Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Neuromolecular Med ; 25(3): 350-359, 2023 09.
Article in English | MEDLINE | ID: mdl-36857006

ABSTRACT

Interleukin-17A plays a crucial role in multiple sclerosis and other autoimmune diseases. Although the link between IL-17 and disease activity has been clearly demonstrated, the precise function of this cytokine remains elusive. Here, we investigated the function of astrocyte-targeted IL-17A production in GF/IL-17 transgenic mice during EAE. In particular, IL-17A is important during disease induction. In mice with transgenic IL-17A production, disease occurs earlier and peak disease is more severe, whereas remission is unimpaired. IL-17A synthesis is associated with increased infiltration of granulocytes into the CNS and microglial activation. Moreover, IL-17A synthesis allows induction of MOG-EAE without the additional administration of the co-adjuvant pertussis toxin. Examination of double transgenic GF/IL-17 2D2 mice revealed that, in addition, local IL-17A production facilitates spontaneous infiltration of immune cells into the CNS in mice expressing a MOG-specific T-cell receptor. Overall, we provide evidence for a crucial effect of IL-17A in the induction phase of EAE, facilitating the infiltration of granulocytes and autoreactive T-cells into the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Interleukin-17 , T-Lymphocytes , Mice, Transgenic , Granulocytes , Mice, Inbred C57BL
2.
Int J Stroke ; 18(2): 242-247, 2023 02.
Article in English | MEDLINE | ID: mdl-35361026

ABSTRACT

RATIONALE: Aneurysmal subarachnoid hemorrhage (SAH) has high morbidity and mortality. While the primary injury results from the initial bleeding cannot currently be influenced, secondary injury through vasospasm and delayed cerebral ischemia worsens outcome and might be a target for interventions to improve outcome. To date, beside the aneurysm treatment to prevent re-bleeding and the administration of oral nimodipine, there is no therapy available, so novel treatment concepts are needed. Evidence suggests that inflammation contributes to delayed cerebral ischemia and poor outcome in SAH. Some studies suggest a beneficial effect of anti-inflammatory glucocorticoids, but there are no data from randomized controlled trials examining the efficacy of glucocorticoids. Therefore, current guidelines do not recommend the use of glucocorticoids in SAH. AIM: The Fight INflammation to Improve outcome after aneurysmal Subarachnoid HEmorRhage (FINISHER) trial aims to determine whether dexamethasone improves outcome in a clinically relevant endpoint in SAH patients. METHODS AND DESIGN: FINISHER is a multicenter, prospective, randomized, double-blinded, placebo-controlled clinical phase III trial which is testing the outcome and safety of anti-inflammatory treatment with dexamethasone in SAH patients. SAMPLE SIZE ESTIMATES: In all, 334 patients will be randomized to either dexamethasone or placebo within 48 h after SAH. The dexamethasone dose is 8 mg tds for days 1-7 and then 8 mg od for days 8-21. STUDY OUTCOME: The primary outcome is the modified Rankin Scale (mRS) at 6 months, which is dichotomized to favorable (mRS 0-3) versus unfavorable (mRS 4-6). DISCUSSION: The results of this study will provide the first phase III evidence as to whether dexamethasone improves outcome in SAH.


Subject(s)
Brain Ischemia , Stroke , Subarachnoid Hemorrhage , Vasospasm, Intracranial , Humans , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Prospective Studies , Treatment Outcome , Stroke/complications , Brain Ischemia/complications , Brain Ischemia/drug therapy , Cerebral Infarction/complications , Inflammation/complications , Dexamethasone/therapeutic use , Vasospasm, Intracranial/prevention & control , Randomized Controlled Trials as Topic , Multicenter Studies as Topic , Clinical Trials, Phase III as Topic
3.
J Neuroinflammation ; 18(1): 101, 2021 Apr 27.
Article in English | MEDLINE | ID: mdl-33906683

ABSTRACT

BACKGROUND: Interleukin 23 is a critical cytokine in the pathogenesis of multiple sclerosis. But the local impact of interleukin 23 on the course of neuroinflammation is still not well defined. To further characterize the effect of interleukin 23 on CNS inflammation, we recently described a transgenic mouse model with astrocyte-specific expression of interleukin 23 (GF-IL23 mice). The GF-IL23 mice spontaneously develop a progressive ataxic phenotype with cerebellar tissue destruction and inflammatory infiltrates with high amounts of B cells most prominent in the subarachnoid and perivascular space. METHODS: To further elucidate the local impact of the CNS-specific interleukin 23 synthesis in autoimmune neuroinflammation, we induced a MOG35-55 experimental autoimmune encephalomyelitis (EAE) in GF-IL23 mice and WT mice and analyzed the mice by histology, flow cytometry, and transcriptome analysis. RESULTS: We were able to demonstrate that local interleukin 23 production in the CNS leads to aggravation and chronification of the EAE course with a severe paraparesis and an ataxic phenotype. Moreover, enhanced multilocular neuroinflammation was present not only in the spinal cord, but also in the forebrain, brainstem, and predominantly in the cerebellum accompanied by persisting demyelination. Thereby, interleukin 23 creates a pronounced proinflammatory response with accumulation of leukocytes, in particular B cells, CD4+ cells, but also γδ T cells and activated microglia/macrophages. Furthermore, transcriptome analysis revealed an enhanced proinflammatory cytokine milieu with upregulation of lymphocyte activation markers, co-stimulatory markers, chemokines, and components of the complement system. CONCLUSION: Taken together, the GF-IL23 model allowed a further breakdown of the different mechanisms how IL-23 drives neuroinflammation in the EAE model and proved to be a useful tool to further dissect the impact of interleukin 23 on neuroinflammatory models.


Subject(s)
Astrocytes/immunology , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Interleukin-23/immunology , Animals , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/metabolism , Interleukin-23/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
4.
Mol Neurobiol ; 56(12): 7977-7993, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31154574

ABSTRACT

Interleukin 23 (IL-23) is a key mediator in neuroinflammation in numerous autoimmune diseases including multiple sclerosis (MS). However, the pathophysiology of IL-23 and how it contributes to neuroinflammation is poorly defined. To further clarify the role of IL-23 in CNS inflammation, we generated a transgenic mouse model (GF-IL23) with astrocyte-targeted expression of both IL-23 subunits, IL-23p19, and IL-23p40. These GF-IL23 mice spontaneously develop a progressive ataxic phenotype, which corresponds to cerebellar tissue destruction, and inflammatory infiltrates most prominent in the subarachnoidal and perivascular space. The CNS-cytokine milieu was characterized by numerous inflammatory mediators such as IL-17a and IFNγ. However, the leukocytic infiltrates were surprisingly predominated by B cells. To further examine the impact of the CNS-specific IL-23 synthesis on an additional systemic inflammatory stimulus, we applied the LPS-induced endotoxemia model. Administration of LPS in GF-IL23 mice resulted in early and pronounced microglial activation, enhanced cytokine production and, in sharp contrast to control animals, in the formation of lymphocytic infiltrates. Our model confirms a critical role for IL-23 in the induction of inflammation in the CNS, in particular facilitating the accumulation of lymphocytes in and around the brain. Thereby, CNS-specific synthesis of IL-23 is able to induce a cascade of inflammatory cytokines leading to microglia activation, astrocytosis, and ultimately tissue damage. The presented transgenic model will be a useful tool to further dissect the role of IL-23 in neuroinflammation.


Subject(s)
B-Lymphocytes/metabolism , Brain/metabolism , Cerebellar Ataxia/metabolism , Disease Progression , Interleukin-23/biosynthesis , T-Lymphocytes/metabolism , Animals , Brain/diagnostic imaging , Cerebellar Ataxia/diagnostic imaging , Cerebellar Ataxia/etiology , Disease Models, Animal , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic
5.
Mol Neurobiol ; 55(2): 946-957, 2018 02.
Article in English | MEDLINE | ID: mdl-28084589

ABSTRACT

Recent evidence suggests a pivotal role of the proinflammatory cytokine interleukin - 17A (IL-17) in demyelinating autoimmune diseases of the central nervous system (CNS) such as multiple sclerosis (MS). Nevertheless, it remains unclear if this cytokine exerts direct effects on CNS resident cells during MS or modulates the function of infiltrating immune cells towards a more detrimental phenotype. Here, we investigated the effects of locally produced IL-17 during experimental demyelination of the CNS using the cuprizone (CPZ) model in mice with (GF/IL17) or without transgenic production of IL-17 by astrocytes in the CNS. During early demyelination, GF/IL17 mice demonstrated enhanced activity and decreased anxiety-related behavior in the elevated plus maze suggesting a more severe disease course. Furthermore, in GF/IL17 mice, toxic demyelination was accelerated and synthesis of myelin proteins was reduced. Early demyelination was accompanied by an increased ratio of infiltrating granulocytes in GF/ILl17 mice. The presence of IL-17 during CPZ treatment increased the accumulation of activated microglia and sustained microglial proliferation during myelin loss. Taken together, our results argue for a detrimental role of IL-17 during demyelinating diseases.


Subject(s)
Astrocytes/metabolism , Behavior, Animal/physiology , Demyelinating Diseases/metabolism , Granulocytes/metabolism , Interleukin-17/metabolism , Microglia/metabolism , Myelin Sheath/metabolism , Animals , Anxiety/metabolism , Anxiety/pathology , Astrocytes/drug effects , Astrocytes/pathology , Behavior, Animal/drug effects , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Granulocytes/drug effects , Granulocytes/pathology , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Myelin Sheath/drug effects , Myelin Sheath/pathology
6.
J Clin Invest ; 125(1): 365-78, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25500888

ABSTRACT

Chemokines are important modulators of neuroinflammation and neurodegeneration. In the brains of Alzheimer's disease (AD) patients and in AD animal models, the chemokine CXCL10 is found in high concentrations, suggesting a pathogenic role for this chemokine and its receptor, CXCR3. Recent studies aimed at addressing the role of CXCR3 in neurological diseases indicate potent, but diverse, functions for CXCR3. Here, we examined the impact of CXCR3 in the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model of AD. We found that, compared with control APP/PSI animals, plaque burden and Aß levels were strongly reduced in CXCR3-deficient APP/PS1 mice. Analysis of microglial phagocytosis in vitro and in vivo demonstrated that CXCR3 deficiency increased the microglial uptake of Aß. Application of a CXCR3 antagonist increased microglial Aß phagocytosis, which was associated with reduced TNF-α secretion. Moreover, in CXCR3-deficient APP/PS1 mice, microglia exhibited morphological activation and reduced plaque association, and brain tissue from APP/PS1 animals lacking CXCR3 had reduced concentrations of proinflammatory cytokines compared with controls. Further, loss of CXCR3 attenuated the behavioral deficits observed in APP/PS1 mice. Together, our data indicate that CXCR3 signaling mediates development of AD-like pathology in APP/PS1 mice and suggest that CXCR3 has potential as a therapeutic target for AD.


Subject(s)
Alzheimer Disease/metabolism , Plaque, Amyloid/pathology , Receptors, CXCR3/physiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Female , Male , Maze Learning , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Phagocytosis , Signal Transduction , Transcriptional Activation , Tumor Necrosis Factor-alpha/metabolism
7.
J Neuroinflammation ; 11: 109, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24930935

ABSTRACT

BACKGROUND: The functional state of glial cells, like astrocytes and microglia, critically modulates the course of neuroinflammatory and neurodegenerative diseases and can have both detrimental and beneficial effects. Glial cell function is tightly controlled by cellular interactions in which cytokines are important messengers. Recent studies provide evidence that in particular chemokines are important modulators of glial cell function. During the course of CNS diseases like multiple sclerosis or Alzheimer's disease, and in the corresponding animal models, the chemokines CXCL9 and CXCL10 are abundantly expressed at sites of glial activation, arguing for an important role of these chemokines and their corresponding receptor CXCR3 in glial activation. To clarify the role of this chemokine system in glial cell activation, we characterized the impact of CXCR3 on glial activation in a model of toxic demyelination in which glial activation without a prominent influx of hematogenous cells is prototypical. METHODS: We investigated the impact of CXCR3 on cuprizone-induced demyelination, comparing CXCR3-deficient mice with wild type controls. The clinical course during cuprizone feeding was documented for five weeks and for the subsequent four days withdrawal of the cuprizone diet (5.5 weeks). Glial activation was characterized using histological, histomorphometric and phenotypic analysis. Molecular analysis for (de)myelination and neuroinflammation was applied to characterize the effect of cuprizone on CXCR3-deficient mice and control animals. RESULTS: CXCR3-deficient mice displayed a milder clinical course during cuprizone feeding and a more rapid body weight recovery after offset of diet. In the CNS, CXCR3 deficiency significantly attenuated the accumulation and activation of microglia and astrocytes. Moreover, a deficiency of CXCR3 reduced the expression of the microglial activation markers CD45 and CD11b. Compared to controls, we observed a vast reduction of RNA levels for proinflammatory cytokines and chemokines like Ccl2, Cxcl10, Tnf and Il6 within the CNS of cuprizone-treated mice. Lastly, CXCR3 deficiency had no major effects on the course of demyelination during cuprizone feeding. CONCLUSIONS: The CXCR3 chemokine system is critically involved in the intrinsic glial activation during cuprizone-induced demyelination, which significantly modulates the distribution of glial cells and the local cytokine milieu.


Subject(s)
Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Monoamine Oxidase Inhibitors/toxicity , Neuroglia/metabolism , Receptors, CXCR3/genetics , Analysis of Variance , Animals , Antigens, CD/metabolism , Body Weight/drug effects , Cuprizone/administration & dosage , Cytokines/genetics , Cytokines/metabolism , Flow Cytometry , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monoamine Oxidase Inhibitors/administration & dosage , Neuroglia/drug effects , RNA, Messenger/metabolism , Receptors, CXCR3/deficiency
8.
Mol Neurobiol ; 50(2): 534-44, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24567119

ABSTRACT

This article gives a comprehensive overview of cytokine and other inflammation associated protein levels in plasma, serum and cerebrospinal fluid (CSF) of patients with Alzheimer's disease (AD) and mild cognitive impairment (MCI). We reviewed 118 research articles published between 1989 and 2013 to compare the reported levels of 66 cytokines and other proteins related to regulation and signaling in inflammation in the blood or CSF obtained from MCI and AD patients. Several cytokines are evidently regulated in (neuro-) inflammatory processes associated with neurodegenerative disorders. Others do not display changes in the blood or CSF during disease progression. However, many reports on cytokine levels in MCI or AD are controversial or inconclusive, particularly those which provide data on frequently investigated cytokines like tumor necrosis factor alpha (TNF-α) or interleukin-6 (IL-6). The levels of several cytokines are possible indicators of neuroinflammation in AD. Some of them might increase steadily during disease progression or temporarily at the time of MCI to AD conversion. Furthermore, elevated body fluid cytokine levels may correlate with an increased risk of conversion from MCI to AD. Yet, research results are conflicting. To overcome interindividual variances and to obtain a more definite description of cytokine regulation and function in neurodegeneration, a high degree of methodical standardization and patients collective characterization, together with longitudinal sampling over years is essential.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Body Fluids/metabolism , Cognitive Dysfunction/metabolism , Cytokines , Alzheimer Disease/blood , Amyloid beta-Peptides/blood , Animals , Cognitive Dysfunction/blood , Cytokines/blood , Cytokines/metabolism , Humans , Inflammation/blood , Inflammation/metabolism
9.
PLoS One ; 8(2): e57307, 2013.
Article in English | MEDLINE | ID: mdl-23468966

ABSTRACT

Interleukin-17A (IL-17A) is a key cytokine modulating the course of inflammatory diseases. Whereas effector functions of IL-17A like induction of antimicrobial peptides and leukocyte infiltration could clearly be demonstrated for peripheral organs, CNS specific effects are not well defined and appear controversial. To further clarify the functional significance of IL-17A in the CNS, we generated a transgenic mouse line with astrocyte-restricted expression of the IL-17A gene. GFAP/IL-17A transgenic mice develop normally and do not show any signs of neurological dysfunction. However, histological characterization revealed astrocytosis and activation of microglia. Demyelination, neurodegeneration or prominent tissue damage was not observed but a vascular pathology mimicking microangiopathic features was evident. Histological and flow cytometric analysis demonstrated the absence of parenchymal infiltration of immune cells into the CNS of GFAP/IL-17A transgenic mice. In GFAP/IL-17A mice, LPS-induced endotoxemia led to a more pronounced microglial activation with expansion of a distinct CD45(high)/CD11b(+) population and increased induction of proinflammatory cytokines compared with controls. Our data argues against a direct role of IL-17A in mediating tissue damage during neuroinflammation. More likely IL-17A acts as a modulating factor in the network of induced cytokines. This novel mouse model will be a very useful tool to further characterize the role of IL-17A in neuroinflammatory disease models.


Subject(s)
Central Nervous System/metabolism , Endotoxemia/pathology , Inflammation/pathology , Interleukin-17/biosynthesis , Microvessels/pathology , Neuroglia/pathology , Animals , Blood-Brain Barrier , Cells, Cultured , Central Nervous System/pathology , Flow Cytometry , Glial Fibrillary Acidic Protein/genetics , Interleukin-17/genetics , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron, Transmission , Reverse Transcriptase Polymerase Chain Reaction
10.
Am J Pathol ; 179(5): 2346-59, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21925471

ABSTRACT

CXCR3 and its ligands are important for the trafficking of activated CD4(+) T(H)1 T cells, CD8(+) T cells, and natural killer cells during inflammation. Recent functional studies demonstrate a more diverse role of CXCR3 in inflammatory diseases of the central nervous system (CNS). We examined the impact of CXCR3 on a less complex interferon-γ-dependent, type 1 cell-mediated immune response in the CNS, induced in mice by the transgenic production of glial fibrillary acidic protein IL-12 (GF-IL12) by astrocytes and retinal Müller cells. GF-IL12 mice develop ataxia because of severe cerebellar inflammation but have little overt ocular disease. Surprisingly, CXCR3-deficient GF-IL12 mice (GF-IL12/CXCR3KO) have drastically reduced ataxia but developed cataracts, severe ocular inflammation, and eye atrophy. Most GF-IL12/CXCR3KO mice had minimal cerebellar inflammation but severe retinal disorganization, loss of photoreceptors, and lens destruction in the eye. The number of CD3(+), CD11b(+), and natural killer 1.1(+) cells were reduced in the CNS but highly increased in the eyes of GF-IL12/CXCR3KO compared with GF-IL12 mice. High levels of interferon-γ, IL-1, tumor necrosis factor α, CXCL9, CXCL10, and CCL5 were found in GF-IL12 cerebelli and GF-IL12/CXCR3KO eyes. Our findings demonstrate key but paradoxical functions for CXCR3 in IL-12-induced immune disease in the CNS, promoting inflammation in the brain yet restricting it in the eye. We conclude that the function of CXCR3 in cellular immune disease is driven by a common trigger and is controlled by tissue-specific factors.


Subject(s)
Astrocytes/metabolism , Cerebellar Ataxia/immunology , Eye Diseases/immunology , Immunity, Cellular/immunology , Interleukin-12/biosynthesis , Nerve Tissue Proteins/metabolism , Receptors, CXCR3/deficiency , Animals , Blindness/immunology , Cerebellar Ataxia/pathology , Encephalitis/immunology , Encephalitis/pathology , Endophthalmitis/immunology , Endophthalmitis/pathology , Eye Diseases/pathology , Genotype , Glial Fibrillary Acidic Protein , Interferon-gamma/metabolism , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Messenger/metabolism , Receptors, CCR5/metabolism , Receptors, Interleukin-12/metabolism , STAT4 Transcription Factor/metabolism , Signal Transduction
11.
J Peripher Nerv Syst ; 15(4): 357-65, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21199107

ABSTRACT

Macrophages are intimately involved in the pathogenesis of peripheral nervous system (PNS) disorders. Recently, we characterized a resident endoneurial macrophage population, which contributes rapidly to the endoneurial macrophage response in PNS diseases. Unlike microglial cells, resident macrophages undergo a physiological turnover of 50% in the sciatic nerve and 80% in dorsal root ganglia (DRG) within 12 weeks. Further information about the dynamics of this turnover is not available. This study examined the macrophage turnover in the sciatic nerve and DRGs over a longer period and addresses the question whether the turnover of resident macrophages is complete or whether there is a truly resident endoneurial macrophage population. We used chimeric mice carrying GFP(+) bone marrow and immunohistochemistry to detect hematogenous (GFP(+)) endoneurial macrophages after turnover. Non-exchanged, resident macrophages were GFP(-). Quantification of GFP(+) and GFP(-) macrophages revealed a maximal turnover of 75%, reached in DRGs after 12 weeks and in sciatic nerves after 36 weeks. GFP(-) long-term resident macrophages were further characterized after sciatic nerve injury, where they participated in the early macrophage response of Wallerian degeneration. Our results point toward a small but truly resident PNS macrophage population. These macrophages are an interesting target for further characterization and might have a distinct role in peripheral nerve disease.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Macrophages/cytology , Macrophages/physiology , Peripheral Nerves/cytology , Peripheral Nerves/physiology , Peripheral Nervous System/cytology , Peripheral Nervous System/physiology , Animals , Cell Survival/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radiation Chimera/genetics
12.
J Immunol ; 183(3): 2079-88, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19597000

ABSTRACT

IL-6 is crucial for the induction of many murine models of autoimmunity including experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. To establish the role of site-specific production of IL-6 in autoimmunity, we examined myelin oligodendrocyte glycoprotein immunization-induced EAE in transgenic mice (GFAP-IL6) with IL-6 production restricted to the cerebellum. Myelin oligodendrocyte glycoprotein-immunized (Mi-) GFAP-IL6 mice developed severe ataxia but no physical signs of spinal cord involvement, which was in sharp contrast to Mi-wild type (WT) animals that developed classical EAE with ascending paralysis. Immune pathology and demyelination were nearly absent from the spinal cord, but significantly increased in the cerebellum of Mi-GFAP-IL6 mice. Tissue damage in the cerebellum in the Mi-GFAP-IL6 mice was accompanied by increased total numbers of infiltrating leukocytes and increased proportions of both neutrophils and B-cells. With the exception of IL-17 mRNA, which was elevated in both control immunized and Mi-GFAP-IL6 cerebellum, the level of other cytokine and chemokine mRNAs were comparable with Mi-WT cerebellum whereas significantly higher levels of IFN-gamma and TNF-alpha mRNA were found in Mi-WT spinal cord. Thus, site-specific production of IL-6 in the cerebellum redirects trafficking away from the normally preferred antigenic site the spinal cord and acts as a leukocyte "sink" that markedly enhances the inflammatory cell accumulation and disease. The mechanisms underlying this process likely include the induction of specific chemokines, activation of microglia, and activation and loss of integrity of the blood-brain barrier present in the cerebellum of the GFAP-IL6 mice before the induction of EAE.


Subject(s)
Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Inflammation/etiology , Interleukin-6/biosynthesis , Animals , Autoimmunity , Central Nervous System/metabolism , Cerebellum/pathology , Chemotaxis , Cytokines/analysis , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Interleukin-6/immunology , Mice , Mice, Transgenic , Myelin Proteins , Myelin-Associated Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein , Paralysis
SELECTION OF CITATIONS
SEARCH DETAIL
...