Cultivadas , Humanos , Inflamação/imunologia , Inflamação/patologia , Camundongos Endogâmicos C57BL , Neurônios/imunologia , Neurônios/patologia , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia
9.
Brain Behav Immun ; 59: 103-117, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27569659

RESUMO

Myelin loss is a severe pathological hallmark common to a number of neurodegenerative diseases, including multiple sclerosis (MS). Demyelination in the central nervous system appears in the form of lesions affecting both white and gray matter structures. The functional consequences of demyelination on neuronal network and brain function are not well understood. Current therapeutic strategies for ameliorating the course of such diseases usually focus on promoting remyelination, but the effectiveness of these approaches strongly depends on the timing in relation to the disease state. In this study, we sought to characterize the time course of sensory and behavioral alterations induced by de- and remyelination to establish a rational for the use of remyelination strategies. By taking advantage of animal models of general and focal demyelination, we tested the consequences of myelin loss on the functionality of the auditory thalamocortical system: a well-studied neuronal network consisting of both white and gray matter regions. We found that general demyelination was associated with a permanent loss of the tonotopic cortical organization in vivo, and the inability to induce tone-frequency-dependent conditioned behaviors, a status persisting after remyelination. Targeted, focal lysolecithin-induced lesions in the white matter fiber tract, but not in the gray matter regions of cortex, were fully reversible at the morphological, functional and behavioral level. These findings indicate that remyelination of white and gray matter lesions have a different functional regeneration potential, with the white matter being able to regain full functionality while cortical gray matter lesions suffer from permanently altered network function. Therefore therapeutic interventions aiming for remyelination have to consider both region- and time-dependent strategies.


Assuntos
Córtex Cerebral/fisiopatologia , Doenças Desmielinizantes/fisiopatologia , Rede Nervosa/fisiopatologia , Imunidade Adaptativa , Animais , Comportamento Animal , Cuprizona , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/psicologia , Eletrodos Implantados , Substância Cinzenta/patologia , Lisofosfatidilcolinas , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/patologia , Recuperação de Função Fisiológica , Sensação , Substância Branca/patologia
10.
J Neuroinflammation ; 13(1): 160, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27334140

RESUMO

BACKGROUND: Demyelination and remyelination are common pathological processes in many neurological disorders, including multiple sclerosis (MS). Clinical evidence suggests extensive involvement of the thalamocortical (TC) system in patients suffering from MS. METHODS: Using murine brain slices of the primary auditory cortex, we investigated the functional consequences of cuprizone-induced de- and remyelination on neuronal activity and auditory TC synaptic transmission in vitro. RESULTS: Our results revealed an impact of myelin loss and restoration on intrinsic cellular firing patterns, synaptic transmission, and neuronal plasticity in layer 3 and 4 neurons of the auditory TC network. While there was a complex hyper- and depolarizing shift of the resting membrane potential, spontaneous and induced action potential firing was reduced during demyelination and early remyelination. In addition, excitatory postsynaptic potential amplitudes were decreased and induction of LTP was reduced during demyelination. CONCLUSIONS: These data indicate that demyelination-induced impairment of neurons and network activity within the TC system may underlie clinical symptoms observed in demyelinating diseases, corroborating human findings that disease progression is significantly correlated with microstructural tissue damage of the TC system. Further investigation into focal inflammation-induced demyelination models ex vivo and in vivo are needed to understand the functional implication of local and remote lesion formation on TC network activity in MS.


Assuntos
Córtex Auditivo/patologia , Vias Auditivas/efeitos dos fármacos , Cuprizona/toxicidade , Doenças Desmielinizantes/induzido quimicamente , Inibidores da Monoaminoxidase/toxicidade , Tálamo/patologia , Potenciais de Ação/efeitos dos fármacos , Animais , Vias Auditivas/fisiopatologia , Biofísica , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Estimulação Elétrica , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Proteína Proteolipídica de Mielina , Neurônios/efeitos dos fármacos , Neurônios/patologia , Técnicas de Patch-Clamp , Potenciais Sinápticos/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Fatores de Tempo
11.
Ann Neurol ; 77(5): 784-803, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25628066

RESUMO

OBJECTIVE: Recent evidence suggests that ischemic stroke is a thromboinflammatory disease. Plasma kallikrein (PK) cleaves high-molecular-weight kininogen to release bradykinin (BK) and is a key constituent of the proinflammatory contact-kinin system. In addition, PK can activate coagulation factor XII, the origin of the intrinsic coagulation cascade. Thus, PK triggers 2 important pathological pathways of stroke formation, thrombosis and inflammation. METHODS: We investigated the consequences of PK inhibition in transient and permanent models of ischemic stroke. RESULTS: PK-deficient mice of either sex challenged with transient middle cerebral artery occlusion developed significantly smaller brain infarctions and less severe neurological deficits compared with controls without an increase in infarct-associated hemorrhage. This protective effect was preserved at later stages of infarctions as well as after permanent stroke. Reduced intracerebral thrombosis and improved cerebral blood flow could be identified as underlying mechanisms. Moreover, blood-brain barrier function was maintained in mice lacking PK, and the local inflammatory response was reduced. PK-deficient mice reconstituted with PK or BK again developed brain infarctions similar to wild-type mice. Important from a translational perspective, inhibition of PK in wild-type mice using a PK-specific antibody was likewise effective even when performed in a therapeutic setting up to 3 hours poststroke. INTERPRETATION: PK drives thrombus formation and inflammation via activation of the intrinsic coagulation cascade and the release of BK but appears to be dispensable for hemostasis. Hence, PK inhibition may offer a safe strategy to combat thromboembolic disorders including ischemic stroke.


Assuntos
Calicreína Plasmática/metabolismo , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/prevenção & controle , Trombose/sangue , Trombose/prevenção & controle , Animais , Infarto Encefálico/sangue , Infarto Encefálico/genética , Infarto Encefálico/prevenção & controle , Feminino , Inflamação/sangue , Inflamação/genética , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/genética , Acidente Vascular Cerebral/genética , Trombose/genética
12.
Int J Mol Sci ; 17(10)2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27754414

RESUMO

Beyond their indispensable role in hemostasis, platelets have shown to affect the development of inflammatory disorders, as they have been epidemiologically and mechanistically linked to diseases featuring an inflammatory reaction in inflammatory diseases like multiple sclerosis, rheumatoid arthritis and inflammatory bowel disorders. The identification of novel molecular mechanisms linking inflammation and to platelets has highlighted them as new targets for therapeutic interventions. In particular, genetic and pharmacological studies have identified an important role for platelets in neuroinflammation. This review summarizes the main molecular links between platelets and inflammation, focusing on immune regulatory factors, receptors, cellular targets and signaling pathways by which they can amplify inflammatory reactions and that make them potential therapeutic targets.


Assuntos
Doenças Autoimunes/imunologia , Plaquetas/imunologia , Plaquetas/patologia , Inflamação/imunologia , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Doenças Autoimunes/patologia , Humanos , Inflamação/patologia , Doenças Inflamatórias Intestinais/imunologia , Doenças Inflamatórias Intestinais/patologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia
13.
Pflugers Arch ; 467(5): 973-87, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25315980

RESUMO

Potassium channels can fulfill both beneficial and detrimental roles in neuronal damage during ischemic stroke. Earlier studies have characterized a neuroprotective role of the two-pore domain potassium channels KCNK2 (TREK1) and KCNK3 (TASK1). Protective neuronal hyperpolarization and prevention of intracellular Ca(2+) overload and glutamate excitotoxicity were suggested to be the underlying mechanisms. We here identify an unexpected role for the related KCNK5 channel in a mouse model of transient middle cerebral artery occlusion (tMCAO). KCNK5 is strongly upregulated on neurons upon cerebral ischemia, where it is most likely involved in the induction of neuronal apoptosis. Hypoxic conditions elevated neuronal expression levels of KCNK5 in acute brain slices and primary isolated neuronal cell cultures. In agreement, KCNK5 knockout mice had significantly reduced infarct volumes and improved neurologic function 24 h after 60 min of tMCAO and this protective effect was preserved at later stages of infarct development. KCNK5 deficiency resulted in a significantly reduced number of apoptotic neurons, a downregulation of pro-apoptotic and upregulation of anti-apoptotic factors. Results of adoptive transfer experiments of wild-type and Kcnk5 (-/-) immune cells into Rag1 (-/-) mice prior to tMCAO exclude a major role of KCNK5 in poststroke inflammatory reactions. In summary, KCNK5 expression is induced on neurons under ischemic conditions where it most likely exerts pro-apoptotic effects. Hence, pharmacological blockade of KCNK5 might have therapeutic potential in preventing ischemic neurodegeneration.


Assuntos
Isquemia Encefálica/metabolismo , Neurônios/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Humanos , Infarto da Artéria Cerebral Média/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia
14.
Eur J Immunol ; 44(8): 2295-305, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24811005

RESUMO

Lymphocyte adhesion and subsequent trafficking across endothelial barriers are essential steps in various immune-mediated disorders of the CNS, including MS. The molecular mechanisms underlying these processes, however, are still unknown. Phospholipase D1 (PLD1), an enzyme that generates phosphatidic acid through hydrolysis of phosphatidylcholine and additionally yields choline as a product, has been described as regulator of the cell mobility. By using PLD1-deficient mice, we investigated the functional significance of PLD1 for lymphocyte adhesion and migration in vitro and after myelin oligodendrocyte glycoprotein (MOG)35-55 -induced EAE, a model of human MS. The lack of PLD1 reduced chemokine-mediated static adhesion of lymphocytes to the endothelial adhesion molecules vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) in vitro, and was accompanied by a decreased migratory capacity in both blood brain barrier and cell migration models. Importantly, PLD1 is also relevant for the recruitment of immune cells into the CNS in vivo since disease severity after EAE was significantly attenuated in PLD1-deficient mice. Furthermore, PLD1 expression could be detected on lymphocytes in MS patients. Our findings suggest a critical function of PLD1-dependent intracellular signaling cascades in regulating lymphocyte trafficking during autoimmune CNS inflammation.


Assuntos
Adesão Celular/imunologia , Movimento Celular/imunologia , Encefalomielite Autoimune Experimental/imunologia , Linfócitos/imunologia , Fosfolipase D/imunologia , Animais , Barreira Hematoencefálica/imunologia , Encefalomielite Autoimune Experimental/patologia , Células Endoteliais/imunologia , Feminino , Inflamação/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Linfócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Molécula 1 de Adesão de Célula Vascular/imunologia
15.
Am J Pathol ; 184(7): 2056-65, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24819960

RESUMO

Glatiramer acetate (GA) (Copaxone), a well-established drug for the treatment of multiple sclerosis, is believed to modulate numerous pathways including antigen-presenting cells or cytokine responses. A new generation of spontaneous experimental autoimmune encephalomyelitis mouse models has been developed that mimic certain aspects of multiple sclerosis spectrum disorders. We assessed the effects of GA in the opticospinal encephalomyelitis model, which involves MOG35-55 peptide-specific T cells and B cells. A nonsignificant trend toward lower disease incidence was found for GA treatment (started on postnatal day 20). Immunohistochemical evaluations revealed no significant differences for inflammatory lesions and demyelination, cytokine production, proliferation, and cell surface markers of immune cells between GA-treated and PBS-treated (control) mice. Although a good correlation was found between the disease score of individual mice and some readout parameters (eg, immunohistochemical staining), this was not the case for others (eg, IFN-γ production). It seems plausible that a major effect of GA lies on alternative immunological pathways, such as initiating of an immune response that is not sufficiently reflected in this spontaneous experimental autoimmune encephalomyelitis model. Thus, the main advantage of the opticospinal encephalomyelitis model in our hands lies in the elucidation of factors influencing the onset of experimental autoimmune encephalomyelitis (eg, susceptibility factors). The model seems less suitable for investigation of disease severity modifications after therapeutic interventions.


Assuntos
Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/tratamento farmacológico , Peptídeos/farmacologia , Animais , Linfócitos B/imunologia , Encefalomielite Autoimune Experimental/imunologia , Acetato de Glatiramer , Camundongos Transgênicos , Linfócitos T/imunologia
16.
Blood ; 121(4): 679-91, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23160472

RESUMO

We have recently identified T cells as important mediators of ischemic brain damage, but the contribution of the different T-cell subsets is unclear. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) are generally regarded as prototypic anti-inflammatory cells that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. In the present study, we examined the role of Tregs after experimental brain ischemia/reperfusion injury. Selective depletion of Tregs in the DEREG mouse model dramatically reduced infarct size and improved neurologic function 24 hours after stroke and this protective effect was preserved at later stages of infarct development. The specificity of this detrimental Treg effect was confirmed by adoptive transfer experiments in wild-type mice and in Rag1(-/-) mice lacking lymphocytes. Mechanistically, Tregs induced microvascular dysfunction in vivo by increased interaction with the ischemic brain endothelium via the LFA-1/ICAM-1 pathway and platelets and these findings were confirmed in vitro. Ablation of Tregs reduced microvascular thrombus formation and improved cerebral reperfusion on stroke, as revealed by ultra-high-field magnetic resonance imaging at 17.6 Tesla. In contrast, established immunoregulatory characteristics of Tregs had no functional relevance. We define herein a novel and unexpected role of Tregs in a primary nonimmunologic disease state.


Assuntos
Isquemia Encefálica/imunologia , Microvasos/fisiopatologia , Acidente Vascular Cerebral/metabolismo , Linfócitos T Reguladores/metabolismo , Transferência Adotiva , Animais , Plaquetas/imunologia , Plaquetas/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Comunicação Celular , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Depleção Linfocítica , Masculino , Camundongos , Camundongos Knockout , Microvasos/patologia , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/terapia , Linfócitos T Reguladores/imunologia
18.
Int J Mol Sci ; 16(8): 16880-96, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26213925

RESUMO

Lymphocytes express potassium channels that regulate physiological cell functions, such as activation, proliferation and migration. Expression levels of K2P5.1 (TASK2; KCNK5) channels belonging to the family of two-pore domain potassium channels have previously been correlated to the activity of autoreactive T lymphocytes in patients with multiple sclerosis and rheumatoid arthritis. In humans, K2P5.1 channels are upregulated upon T cell stimulation and influence T cell effector functions. However, a further clinical translation of targeting K2P5.1 is currently hampered by a lack of highly selective inhibitors, making it necessary to evaluate the impact of KCNK5 in established preclinical animal disease models. We here demonstrate that K2P5.1 knockout (K2P5.1-/-) mice display no significant alterations concerning T cell cytokine production, proliferation rates, surface marker molecules or signaling pathways. In an experimental model of autoimmune neuroinflammation, K2P5.1-/- mice show a comparable disease course to wild-type animals and no major changes in the peripheral immune system or CNS compartment. A compensatory upregulation of the potassium channels K2P3.1 and KV1.3 seems to counterbalance the deletion of K2P5.1. As an alternative model mimicking autoimmune neuroinflammation, experimental autoimmune encephalomyelitis in the common marmoset has been proposed, especially for testing the efficacy of new potential drugs. Initial experiments show that K2P5.1 is functionally expressed on marmoset T lymphocytes, opening up the possibility for assessing future K2P5.1-targeting drugs.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Canal de Potássio Kv1.3/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Animais , Callithrix , Modelos Animais de Doenças , Progressão da Doença , Deleção de Genes , Sistema Imunitário/patologia , Camundongos , Camundongos Knockout , Fenótipo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Regulação para Cima
19.
Ann Neurol ; 73(3): 419-29, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23424019

RESUMO

OBJECTIVE: The outbreak of hemolytic-uremic syndrome and diarrhea caused by Shiga toxin-producing Escherichia coli O104:H4 in Germany during May to July 2011 involved severe and characteristic neurologic manifestations with a strong female preponderance. Owing to these observations, we designed a series of experimental studies to evaluate the underlying mechanism of action of this clinical picture. METHODS: A magnetic resonance imaging and electroencephalographic study of patients was performed to evaluate the clinical picture in detail. Thereafter, combinations of different experimental settings, including electrophysiological and histological analyses, as well as calcium imaging in brain slices of rats, were conducted. RESULTS: We report on 7 female patients with neurologic symptoms and signs including bilateral thalamic lesions and encephalopathic changes indicative of a predominant involvement of the thalamus. Experimental studies in rats revealed an enhanced expression of the Shiga toxin receptor globotriaosylceramide on thalamic neurons in female rats as compared to other brain regions in the same rats and to male animals. Incubation of brain slices with Shiga toxin 2 evoked a strong membrane depolarization and intracellular calcium accumulation in neurons, associated with neuronal apoptosis, predominantly in the thalamic area. INTERPRETATION: These findings suggest that the direct cytotoxic effect of Shiga toxin 2 in the thalamus might contribute to the pathophysiology of neuronal complications in hemolytic-uremic syndrome.


Assuntos
Infecções por Escherichia coli/complicações , Doenças do Sistema Nervoso/etiologia , Doenças do Sistema Nervoso/patologia , Toxina Shiga II/toxicidade , Tálamo/patologia , Adulto , Idoso , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Cálcio/metabolismo , Eletroencefalografia , Infecções por Escherichia coli/líquido cefalorraquidiano , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas In Vitro , Imageamento por Ressonância Magnética , Potenciais da Membrana/efeitos dos fármacos , Doenças do Sistema Nervoso/líquido cefalorraquidiano , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , RNA Mensageiro/metabolismo , Ratos , Ratos Long-Evans , Caracteres Sexuais , Tálamo/fisiopatologia , Triexosilceramidas/metabolismo , Adulto Jovem
20.
Blood ; 120(19): 4082-92, 2012 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22936662

RESUMO

Thrombosis and inflammation are hallmarks of ischemic stroke still unamenable to therapeutic interventions. High-molecular-weight kininogen (KNG) is a central constituent of the contact-kinin system which represents an interface between thrombotic and inflammatory circuits and is critically involved in stroke development. Kng(-/-) mice are protected from thrombosis after artificial vessel wall injury and lack the proinflammatory mediator bradykinin. We investigated the consequences of KNG deficiency in models of ischemic stroke. Kng(-/-) mice of either sex subjected to transient middle cerebral artery occlusion developed dramatically smaller brain infarctions and less severe neurologic deficits without an increase in infarct-associated hemorrhage. This protective effect was preserved at later stages of infarction as well as in elderly mice. Targeting KNG reduced thrombus formation in ischemic vessels and improved cerebral blood flow, and reconstitution of KNG-deficient mice with human KNG or bradykinin restored clot deposition and infarct susceptibility. Moreover, mice deficient in KNG showed less severe blood-brain barrier damage and edema formation, and the local inflammatory response was reduced compared with controls. Because KNG appears to be instrumental in pathologic thrombus formation and inflammation but dispensable for hemostasis, KNG inhibition may offer a selective and safe strategy for combating stroke and other thromboembolic diseases.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Isquemia Encefálica/prevenção & controle , Cininogênios/deficiência , Trombose/fisiopatologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Edema Encefálico/genética , Edema Encefálico/prevenção & controle , Isquemia Encefálica/genética , Isquemia Encefálica/mortalidade , Modelos Animais de Doenças , Feminino , Inflamação/genética , Inflamação/patologia , Hemorragias Intracranianas/diagnóstico , Cininogênios/genética , Cininogênios/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Knockout , Fluxo Sanguíneo Regional , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/mortalidade , Acidente Vascular Cerebral/prevenção & controle , Trombose/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa