Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 101: 194-210, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35032575

RESUMO

BACKGROUND: Antigen-specific neuroinflammation and neurodegeneration are characteristic for neuroimmunological diseases. In Parkinson's disease (PD) pathogenesis, α-synuclein is a known culprit. Evidence for α-synuclein-specific T cell responses was recently obtained in PD. Still, a causative link between these α-synuclein responses and dopaminergic neurodegeneration had been lacking. We thus addressed the functional relevance of α-synuclein-specific immune responses in PD in a mouse model. METHODS: We utilized a mouse model of PD in which an Adeno-associated Vector 1/2 serotype (AAV1/2) expressing human mutated A53T-α-Synuclein was stereotactically injected into the substantia nigra (SN) of either wildtype C57BL/6 or Recombination-activating gene 1 (RAG1)-/- mice. Brain, spleen, and lymph node tissues from different time points following injection were then analyzed via FACS, cytokine bead assay, immunohistochemistry and RNA-sequencing to determine the role of T cells and inflammation in this model. Bone marrow transfer from either CD4+/CD8-, CD4-/CD8+, or CD4+/CD8+ (JHD-/-) mice into the RAG-1-/- mice was also employed. In addition to the in vivo studies, a newly developed A53T-α-synuclein-expressing neuronal cell culture/immune cell assay was utilized. RESULTS: AAV-based overexpression of pathogenic human A53T-α-synuclein in dopaminergic neurons of the SN stimulated T cell infiltration. RNA-sequencing of immune cells from PD mouse brains confirmed a pro-inflammatory gene profile. T cell responses were directed against A53T-α-synuclein-peptides in the vicinity of position 53 (68-78) and surrounding the pathogenically relevant S129 (120-134). T cells were required for α-synuclein-induced neurodegeneration in vivo and in vitro, while B cell deficiency did not protect from dopaminergic neurodegeneration. CONCLUSIONS: Using T cell and/or B cell deficient mice and a newly developed A53T-α-synuclein-expressing neuronal cell culture/immune cell assay, we confirmed in vivo and in vitro that pathogenic α-synuclein peptide-specific T cell responses can cause dopaminergic neurodegeneration and thereby contribute to PD-like pathology.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Animais , Modelos Animais de Doenças , Dopamina , Neurônios Dopaminérgicos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doença de Parkinson/patologia , RNA , Substância Negra/metabolismo , Linfócitos T/metabolismo , alfa-Sinucleína/metabolismo
2.
Exp Neurol ; 350: 113978, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35026227

RESUMO

Deep Brain Stimulation (DBS) is an efficacious treatment option for an increasing range of brain disorders. To enhance our knowledge about the mechanisms of action of DBS and to probe novel targets, basic research in animal models with DBS is an essential research base. Beyond nonhuman primate, pig, and mouse models, the rat is a widely used animal model for probing DBS effects in basic research. Reconstructing DBS electrode placement after surgery is crucial to associate observed effects with modulating a specific target structure. Post-mortem histology is a commonly used method for reconstructing the electrode location. In humans, however, neuroimaging-based electrode localizations have become established. For this reason, we adapt the open-source software pipeline Lead-DBS for DBS electrode localizations from humans to the rat model. We validate our localization results by inter-rater concordance and a comparison with the conventional histological method. Finally, using the open-source software pipeline OSS-DBS, we demonstrate the subject-specific simulation of the VTA and the activation of axon models aligned to pathways representing neuronal fibers, also known as the pathway activation model. Both activation models yield a characterization of the impact of DBS on the target area. Our results suggest that the proposed neuroimaging-based method can precisely localize DBS electrode placements that are essentially rater-independent and yield results comparable to the histological gold standard. The advantages of neuroimaging-based electrode localizations are the possibility of acquiring them in vivo and combining electrode reconstructions with advanced imaging metrics, such as those obtained from diffusion or functional magnetic resonance imaging (MRI). This paper introduces a freely available open-source pipeline for DBS electrode reconstructions in rats. The presented initial validation results are promising.


Assuntos
Estimulação Encefálica Profunda , Modelos Neurológicos , Animais , Axônios , Eletrodos Implantados , Imageamento por Ressonância Magnética , Masculino , Modelos Animais , Neuroimagem , Ratos , Reprodutibilidade dos Testes , Software , Área Tegmentar Ventral/diagnóstico por imagem
3.
J Vis Exp ; (163)2020 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-32986028

RESUMO

Genetically modified mouse models face limitations, especially when studying movement disorders, where most of the available transgenic rodent models do not present a motor phenotype resembling the clinical aspects of the human disease. Pharmacological mouse models allow for a more direct study of the pathomechanisms and their effect on the behavioral phenotype. Osmotic pumps connected to brain cannulas open up the possibility of creating pharmacological mouse models via local and chronic drug delivery. For the hereditary movement disorder of rapid-onset dystonia-parkinsonism, the loss-of-function mutation in the α3-subunit of the Na+/K+-ATPase can be simulated by a highly specific blockade via the glycoside ouabain. In order to locally block the α3-subunit in the basal ganglia and the cerebellum, which are the two brain structures believed to be heavily involved in the pathogenesis of rapid-onset dystonia-parkinsonism, a bilateral cannula is stereotaxically implanted into the striatum and an additional single cannula is introduced into the cerebellum. The cannulas are connected via vinyl tubing to two osmotic pumps, which are subcutaneously implanted on the back of the animals and allow for the chronic and precise delivery of ouabain. The pharmacological mouse model for rapid-onset dystonia-parkinsonism carries the additional advantage of recapitulating the clinical and pathological features of asymptomatic and symptomatic mutation carriers. Just like mutation carriers of rapid-onset dystonia parkinsonism, the ouabain-perfused mice develop dystonia-like movements only after additional exposure to stress. We demonstrate a mild stress paradigm and introduce two modified scoring systems for the assessment of a motor phenotype.


Assuntos
Distúrbios Distônicos/patologia , Bombas de Infusão , ATPase Trocadora de Sódio-Potássio/genética , Estresse Fisiológico , Animais , Modelos Animais de Doenças , Distúrbios Distônicos/fisiopatologia , Elevação dos Membros Posteriores , Masculino , Camundongos Endogâmicos C57BL , Atividade Motora , Movimento , Mutação/genética , Osmose
4.
J Neural Transm (Vienna) ; 126(7): 905-912, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30725186

RESUMO

Delirium is an acute and fluctuating disturbance of attention and awareness. Pre-existing cognitive disturbances or dementia are the most significant risk factors for developing delirium and precipitating factors such as drug treatment, infections, trauma, or surgery may trigger delirium. Patients with Parkinson's disease (PD) are at an increased risk for delirium which may be underdiagnosed due to phenomenological overlap between delirium and chronic neuropsychiatric features of PD or side effects of dopaminergic medication. Prognosis of delirium is detrimental in many cases including permanent cognitive decline, motor impairment, and increased mortality. Management of delirium comprises of pharmacological and non-pharmacological measures. Pharmacotherapy is aimed at treating medical precipitating factors such as infections, pain, and sleep deprivation. Adjustments of anti-parkinsonian medication are recommended to prevent or treat delirium, but no hard evidence in this respect is available from controlled studies. Administration of neuroleptics and other psychoactive drugs in the treatment of delirium is controversially discussed and should be reserved for patients with severe agitation or distressing psychosis. Non-pharmacological interventions to prevent or palliate delirium are based on withdrawing precipitating or distressing factors, and to provide sensory, emotional and environmental support. Appropriate instruments to detect and assess delirium in PD are needed, and efforts are warranted to improve understanding and treatment of this severe and common disorder.


Assuntos
Delírio/etiologia , Doença de Parkinson/complicações , Doença de Parkinson/psicologia , Humanos
5.
Ann Neurol ; 81(6): 825-836, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28470693

RESUMO

OBJECTIVE: Deep brain stimulation (DBS) of the subthalamic nucleus (STN) is a highly effective symptomatic therapy for motor deficits in Parkinson's disease (PD). An additional, disease-modifying effect has been suspected from studies in toxin-based PD animal models, but these models do not reflect the molecular pathology and progressive nature of PD that would be required to evaluate a disease-modifying action. Defining a disease-modifying effect could radically change the way in which DBS is used in PD. METHODS: We applied STN-DBS in an adeno-associated virus (AAV) 1/2-driven human mutated A53T α-synuclein (aSyn)-overexpressing PD rat model (AAV1/2-A53T-aSyn). Rats were injected unilaterally, in the substantia nigra (SN), with AAV1/2-A53T-aSyn or control vector. Three weeks later, after behavioral and nigrostriatal dopaminergic deficits had developed, rats underwent STN-DBS electrode implantation ipsilateral to the vector-injected SN. Stimulation lasted for 3 weeks. Control groups remained OFF stimulation. Animals were sacrificed at 6 weeks. RESULTS: Motor performance in the single pellet reaching task was impaired in the AAV1/2-A53T-aSyn-injected stim-OFF group, 6 weeks after AAV1/2-A53T-aSyn injection, compared to preoperative levels (-82%; p < 0.01). Deficits were reversed in AAV1/2-A53T-aSyn, stim-ON rats after 3 weeks of active stimulation, compared to the AAV1/2-A53T-aSyn stim-OFF rats (an increase of ∼400%; p < 0.05), demonstrating a beneficial effect of DBS. This motor improvement was maintained when the stimulation was turned off and was accompanied by a higher number of tyrosine hydroxylase+ SN neurons (increase of ∼29%), compared to AAV1/2-A53T-aSyn stim-OFF rats (p < 0.05). INTERPRETATION: Our data support the putative neuroprotective and disease-modifying effect of STN-DBS in a mechanistically relevant model of PD. Ann Neurol 2017;81:825-836.


Assuntos
Estimulação Encefálica Profunda/métodos , Doença de Parkinson/terapia , Núcleo Subtalâmico , alfa-Sinucleína/administração & dosagem , Animais , Comportamento Animal , Dependovirus , Modelos Animais de Doenças , Vetores Genéticos , Humanos , Masculino , Mutação , Ratos , Ratos Sprague-Dawley , Núcleo Subtalâmico/citologia , Núcleo Subtalâmico/metabolismo , Núcleo Subtalâmico/fisiopatologia
6.
Acta Neuropathol Commun ; 5(1): 11, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-28143577

RESUMO

α-Synuclein is a protein implicated in the etiopathogenesis of Parkinson's disease (PD). AAV1/2-driven overexpression of human mutated A53T-α-synuclein in rat and monkey substantia nigra (SN) induces degeneration of nigral dopaminergic neurons and decreases striatal dopamine and tyrosine hydroxylase (TH). Given certain advantages of the mouse, especially it being amendable to genetic manipulation, translating the AAV1/2-A53T α-synuclein model to mice would be of significant value. AAV1/2-A53T α-synuclein or AAV1/2 empty vector (EV) at a concentration of 5.16 x 1012 gp/ml were unilaterally injected into the right SN of male adult C57BL/6 mice. Post-mortem examinations included immunohistochemistry to analyze nigral α-synuclein, Ser129 phosphorylated α-synuclein and TH expression, striatal dopamine transporter (DAT) levels by autoradiography and dopamine levels by high performance liquid chromatography. At 10 weeks, in AAV1/2-A53T α-synuclein mice there was a 33% reduction in TH+ dopaminergic nigral neurons (P < 0.001), 29% deficit in striatal DAT binding (P < 0.05), 38% and 33% reductions in dopamine (P < 0.001) and DOPAC (P < 0.01) levels and a 60% increase in dopamine turnover (homovanilic acid/dopamine ratio; P < 0.001). Immunofluorescence showed that the AAV1/2-A53T α-synuclein injected mice had widespread nigral and striatal expression of vector-delivered A53T-α-synuclein. Concurrent staining with human PD SN samples using gold standard histological methodology for Lewy pathology detection by proteinase K digestion and application of specific antibody raised against human Lewy body α-synuclein (LB509) and Ser129 phosphorylated α-synuclein (81A) revealed insoluble α-synuclein aggregates in AAV1/2-A53T α-synuclein mice resembling Lewy-like neurites and bodies. In the cylinder test, we observed significant paw use asymmetry in the AAV1/2-A53T α-synuclein group when compared to EV controls at 5 and 9 weeks post injection (P < 0.001; P < 0.05). These data show that unilateral injection of AAV1/2-A53T α-synuclein into the mouse SN leads to persistent motor deficits, neurodegeneration of the nigrostriatal dopaminergic system and development of Lewy-like pathology, thereby reflecting clinical and pathological hallmarks of human PD.


Assuntos
Atividade Motora/fisiologia , Mutação , Transtornos Parkinsonianos/metabolismo , Substância Negra/metabolismo , alfa-Sinucleína/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Dependovirus/genética , Dopamina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Vetores Genéticos , Ácido Homovanílico/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , Transtornos Parkinsonianos/patologia , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/genética
7.
J Neural Transm (Vienna) ; 122(12): 1633-43, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26290125

RESUMO

Neuroinflammation is a well-known neuropathological feature of Parkinson's disease (PD), but it remains controversial whether it is causal or consequential to neurodegeneration. While the role of microglia in the pathogenesis has been thoroughly investigated in human and different rodent models, data concerning the impact of the adaptive immune system on the pathogenesis of PD are still rare, although lymphocyte populations were found in brain tissue of PD patients and have been implicated in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mediated neurodegeneration in mice. To test the hypothesis that the adaptive immune system contributes to the progression of PD in the murine 6-hydroxydopamine (6-OHDA) model, we performed unilateral 6-OHDA injection into the medial forebrain bundle and compared wild-type mice with recombination activating gene-1 deficient mice (RAG-1(-/-)), that lack mature lymphocytes. After 6-OHDA injection, immune-deficient mice moved significantly slower and less often than wild-type mice. Rotarod analysis displayed a shorter latency to fall in RAG-1(-/-) mice. Immunohistochemical analysis in wild-type mice demonstrated a higher CD8+ T cell density in the ipsilesional striatum compared to sham-operated animals. Cell counts of tyrosine hydroxylase positive dopaminergic neurons of the substantia nigra in immune compromised mice were significantly reduced compared to wild-type mice. Wild type bone marrow reconstitution into RAG-1(-/-) recipients rescued the clinical deterioration as well as the neurodegeneration in RAG-1(-/-) deficient recipients ameliorated clinical symptoms and neurodegeneration after 6-OHDA treatment. Our data indicate that lymphocytes reduce the clinical and neuropathological impact of 6-OHDA lesioning and thus may play a protective role in this toxic mouse model of PD.


Assuntos
Corpo Estriado/fisiopatologia , Linfócitos/fisiologia , Transtornos Parkinsonianos/fisiopatologia , Substância Negra/fisiopatologia , Animais , Transplante de Medula Óssea , Corpo Estriado/patologia , Progressão da Doença , Neurônios Dopaminérgicos/patologia , Neurônios Dopaminérgicos/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Linfócitos/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , Oxidopamina , Transtornos Parkinsonianos/patologia , Teste de Desempenho do Rota-Rod , Substância Negra/patologia , Tirosina 3-Mono-Oxigenase/metabolismo
8.
PLoS One ; 7(8): e42554, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22905147

RESUMO

Mice overexpressing proteolipid protein (PLP) develop a leukodystrophy-like disease involving cytotoxic, CD8+ T-lymphocytes. Here we show that these cytotoxic T-lymphocytes perturb retrograde axonal transport. Using fluorogold stereotactically injected into the colliculus superior, we found that PLP overexpression in oligodendrocytes led to significantly reduced retrograde axonal transport in retina ganglion cell axons. We also observed an accumulation of mitochondria in the juxtaparanodal axonal swellings, indicative for a disturbed axonal transport. PLP overexpression in the absence of T-lymphocytes rescued retrograde axonal transport defects and abolished axonal swellings. Bone marrow transfer from wildtype mice, but not from perforin- or granzyme B-deficient mutants, into lymphocyte-deficient PLP mutant mice led again to impaired axonal transport and the formation of axonal swellings, which are predominantly located at the juxtaparanodal region. This demonstrates that the adaptive immune system, including cytotoxic T-lymphocytes which release perforin and granzyme B, are necessary to perturb axonal integrity in the PLP-transgenic disease model. Based on our observations, so far not attended molecular and cellular players belonging to the immune system should be considered to understand pathogenesis in inherited myelin disorders with progressive axonal damage.


Assuntos
Axônios/patologia , Inflamação/patologia , Oligodendroglia/citologia , Linfócitos T Citotóxicos/citologia , Animais , Axônios/metabolismo , Transplante de Medula Óssea , Linfócitos T CD8-Positivos/citologia , Granzimas/metabolismo , Heterozigoto , Sistema Imunitário , Camundongos , Camundongos Transgênicos , Mutação , Neurônios/patologia , Proteolipídeos/metabolismo , Células Ganglionares da Retina/citologia
9.
Glia ; 58(4): 469-80, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19780193

RESUMO

Demyelination and death of oligodendrocytes accompanied by transection of neurites and neuronal apoptosis are pathological hallmarks of cortical and subcortical gray matter lesions in demyelinating viral and autoimmune inflammatory CNS disorders. In these disorders, leukocortical lesions, containing the perikarya of most efferent neurons, display pronounced infiltration by CD8(+) T cells of putative specificity for oligodendrocyte- and myelin-related antigens. Hence, neuronal apoptosis in gray matter lesions may be a collateral effect of an oligodendrocyte-directed attack by CD8(+) T cells. To challenge this hypothesis, we transferred activated antigen-specific CD8(+) T cells (OT-I T cells) into acute coronal brain slices from mice selectively expressing ovalbumin as a cytosolic neo-self-antigen in oligodendrocytes (ODC-OVA mice). We studied mechanisms and kinetics of oligodendroglial and neuronal apoptosis in the neocortex and hippocampus, using multicolor staining for different cell types and activated caspase-3. Within the gray matter, a single OT-I T cell caused simultaneous caspase-3 activation in about 30 ODCs and 10 neurons within 6 h in a strictly antigen-dependent manner. Experiments with OT-I T cells genetically deficient for perforin or the granzyme B-cluster and with blocking anti-FasL antibodies as well as proinflammatory cytokines revealed, that collateral apoptosis of neurons was likely due to a spillover of perforin and granzyme(s) from the OT-I T cell itself or the immunological synapse that it selectively formed with antigen-presenting oligodendrocytes. Collateral neuronal apoptosis could contribute to substantial neuronal loss in gray matter lesions and cause persistent neurological impairment in both acute and chronic gray matter lesions in various inflammatory CNS disorders.


Assuntos
Apoptose/fisiologia , Encéfalo/fisiologia , Antígenos CD8/metabolismo , Neurônios/fisiologia , Oligodendroglia/fisiologia , Linfócitos T/fisiologia , Animais , Caspase 3/metabolismo , Movimento Celular , Citocinas/metabolismo , Proteína Ligante Fas/metabolismo , Granzimas/genética , Granzimas/metabolismo , Hipocampo/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/fisiologia , Fibras Nervosas Amielínicas/fisiologia , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Serpinas/metabolismo
10.
Am J Pathol ; 174(6): 2290-9, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19443704

RESUMO

It is assumed that the onset and course of autoimmune inflammatory central nervous system (CNS) disorders (eg, multiple sclerosis) are influenced by factors that afflict immune regulation as well as CNS vulnerability. We challenged this concept experimentally by investigating how genetic alterations that affect myelin (primary oligodendrocyte damage in PLPtg mice) and/or T-cell regulation (deficiency of PD-1) influence both the onset and course of an experimental autoimmune CNS inflammatory disease [MOG(35-55)-induced experimental autoimmune encephalomyelitis (EAE)]. We observed that double pathology was associated with a significantly earlier onset of disease, a slight increase in the neurological score, an increase in the number of infiltrating cells, and enhanced axonal degeneration compared with wild-type mice and the respective, single mutant controls. Double-mutant PLPtg/PD-1(-/-) mice showed an increased production of interferon-gamma by CNS immune cells at the peak of disease. Neither PD-1 deficiency nor oligodendropathy led to detectable spread of antigenic MHC class I- or class II-restricted epitopes during EAE. However, absence of PD-1 clearly increased the propensity of T lymphocytes to expand, and the number of clonal expansions reliably reflected the severity of the EAE disease course. Our data show that the interplay between immune dysregulation and myelinopathy results in a stable exacerbation of actively induced autoimmune CNS inflammation, suggesting that the combination of several pathological issues contributes significantly to disease susceptibility or relapses in human disease.


Assuntos
Antígenos de Diferenciação/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , Animais , Antígenos de Diferenciação/genética , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/genética , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Oligodendroglia/patologia , Receptor de Morte Celular Programada 1 , Linfócitos T/imunologia
11.
PLoS One ; 4(2): e4405, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19197390

RESUMO

We investigated the impact of immune regulatory mechanisms involved in the modulation of the recently presented, CD8+ lymphocyte mediated immune response in a mouse model of oligodendropathy-induced inflammation (PLPtg-mutants). The focus was on the role of the co-inhibitory molecule PD-1, a CD28-related receptor expressed on activated T- and B-lymphocytes associated with immune homeostasis and autoimmunity. PLPtg/PD-1-deficient double mutants and the corresponding bone marrow chimeras were generated and analysed using immunohistochemistry, light- and electron microscopy, with particular emphasis on immune-cell number and neural damage. In addition, the immune cells in both the CNS and the peripheral immune system were investigated by IFN-gamma elispot assays and spectratype analysis. We found that mice with combined pathology exhibited significantly increased numbers of CD4+ and CD8+ T-lymphocytes in the CNS. Lack of PD-1 substantially aggravated the pathological phenotype of the PLPtg mutants compared to genuine PLPtg mutants, whereas the PD-1 deletion alone did not cause alterations in the CNS. CNS T-lymphocytes in PLPtg/PD-1-/- double mutants exhibited massive clonal expansions. Furthermore, PD-1 deficiency was associated with a significantly higher propensity of CNS but not peripheral CD8+ T-cells to secrete proinflammatory cytokines. PD-1 could be identified as a crucial player of tissue homeostasis and immune-mediated damage in a model of oligodendropathy-induced inflammation. Alterations of this regulatory pathway lead to overt neuroinflammation of high pathogenetic impact. Our finding may have implications for understanding the mechanisms leading to the high clinical variability of polygenic or even monogenic disorders of the nervous system.


Assuntos
Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Inflamação/patologia , Neurônios/patologia , Oligodendroglia/patologia , Animais , Transplante de Medula Óssea , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Contagem de Células , Proliferação de Células , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Quimera , Células Clonais , Imuno-Histoquímica , Interferon gama/metabolismo , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Proteína Proteolipídica de Mielina , Neurônios/metabolismo , Receptor de Morte Celular Programada 1
12.
Neurobiol Dis ; 33(2): 207-12, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19028581

RESUMO

Mice hetero- or homozygously deficient for myelin protein zero (P0+/-, P0-/- mice) are models for distinct forms of inherited de- or dysmyelinating neuropathies, respectively. P0+/- mice show a demyelinating neuropathy with a pathogenetic implication of CD8+ T-lymphocytes and macrophages, while P0-/- mice show dysmyelination with axonal loss. It was, therefore, of interest to treat both mutants with FK506 (Tacrolimus), an agent with immunosuppressive and neuroprotective properties. Treatment of P0+/- mice led to an aggravation of demyelination, without affecting nervous CD8+ T-lymphocytes, but reducing splenic CD4+ cells. Treatment of P0-/- mice resulted in a substantial increase of the dysmyelination-related axon loss. Treatment of wildtype mice did not cause pathological changes in peripheral nerves. Our study shows that FK506 may not be suitable for the treatment of the human nerve disorders. Furthermore, when used as an immunosuppressant, the drug may generate detrimental neurological side effects in patients with an additional hereditary neuropathy.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Imunossupressores/toxicidade , Bainha de Mielina/efeitos dos fármacos , Nervos Periféricos/efeitos dos fármacos , Tacrolimo/toxicidade , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Doença de Charcot-Marie-Tooth/tratamento farmacológico , Doença de Charcot-Marie-Tooth/fisiopatologia , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Imuno-Histoquímica , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Proteína P0 da Mielina/genética , Bainha de Mielina/patologia , Condução Nervosa , Nervos Periféricos/patologia , Nervos Periféricos/fisiopatologia , Baço/metabolismo
13.
Neurobiol Dis ; 33(1): 96-103, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18996482

RESUMO

We have previously shown that mice heterozygously deficient for P0 are characterized by a late onset myelin disorder implicating CD8+ T-lymphocytes and macrophages. We now investigated the impact of the co-inhibitory molecule "programmed death" (PD)-1 (CD279), a CD28-related receptor expressed on activated T- and B-lymphocytes on the pathogenic phenotype of CD8+ T-lymphocytes in the P0 myelin mutants. PD-1 deficiency in P0+/- mice leads to a stronger increase of CD8+ T-lymphocytes and a substantially aggravated histological phenotype in the PNS compared to P0+/- mice expressing PD-1. Correspondingly, functional down-stream features, such as electrophysiological parameters, walking coordination and mechano-sensation are more affected than in PD-1-expressing myelin mutants. Our study demonstrates that a monogenic nerve disorder can be substantially modified by immune-controlling mechanisms. Thus, understanding the implication of disease-modifiers in inherited demyelination could be of pivotal interest for limiting the detrimental impact of primarily genetically-mediated myelin disorders by fostering immuno-regulatory pathways.


Assuntos
Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Doenças Desmielinizantes/fisiopatologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Animais , Linfócitos T CD8-Positivos/imunologia , Doença de Charcot-Marie-Tooth , Quimera , Doenças Desmielinizantes/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Apraxia da Marcha/fisiopatologia , Interferon gama/metabolismo , Ativação Linfocitária , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Doenças do Sistema Nervoso Periférico/imunologia , Receptor de Morte Celular Programada 1 , Músculo Quadríceps/inervação , Nervo Isquiático/fisiopatologia , Nervo Isquiático/ultraestrutura , Raízes Nervosas Espinhais/fisiopatologia , Raízes Nervosas Espinhais/ultraestrutura , Estatísticas não Paramétricas , Tato/fisiologia
14.
Mol Cell Neurosci ; 38(4): 489-94, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18555697

RESUMO

We have recently reported that overexpression of proteolipid protein in oligodendrocytes leads to a pathologically relevant increase of both CD8+ T-lymphocytes and CD11b+ cells in the CNS. We now focussed on the origin of the CD11b+ cells in the optic nerve, a well established structure for the analysis of the mutant, using bone marrow chimeric mice. Although there is an age-related increase in CD11b+ cells in the myelinated part of the optic nerve of the mutants, the percentage of infiltrating cells was not increased, but enhanced proliferation was detectable. In the non-myelinated optic nerve head, the rate of infiltrating CD11b+ cells and albumin extravasation was high in both genotypes. However, albumin extravasation was also high in the rostral myelinated part, where CD11b+ cell influx was low. Our study demonstrates an intrinsic origin of CD11b+ cells in the presence of an unchanged blood-brain-barrier in a CNS myelin mutant.


Assuntos
Barreira Hematoencefálica/patologia , Antígeno CD11b/biossíntese , Diferenciação Celular/genética , Macrófagos/patologia , Proteína Proteolipídica de Mielina/genética , Nervo Óptico/patologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Transplante de Medula Óssea/métodos , Antígeno CD11b/genética , Diferenciação Celular/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/biossíntese , Nervo Óptico/imunologia , Nervo Óptico/metabolismo
15.
Mol Cell Neurosci ; 37(2): 359-66, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18326085

RESUMO

Macrophages are critically involved in the pathogenesis of genetically caused demyelination, as it occurs in models for inherited demyelinating neuropathies. It is presently unknown which factors link the Schwann cell-based myelin mutation to the activation of endoneurial macrophages. Here we identified the chemokine monocyte chemoattractant protein-1 (MCP-1) as a first and crucial factor upregulated in Schwann cells of mice heterozygously deficient for the myelin protein zero. The chemokine could be identified as an important mediator of macrophage immigration into peripheral nerves. Furthermore, a 50% reduction of chemokine expression by crossbreeding with MCP-1-deficient mice reduced the increase in macrophage numbers in the mutant nerves and lead to a robust amelioration of pathology. Surprisingly, the complete absence of MCP-1 aggravated the disease. Our findings show that reducing but not eliminating chemokine expression can rescue genetically caused demyelination that may be an interesting target in treating demyelinating diseases of the peripheral nervous system.


Assuntos
Quimiocina CCL2/genética , Macrófagos/imunologia , Nervos Periféricos/imunologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/imunologia , Células de Schwann/imunologia , Animais , Quimiocina CCL2/imunologia , Quimiocina CCL2/metabolismo , Quimiocinas/genética , Quimiocinas/imunologia , Quimiocinas/metabolismo , Quimiotaxia de Leucócito/genética , Modelos Animais de Doenças , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteína P0 da Mielina/genética , Bainha de Mielina/imunologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Fibras Nervosas Mielinizadas/imunologia , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Nervos Periféricos/metabolismo , Nervos Periféricos/fisiopatologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Polirradiculoneuropatia/genética , Polirradiculoneuropatia/imunologia , Polirradiculoneuropatia/fisiopatologia , Células de Schwann/metabolismo
16.
Neurobiol Dis ; 25(1): 105-11, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17064921

RESUMO

PLP overexpressing mice display demyelination and axonopathic changes, accompanied by an elevation of CD8+ T-lymphocytes and CD11b+ macrophages in the CNS. By crossbreeding these mutants with RAG-1-deficient mice lacking mature lymphocytes, we could recently demonstrate a pathogenetic impact of the CD8+ cells. In the present study, we investigated the pathogenetic impact of CD11b+ macrophages by crossbreeding the myelin mutants with knockout mice deficient for the macrophage-restricted adhesion molecule sialoadhesin (Sn). In the wild-type mice, Sn is barely detectable on CD11b+ cells, whereas in the myelin mutants, almost all CD11b+ cells express Sn. In the double mutants, upregulation of CD8+ T-cells and CD11b+ macrophages is reduced and pathological alterations are ameliorated. These data indicate that in a primarily genetically caused myelin disorder of the CNS macrophages expressing Sn partially mediate pathogenesis. These findings may have substantial impact on treatment strategies for leukodystrophic disorders and some forms of multiple sclerosis.


Assuntos
Axônios/patologia , Sistema Nervoso Central/patologia , Glicoproteínas de Membrana/fisiologia , Bainha de Mielina/patologia , Fosfolipases/genética , Fosfolipases/fisiologia , Receptores Imunológicos/fisiologia , Animais , Axônios/fisiologia , Antígeno CD11b/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Genótipo , Imuno-Histoquímica , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Bainha de Mielina/metabolismo , Fosfolipases/biossíntese , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA