Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Neuropathol ; 141(6): 881-899, 2021 06.
Article in English | MEDLINE | ID: mdl-33779783

ABSTRACT

Meningeal inflammation strongly associates with demyelination and neuronal loss in the underlying cortex of progressive MS patients, thereby contributing significantly to clinical disability. However, the pathological mechanisms of meningeal inflammation-induced cortical pathology are still largely elusive. By extensive analysis of cortical microglia in post-mortem progressive MS tissue, we identified cortical areas with two MS-specific microglial populations, termed MS1 and MS2 cortex. The microglial population in MS1 cortex was characterized by a higher density and increased expression of the activation markers HLA class II and CD68, whereas microglia in MS2 cortex showed increased morphological complexity and loss of P2Y12 and TMEM119 expression. Interestingly, both populations associated with inflammation of the overlying meninges and were time-dependently replicated in an in vivo rat model for progressive MS-like chronic meningeal inflammation. In this recently developed animal model, cortical microglia at 1-month post-induction of experimental meningeal inflammation resembled microglia in MS1 cortex, and microglia at 2 months post-induction acquired a MS2-like phenotype. Furthermore, we observed that MS1 microglia in both MS cortex and the animal model were found closely apposing neuronal cell bodies and to mediate pre-synaptic displacement and phagocytosis, which coincided with a relative sparing of neurons. In contrast, microglia in MS2 cortex were not involved in these synaptic alterations, but instead associated with substantial neuronal loss. Taken together, our results show that in response to meningeal inflammation, microglia acquire two distinct phenotypes that differentially associate with neurodegeneration in the progressive MS cortex. Furthermore, our in vivo data suggests that microglia initially protect neurons from meningeal inflammation-induced cell death by removing pre-synapses from the neuronal soma, but eventually lose these protective properties contributing to neuronal loss.


Subject(s)
Cerebral Cortex/pathology , Meninges/pathology , Microglia/pathology , Multiple Sclerosis/pathology , Neurodegenerative Diseases/pathology , Neuroinflammatory Diseases/pathology , Neurons/pathology , Adult , Aged , Animals , Cell Death , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Female , Humans , Meninges/immunology , Microglia/classification , Microglia/immunology , Microglia/metabolism , Middle Aged , Multiple Sclerosis/immunology , Neurodegenerative Diseases/immunology , Phenotype , Rats
2.
Acta Neuropathol Commun ; 8(1): 66, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32398070

ABSTRACT

Analysis of isolated meninges and cerebrospinal fluid (CSF) of post-mortem MS cases has shown increased gene and protein expression for the pro-inflammatory cytokines: tumour necrosis factor (TNF) and interferon-γ (IFNγ). Here we tested the hypothesis that persistent production of these cytokines in the meningeal compartment and diffusion into underlying GM can drive chronic MS-like GM pathology. Lentiviral transfer vectors were injected into the sagittal sulcus of DA rats to deliver continuous expression of TNF + IFNγ transgenes in the meninges and the resulting neuropathology analysed after 1 and 2 months. Injection of TNF + IFNγ viral vectors, with or without prior MOG immunisation, induced extensive immune cell infiltration (CD4+ and CD8+ T-cells, CD79a + B-cells and macrophages) in the meninges by 28 dpi, which remained at 2 months. Control GFP viral vector did not induce infiltration. Subpial demyelination was seen underlying these infiltrates, which was partly dependant on prior myelin oligodendrocyte glycoprotein (MOG) immunisation. A significant decrease in neuronal numbers was seen at 28 and 56 days in cortical layers II-V that was independent of MOG immunisation. RNA analysis at 28 dpi showed an increase in expression of necroptotic pathway genes, including RIP3, MLKL, cIAP2 and Nox2. PhosphoRIP3+ and phosphoMLKL+ neurons were present in TNF + IFNγ vector injected animals, indicating activation of necroptosis. Our results suggest that persistent expression of TNF in the presence of IFNγ is a potent inducer of meningeal inflammation and can activate TNF signalling pathways in cortical cells leading to neuronal death and subpial demyelination and thus may contribute to clinical progression in MS.


Subject(s)
Demyelinating Diseases/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Interferon-gamma/metabolism , Nerve Degeneration/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cerebral Cortex/immunology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cytokines , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/immunology , Meninges/immunology , Meninges/metabolism , Meninges/pathology , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Nerve Degeneration/immunology , Rats , Tumor Necrosis Factor-alpha/immunology
3.
Glia ; 64(1): 105-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26337870

ABSTRACT

Multiple sclerosis (MS) frequently starts near the lateral ventricles, which are lined by subventricular zone (SVZ) progenitor cells that can migrate to lesions and contribute to repair. Because MS-induced inflammation may decrease SVZ proliferation and thus limit repair, we studied the role of galectin-3 (Gal-3), a proinflammatory protein. Gal-3 expression was increased in periventricular regions of human MS in post-mortem brain samples and was also upregulated in periventricular regions in a murine MS model, Theiler's murine encephalomyelitis virus (TMEV) infection. Whereas TMEV increased SVZ chemokine (CCL2, CCL5, CCL, and CXCL10) expression in wild type (WT) mice, this was inhibited in Gal-3(-/-) mice. Though numerous CD45+ immune cells entered the SVZ of WT mice after TMEV infection, their numbers were significantly diminished in Gal-3(-/-) mice. TMEV also reduced neuroblast and proliferative SVZ cell numbers in WT mice but this was restored in Gal-3(-/-) mice and was correlated with increased numbers of doublecortin+ neuroblasts in the corpus callosum. In summary, our data showed that loss of Gal-3 blocked chemokine increases after TMEV, reduced immune cell migration into the SVZ, reestablished SVZ proliferation and increased the number of progenitors in the corpus callosum. These results suggest Gal-3 plays a central role in modulating the SVZ neurogenic niche's response to this model of MS.


Subject(s)
Brain/metabolism , Galectin 3/metabolism , Multiple Sclerosis/metabolism , Nervous System Autoimmune Disease, Experimental/metabolism , Neurogenesis , Stem Cell Niche/physiology , Adolescent , Adult , Aged , Animals , Brain/immunology , Brain/pathology , Cell Movement , Child , Female , Galectin 3/genetics , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Nervous System Autoimmune Disease, Experimental/immunology , Nervous System Autoimmune Disease, Experimental/pathology , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Poliomyelitis/metabolism , Poliomyelitis/pathology , Theilovirus , Young Adult
4.
Eur J Neurosci ; 32(4): 684-92, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20646047

ABSTRACT

Many speech sounds, such as vowels, exhibit a characteristic pattern of spectral peaks, referred to as formants, the frequency positions of which depend both on the phonological identity of the sound (e.g. vowel type) and on the vocal-tract length of the speaker. This study investigates the processing of formant information relating to vowel type and vocal-tract length in human auditory cortex by measuring electroencephalographic (EEG) responses to synthetic unvoiced vowels and spectrally matched noises. The results revealed specific sensitivity to vowel formant information in both anterior (planum polare) and posterior (planum temporale) regions of auditory cortex. The vowel-specific responses in these two areas appeared to have different temporal dynamics; the anterior source produced a sustained response for as long as the incoming sound was a vowel, whereas the posterior source responded transiently when the sound changed from a noise to a vowel, or when there was a change in vowel type. Moreover, the posterior source appeared to be largely invariant to changes in vocal-tract length. The current findings indicate that the initial extraction of vowel type from formant information is complete by the level of non-primary auditory cortex, suggesting that speech-specific processing may involve primary auditory cortex, or even subcortical structures. This challenges the view that specific sensitivity to speech emerges only beyond unimodal auditory cortex.


Subject(s)
Auditory Cortex/anatomy & histology , Auditory Cortex/physiology , Phonetics , Speech/physiology , Acoustic Stimulation/methods , Adolescent , Adult , Electroencephalography , Humans , Male , Time Factors , Young Adult
5.
J Neuroinflammation ; 5: 44, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18922161

ABSTRACT

BACKGROUND: The periventricular subventricular zone (SVZ) contains stem cells and is an area of active neurogenesis and migration. Since inflammation can reduce neurogenesis, we tested whether Theiler's murine encephalomyelitis virus (TMEV) induces inflammation and reduces neurogenesis in the SVZ. METHODS: We performed immmunohistochemistry for the hematopoietic cell marker CD45 throughout the central nervous system and then examined neuroblasts in the SVZ. RESULTS: CD45+ activation (inflammation) occurred early in the forebrain and preceded cerebellar and spinal cord inflammation. Inflammation in the brain was regionally stochastic except for the SVZ and surrounding periventricular regions where it was remarkably pronounced and consistent. In preclinical mice, SVZ neuroblasts emigrated into inflamed periventricular regions. The number of proliferating phoshpohistone3+ cells and Doublecortin+ (Dcx) SVZ neuroblasts was overall unaffected during the periods of greatest inflammation. However the number of Dcx+ and polysialylated neural cell adhesion molecule (PSA-NCAM+) SVZ neuroblasts decreased only after periventricular inflammation abated. CONCLUSION: Our results suggest that after TMEV infection, the SVZ may mount an attempt at neuronal repair via emigration, a process dampened by decreases in neuroblast numbers.


Subject(s)
Cardiovirus Infections/immunology , Encephalitis/immunology , Hematopoietic Stem Cells/immunology , Leukocyte Common Antigens/immunology , Neurogenesis/immunology , Theilovirus/immunology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cardiovirus Infections/physiopathology , Cell Lineage/immunology , Cell Movement/immunology , Cell Proliferation , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Encephalitis/physiopathology , Female , Hematopoietic Stem Cells/virology , Histones/metabolism , Lateral Ventricles , Mice , Microtubule-Associated Proteins/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/physiopathology , Nerve Regeneration/immunology , Neuronal Plasticity/immunology , Neurons/cytology , Neurons/immunology , Neuropeptides/metabolism , Prosencephalon/immunology , Prosencephalon/pathology , Prosencephalon/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...