Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Article in English | MEDLINE | ID: mdl-38836920

ABSTRACT

Telehealth services were rapidly adopted during the COVID-19 pandemic, but evidence regarding the effectiveness and feasibility of telehealth services in child and adolescent mental healthcare is sparse. This study aims to investigate feasibility, satisfaction, and goal attainment in video-delivered consultations in routine care child and adolescent psychiatry and psychotherapy. A total of 1046 patients from four university child and adolescent outpatient psychiatric clinics and one university outpatient unit for child and adolescent psychotherapy were screened for study participation. We examined a) the percentage of patients considered eligible for video-delivered consultation, b) clinicians', parents' and patients' satisfaction with video consultation, c) clinicians' ratings of goal attainment in video consultation, and d) factors associated with satisfaction and goal attainment. 59% of the screening sample (n = 621) fulfilled eligibility criteria and were considered eligible for video consultation. A total of 267 patients consented to participate in the study and received a video consultation. Clinicians reported high levels of satisfaction with video consultation and high levels of goal attainment in video consultations, especially for patients scheduled for initial patient assessments. Parents and patients were also highly satisfied with the video consultations, especially if patients had less severe emotional and behavioral problems. The present findings suggest that video consultations are a feasible and well-accepted alternative to in-person consultations in child and adolescent mental health care, especially for children with less severe symptoms and for children in early phases of assessment and treatment. Limitations include the lack of a control group. The study was registered at the German Clinical Trials Registry (DRKS00023525).

2.
PLoS One ; 19(2): e0291368, 2024.
Article in English | MEDLINE | ID: mdl-38306361

ABSTRACT

Glioblastoma multiforme (GBM) and the GBM variant gliosarcoma (GS) are among the tumors with the highest morbidity and mortality, providing only palliation. Stem-like glioma cells (SLGCs) are involved in tumor initiation, progression, therapy resistance, and relapse. The identification of general features of SLGCs could contribute to the development of more efficient therapies. Commercially available protein arrays were used to determine the cell surface signature of eight SLGC lines from GBMs, one SLGC line obtained from a xenotransplanted GBM-derived SLGC line, and three SLGC lines from GSs. By means of non-negative matrix factorization expression metaprofiles were calculated. Using the cophenetic correlation coefficient (CCC) five metaprofiles (MPs) were identified, which are characterized by specific combinations of 7-12 factors. Furthermore, the expression of several factors, that are associated with GBM prognosis, GBM subtypes, SLGC differentiation stages, or neural identity was evaluated. The investigation encompassed 24 distinct SLGC lines, four of which were derived from xenotransplanted SLGCs, and included the SLGC lines characterized by the metaprofiles. It turned out that all SLGC lines expressed the epidermal growth factor EGFR and EGFR ligands, often in the presence of additional receptor tyrosine kinases. Moreover, all SLGC lines displayed a neural signature and the IDH1 wildtype, but differed in their p53 and PTEN status. Pearson Correlation analysis identified a positive association between the pluripotency factor Sox2 and the expression of FABP7, Musashi, CD133, GFAP, but not with MGMT or Hif1α. Spherical growth, however, was positively correlated with high levels of Hif1α, CDK4, PTEN, and PDGFRß, whereas correlations with stemness factors or MGMT (MGMT expression and promoter methylation) were low or missing. Factors highly expressed by all SLGC lines, irrespective of their degree of stemness and growth behavior, are Cathepsin-D, CD99, EMMPRIN/CD147, Intß1, the Galectins 3 and 3b, and N-Cadherin.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Gliosarcoma , Humans , Glioblastoma/metabolism , Gliosarcoma/genetics , Gliosarcoma/metabolism , Gliosarcoma/pathology , Brain Neoplasms/metabolism , Neoplasm Recurrence, Local/pathology , Glioma/pathology , Neoplastic Stem Cells/metabolism , ErbB Receptors/metabolism , Cell Line, Tumor
3.
Article in German | MEDLINE | ID: mdl-34751790

ABSTRACT

BACKGROUND AND OBJECTIVE: The infection protection measures adopted as part of the COVID-19 pandemic led to profound restrictions and changes in the social, (pre-) school, family, and leisure areas. The objective of the current study was to examine the mental burden of children and adolescents and their families during the COVID-19 pandemic. Furthermore, this study aimed to identify possible factors that influence the mental burden. MATERIALS AND METHODS: The examinations were carried out between autumn 2020 and spring 2021 in a clinical sample (n = 280 patients aged 4-17 years) and a community sample (n = 1958 children and adolescents aged 4-19 years recruited via schools and preschools). Ratings of parents as well as children and adolescents via questionnaires were assessed. RESULTS: Mental burden due to the corona pandemic was assessed as slightly to moderately increased across both rating perspectives and both samples. Overall, around 60 to 70% of the parents, children, and adolescents describe an increase in mental burden; in contrast, up to 12% of parents as well as children and adolescents describe relief. When comparing both samples, a slightly higher burden on children and adolescents can only be seen in the self-assessment of the clinical sample. None of the socio-demographic factors analyzed influences the mental burden statistically significant. However, low to moderate correlations between the subjectively experienced deterioration in the family and social situation and an increased level of stress is found. DISCUSSION: Targeted interventions for exposed subgroups should be offered during a pandemic. Universal interventions are not indicated.


Subject(s)
COVID-19 , Problem Behavior , Adolescent , Child , Child, Preschool , Germany/epidemiology , Humans , Pandemics , SARS-CoV-2
4.
Brain Pathol ; 30(3): 641-652, 2020 05.
Article in English | MEDLINE | ID: mdl-31916298

ABSTRACT

Cortical demyelinated lesions are frequent and widespread in chronic multiple sclerosis (MS) patients, and may contribute to disease progression. Inflammation and related oxidative stress have been proposed as central mediators of cortical damage, yet meningeal and cortical inflammation is not specific to MS, but also occurs in other diseases. The first aim of this study was to test whether cortical demyelination was specific for demyelinating CNS diseases compared to other CNS disorders with prominent meningeal and cortical inflammation. The second aim was to assess whether oxidative tissue damage was associated with the extent of neuroaxonal damage. We studied a large cohort of patients diagnosed with demyelinating CNS diseases and non-demyelinating diseases of autoimmune, infectious, neoplastic or metabolic origin affecting the meninges and the cortex. Included were patients with MS, acute disseminated encephalomyelitis (ADEM), neuromyelitis optica (NMO), viral and bacterial meningoencephalitis, progressive multifocal leukoencephalopathy (PML), subacute sclerosing panencephalitis (SSPE), carcinomatous and lymphomatous meningitis and metabolic disorders such as extrapontine myelinolysis, thus encompassing a wide range of adaptive and innate cytokine signatures. Using myelin protein immunohistochemistry, we found cortical demyelination in MS, ADEM, PML and extrapontine myelinolysis, whereby each condition showed a disease-specific histopathological pattern. Remarkably, extensive ribbon-like subpial demyelination was only observed in MS, thus providing an important pathogenetic and diagnostic cue. Cortical oxidative injury was detected in both demyelinating and non-demyelinating CNS disorders. Our data demonstrate that meningeal and cortical inflammation alone accompanied by oxidative stress are not sufficient to generate the extensive subpial cortical demyelination found in MS, but require other MS-specific factors.


Subject(s)
Cerebral Cortex/pathology , Demyelinating Diseases/pathology , Meninges/pathology , Multiple Sclerosis/pathology , Myelin Sheath/pathology , Disease Progression , Humans , Inflammation/pathology
5.
J Neuroinflammation ; 16(1): 49, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30808363

ABSTRACT

BACKGROUND: Quinoline-3-carboxamides, such as laquinimod, ameliorate CNS autoimmunity in patients and reduce tumor cell metastasis experimentally. Previous studies have focused on the immunomodulatory effect of laquinimod on myeloid cells. The data contained herein suggest that quinoline-3-carboxamides improve the immunomodulatory and anti-tumor effects of NK cells by upregulating the adhesion molecule DNAX accessory molecule-1 (DNAM-1). METHODS: We explored how NK cell activation by laquinimod inhibits CNS autoimmunity in experimental autoimmune encephalomyelitis (EAE), the most utilized model of MS, and improves immunosurveillance of experimental lung melanoma metastasis. Functional manipulations included in vivo NK and DC depletion experiments and in vitro assays of NK cell function. Clinical, histological, and flow cytometric read-outs were assessed. RESULTS: We demonstrate that laquinimod activates natural killer (NK) cells via the aryl hydrocarbon receptor and increases their DNAM-1 cell surface expression. This activation improves the cytotoxicity of NK cells against B16F10 melanoma cells and augments their immunoregulatory functions in EAE by interacting with CD155+ dendritic cells (DC). Noteworthy, the immunosuppressive effect of laquinimod-activated NK cells was due to decreasing MHC class II antigen presentation by DC and not by increasing DC killing. CONCLUSIONS: This study clarifies how DNAM-1 modifies the bidirectional crosstalk of NK cells with CD155+ DC, which can be exploited to suppress CNS autoimmunity and strengthen tumor surveillance.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Autoimmunity/drug effects , Dendritic Cells/drug effects , Immunologic Surveillance/immunology , Killer Cells, Natural/drug effects , Quinolones/pharmacology , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Autoimmunity/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Quinolines/agonists , Receptors, Aryl Hydrocarbon/agonists , Receptors, Virus/immunology
6.
Glia ; 67(7): 1308-1319, 2019 07.
Article in English | MEDLINE | ID: mdl-30801815

ABSTRACT

Enhanced glial fibrillary acidic protein (GFAP) expression occurs in most diseases of the central nervous system. Thus far, little is known about the effect that GFAP exerts on astrocyte cell signaling. In the present study, we observed that silencing GFAP expression in isolated astrocytes leads to enhanced CCL2 and CXCL10 release, whereas overexpression of GFAP in astrocytes results in a significantly reduced CXCL10 release in vitro. Additionally, we analyzed transgenic mice carrying a full-length copy of the wild-type human GFAP gene. We demonstrate that a persistent GFAP increase alters the astrocytic cell signaling profile, thereby protecting oligodendrocytes, myelin and, subsequently, axons from cuprizone-induced demyelination. Our study revealed that reduced CXCL10 mRNA was accompanied by reduced NF-κB expression in astrocytes. Furthermore, analysis of human tissue from a patient with Alexander disease showed NF-κB activation in astrocytes to be almost completely absent. Our findings indicate that regulation of GFAP expression in astrocytes is crucial for astrocyte signaling and function. Understanding the role of the cytoskeletal protein, GFAP is thus of importance as it is highly regulated in diseases of the central nervous system.


Subject(s)
Astrocytes/metabolism , Chemokines/metabolism , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Glial Fibrillary Acidic Protein/biosynthesis , Adolescent , Animals , Animals, Newborn , Astrocytes/drug effects , Cells, Cultured , Chelating Agents/toxicity , Demyelinating Diseases/genetics , Female , Gene Expression Regulation , Glial Fibrillary Acidic Protein/genetics , Humans , Mice , Mice, Transgenic
7.
Neurosci Biobehav Rev ; 89: 85-98, 2018 06.
Article in English | MEDLINE | ID: mdl-29477616

ABSTRACT

Despite its functional importance and well known clinical impact in Multiple Sclerosis (MS), the cerebellum has only received significant attention over the past few years. It is now established that the cerebellum plays a key role not only in various sensory-motor networks, but also in cognitive-behavioural processes, domains primarily affected in patients with MS. Evidence from histopathological and magnetic resonance imaging (MRI) studies on cerebellar involvement in MS is increasingly available, however linking these pathological findings with clinical dysfunction remains challenging. There are promising advances in technology that are likely to improve the detection of pathological changes within the cerebellum, which may elucidate how pathology relates to disability.


Subject(s)
Cerebellum/pathology , Cognition Disorders/pathology , Cognition/physiology , Multiple Sclerosis/pathology , Animals , Cerebellum/physiopathology , Cognition Disorders/physiopathology , Humans , Magnetic Resonance Imaging/methods , Multiple Sclerosis/physiopathology , Neuropsychological Tests
8.
Clin Neurol Neurosurg ; 160: 137-141, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28734234

ABSTRACT

OBJECTIVES: The etiology and genetic susceptibility of primary central nervous system vasculitis (PCNSV) are still unclear. PATIENTS AND METHODS: We analyzed the DNA of 25 Caucasian patients with PCNSV for human leucocyte antigen genes HLA-A, HLA-B, HLA-DRB1, and HLA-DQB1, respectively. HLA-frequencies of the 25 patients with PCNSV were compared with HLA-frequencies of matched Caucasian controls. RESULTS: No statistically significant associations were found for HLA-B, HLA-DR1 and HLA-DQB1 variant. In the PCNSV group, only the HLA-A*69 variant was found more often than expected statistically. CONCLUSION: The results of this study indicate a potential association of HLA marker with PCNSV in Caucasian patients. Further studies are needed to elucidate the role of genes within the human major histocompatibility complex in the pathogenesis of this angiopathy.


Subject(s)
HLA-A Antigens/genetics , Histocompatibility Antigens Class I/genetics , Vasculitis, Central Nervous System/genetics , Adolescent , Adult , Aged , Child , Female , Germany , Humans , Male , Middle Aged , Vasculitis, Central Nervous System/diagnostic imaging , Young Adult
9.
Acta Neuropathol ; 134(1): 15-34, 2017 07.
Article in English | MEDLINE | ID: mdl-28386765

ABSTRACT

Cortical demyelination is a widely recognized hallmark of multiple sclerosis (MS) and correlate of disease progression and cognitive decline. The pathomechanisms initiating and driving gray matter damage are only incompletely understood. Here, we determined the infiltrating leukocyte subpopulations in 26 cortical demyelinated lesions of biopsied MS patients and assessed their contribution to cortical lesion formation in a newly developed mouse model. We find that conformation-specific anti-myelin antibodies contribute to cortical demyelination even in the absence of the classical complement pathway. T cells and natural killer cells are relevant for intracortical type 2 but dispensable for subpial type 3 lesions, whereas CCR2+ monocytes are required for both. Depleting CCR2+ monocytes in marmoset monkeys with experimental autoimmune encephalomyelitis using a novel humanized CCR2 targeting antibody translates into significantly less cortical demyelination and disease severity. We conclude that biologics depleting CCR2+ monocytes might be attractive candidates for preventing cortical lesion formation and ameliorating disease progression in MS.


Subject(s)
Cerebral Cortex/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Monocytes/immunology , Multiple Sclerosis/immunology , Adult , Animals , Callithrix , Cerebral Cortex/pathology , Cohort Studies , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Male , Meninges/immunology , Meninges/pathology , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Monocytes/pathology , Multiple Sclerosis/pathology , Random Allocation , Receptors, CCR2/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology
10.
Brain Pathol ; 26(4): 452-64, 2016 07.
Article in English | MEDLINE | ID: mdl-26207848

ABSTRACT

Multiple sclerosis (MS) is the most common cause for sustained disability in young adults, yet treatment options remain very limited. Although numerous therapeutic approaches have been effective in rodent models of experimental autoimmune encephalomyelitis (EAE), only few proved to be beneficial in patients with MS. Hence, there is a strong need for more predictive animal models. Within the past decade, EAE in the common marmoset evolved as a potent, alternative model for MS, with immunological and pathological features resembling more closely the human disease. However, an often very rapid and severe disease course hampers its implementation for systematic testing of new treatment strategies. We here developed a new focal model of EAE in the common marmoset, induced by myelin oligodendrocyte glycoprotein (MOG) immunization and stereotactic injections of proinflammatory cytokines. At the injection site of cytokines, confluent inflammatory demyelinating lesions developed that strongly resembled human MS lesions. In a proof-of-principle treatment study with the immunomodulatory compound laquinimod, we demonstrate that targeted EAE in marmosets provides a promising and valid tool for preclinical experimental treatment trials in MS research.


Subject(s)
Callithrix , Encephalomyelitis, Autoimmune, Experimental , Animals , Cytokines/administration & dosage , Cytokines/immunology , Female , Male , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology
11.
J Neuropathol Exp Neurol ; 74(8): 756-66, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26115190

ABSTRACT

Remyelination capacity decreases with age in adult mice, but data comparing remyelination capacity after toxic demyelination in developing mice versus adult mice are not available. We treated 3-week-old and adult C57BL/6 mice with cuprizone for 1 to 5 weeks and studied demyelination/remyelination and cellular reactions in the corpus callosum and motor cortex by histology, immunohistochemistry, and electron microscopy. We compared results between the 2 treated groups and age-matched controls. In juvenile mice, significant demyelination was detectable in the corpus callosum on Week 2 and in the motor cortex on Week 5. Oligodendrocyte loss, microglial activation, and acute axonal damage peaked on Week 2. Increased numbers of oligodendrocyte precursor cells were evident on Week 1, and remyelination was detectable on Week 3. Juvenile mice showed more rapid demyelination than adult mice, which may be related to greater vulnerability of oligodendrocytes, lower myelin content, or dose-dependent cuprizone effects. Earlier activation of microglia and proliferation of oligodendrocyte precursor cells probably contributed to accelerated remyelination and less pronounced axonal damage. Our data indicate that oligodendroglial regeneration and remyelination are enhanced in the maturing rodent brain compared with the young-adult rodent brain.


Subject(s)
Aging/pathology , Brain/pathology , Demyelinating Diseases/pathology , Nerve Regeneration/physiology , Animals , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Monoamine Oxidase Inhibitors/toxicity , Oligodendroglia/pathology
12.
Brain Pathol ; 25(3): 276-86, 2015 May.
Article in English | MEDLINE | ID: mdl-25041171

ABSTRACT

Subpial cortical demyelination (SCD) accounts for the greatest proportion of demyelinated cortex in multiple sclerosis (MS). SCD is already found in biopsy cases with early MS and in marmosets with experimental autoimmune encephalomyelitis (EAE), but the pathogenesis of SCD is not well understood. The objective of this study was to investigate whether and, if so, which meningeal inflammatory cells were associated with early SCD in marmosets with EAE. Immunohistochemistry was performed to analyze brain samples from eight control animals and eight marmosets immunized with myelin oligodendrocyte glycoprotein. Meningeal T, B and plasma cells were quantified adjacent to SCD, normal-appearing EAE cortex (NAC) and control marmoset cortex. SCD areas appeared mostly hypocellular with low-grade microglial activation. In marmosets with EAE, meninges adjacent to SCD showed significantly increased T cells paralleled by elevated plasma cells, but unaltered B cell numbers compared with NAC. The elevation of meningeal T and plasma cells was a specific finding topographically associated with SCD, as the meninges overlying NAC displayed similarly low T, B and plasma cell numbers as control cortex. These findings suggest that local meningeal T and plasma cell infiltration contributes to the pathogenesis of SCD in marmosets with EAE.


Subject(s)
Cerebral Cortex/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Meninges/pathology , Multiple Sclerosis/pathology , Plasma Cells/pathology , T-Lymphocytes/pathology , Animals , Antigens, CD/metabolism , Calgranulin B/metabolism , Callithrix , Case-Control Studies , Disease Models, Animal , Female , Male , Myelin Basic Protein/metabolism , Plasma Cells/metabolism , T-Lymphocytes/metabolism , White Matter/pathology
13.
Acta Neuropathol ; 128(2): 231-46, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24563023

ABSTRACT

Cerebral cortex shows a high endogenous propensity for remyelination. Yet, widespread subpial cortical demyelination (SCD) is a common feature in progressive multiple sclerosis (MS) and can already be found in early MS. In the present study, we compared oligodendroglial loss in SCD in early and chronic MS. Furthermore, we addressed in an experimental model whether repeated episodes of inflammatory SCD could alter oligodendroglial repopulation and subsequently lead to persistently demyelinated cortical lesions. NogoA(+) mature oligodendrocytes and Olig2(+) oligodendrocyte precursor cells were examined in SCD in patients with early and chronic MS, normal-appearing MS cortex, and control cortex as well as in the rat model of repeated targeted cortical experimental autoimmune encephalomyelitis (EAE). NogoA(+) and Olig2(+) cells were significantly reduced in SCD in patients with chronic, but not early MS. Repeated induction of SCD in rats resulted only in a transient loss of NogoA(+), but not Olig2(+) cells during the demyelination phase. This phase was followed by complete oligodendroglial repopulation and remyelination, even after four episodes of demyelination. Our data indicate efficient oligodendroglial repopulation in subpial cortical lesions in rats after repeated SCD that was similar to early, but not chronic MS cases. Accordingly, four cycles of experimental de- and remyelination were not sufficient to induce sustained remyelination failure as found in chronic cortical MS lesions. This suggests that alternative mechanisms contribute to oligodendrocyte depletion in chronic cortical demyelination in MS.


Subject(s)
Cerebral Cortex/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis/pathology , Oligodendroglia/pathology , Adult , Aged , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cerebral Cortex/physiopathology , Chronic Disease , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Humans , Male , Middle Aged , Multiple Sclerosis/physiopathology , Myelin Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nogo Proteins , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/physiology , Rats, Inbred Lew
14.
Acta Neuropathol ; 127(4): 523-38, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24292009

ABSTRACT

Neuromyelitis optica (NMO) is a chronic, mostly relapsing inflammatory demyelinating disease of the CNS characterized by serum anti-aquaporin 4 (AQP4) antibodies in the majority of patients. Anti-AQP4 antibodies derived from NMO patients target and deplete astrocytes in experimental models when co-injected with complement. However, the time course and mechanisms of oligodendrocyte loss and demyelination and the fate of oligodendrocyte precursor cells (OPC) have not been examined in detail. Also, no studies regarding astrocyte repopulation of experimental NMO lesions have been reported. We utilized two rat models using either systemic transfer or focal intracerebral injection of recombinant human anti-AQP4 antibodies to generate NMO-like lesions. Time-course experiments were performed to examine oligodendroglial and astroglial damage and repair. In addition, oligodendrocyte pathology was studied in early human NMO lesions. Apart from early complement-mediated astrocyte destruction, we observed a prominent, very early loss of oligodendrocytes and oligodendrocyte precursor cells (OPCs) as well as a delayed loss of myelin. Astrocyte repopulation of focal NMO lesions was already substantial after 1 week. Olig2-positive OPCs reappeared before NogoA-positive, mature oligodendrocytes. Thus, using two experimental models that closely mimic the human disease, our study demonstrates that oligodendrocyte and OPC loss is an extremely early feature in the formation of human and experimental NMO lesions and leads to subsequent, delayed demyelination, highlighting an important difference in the pathogenesis of MS and NMO.


Subject(s)
Brain/pathology , Neuromyelitis Optica/pathology , Oligodendroglia/pathology , Spinal Cord/pathology , Adult , Aged , Animals , Aquaporin 4/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Male , Middle Aged , Myelin Basic Protein/metabolism , Myelin Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nogo Proteins , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , Rats, Inbred Lew
15.
Clin Neurol Neurosurg ; 115 Suppl 1: S38-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24321153

ABSTRACT

Multiple sclerosis (MS) is the most common inflammatory demyelinating disease of the central nervous system. Traditionally, demyelinating lesions in the white matter have been regarded as the most important pathological feature in MS, but recent pathological and imaging studies confirmed substantial changes in grey matter and normal-appearing white matter. MS lesions are characterized by inflammation, demyelination, axonal damage and astrogliosis. During early MS lesion formation acute axonal injury is extensive and correlates with inflammation. In addition to focal lesions, diffuse wide-spread changes including neuroaxonal degeneration and compartmentalized inflammation are likely to contribute to increasing disability in progressive MS. Neuromyelitis optica (NMO) is classically characterized by severe transverse myelitis and optic neuritis, but brain lesions are also present in the majority of NMO patients. The discovery of the NMO-specific antibody demonstrated that NMO is a disease entity distinct from MS. This antibody binds to aquaporin-4 expressed in astrocytes and ependymal cells. NMO lesions are characterized by inflammation, demyelination, axonal damage and a marked loss of aquaporin-4. Early NMO lesions demonstrate a pronounced humoral inflammatory response and astrocytic cell death with loss of aquaporin-4, followed by inflammatory demyelination and axonal damage. These recent findings contribute to a better understanding of different mechanisms leading to inflammatory demyelination.


Subject(s)
Axons/pathology , Brain/pathology , Multiple Sclerosis/pathology , Neuromyelitis Optica/pathology , Spinal Cord/pathology , Animals , Aquaporin 4/immunology , Aquaporin 4/metabolism , Axons/metabolism , Brain/immunology , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Neuromyelitis Optica/immunology , Neuromyelitis Optica/metabolism , Spinal Cord/immunology
16.
Acta Neuropathol ; 124(3): 411-24, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22766690

ABSTRACT

Laquinimod (LAQ) is a new oral immunomodulatory compound that reduces relapse rate, brain atrophy and disability progression in multiple sclerosis (MS). LAQ has well-documented effects on inflammation in the periphery, but little is known about its direct activity within the central nervous system (CNS). To elucidate the impact of LAQ on CNS-intrinsic inflammation, we investigated the effects of LAQ on cuprizone-induced demyelination in mice in vivo and on primary CNS cells in vitro. Demyelination, inflammation, axonal damage and glial pathology were evaluated in LAQ-treated wild type and Rag-1-deficient mice after cuprizone challenge. Using primary cells we tested for effects of LAQ on oligodendroglial survival as well as on cytokine secretion and NF-κB activation in astrocytes and microglia. LAQ prevented cuprizone-induced demyelination, microglial activation, axonal transections, reactive gliosis and oligodendroglial apoptoses in wild type and Rag-1-deficient mice. LAQ significantly decreased pro-inflammatory factors in stimulated astrocytes, but not in microglia. Oligodendroglial survival was not affected by LAQ in vitro. Astrocytic, but not microglial, NF-κB activation was markedly reduced by LAQ as evidenced by NF-κB reporter assay. LAQ also significantly decreased astrocytic NF-κB activation in cuprizone-treated mice. Our data indicate that LAQ prevents cuprizone-induced demyelination by attenuating astrocytic NF-κB activation. These effects are CNS-intrinsic and not mediated by peripheral immune cells. Therefore, LAQ downregulation of the astrocytic pro-inflammatory response may be an important mechanism underlying its protective effects on myelin, oligodendrocytes and axons. Modulation of astrocyte activation may be an attractive therapeutic target to prevent tissue damage in MS.


Subject(s)
Astrocytes/drug effects , Demyelinating Diseases/prevention & control , NF-kappa B/metabolism , Oligodendroglia/drug effects , Quinolones/pharmacology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Axons/drug effects , Axons/metabolism , Axons/pathology , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Male , Mice , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Myelin Sheath/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology
17.
Mult Scler ; 18(7): 1045-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22086900

ABSTRACT

We report a case of multiple sclerosis-associated fulminant tumefactive demyelinating lesion (TDL) with the special feature of delayed humoral immune response. Plasma exchange (PE) yielded significant benefit in two consecutive steroid-unresponsive relapses, while signs of an intrathecal B-cell response were only present 2 years later at the second relapse. Remission was achieved and sustained thereafter with natalizumab. Our case indicates that PE might be a therapeutic option even when the B-cell response is not fully developed. This delay in the development of a humoral immune response may reflect the step-wise B-cell colonization of the CNS and represent an attractive therapeutic window of opportunity.


Subject(s)
Brain/pathology , Multiple Sclerosis/complications , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Plasma Exchange , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , B-Lymphocytes/immunology , Demyelinating Diseases/etiology , Demyelinating Diseases/immunology , Demyelinating Diseases/therapy , Disease Progression , Flow Cytometry , Humans , Magnetic Resonance Imaging , Male , Multiple Sclerosis/therapy , Natalizumab , T-Lymphocytes/immunology
18.
Eur J Neurosci ; 33(7): 1256-63, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21375601

ABSTRACT

In this multicentre study involving eight European centres, we characterized the spatial pattern of functional connectivity (FC) in the sensorimotor network from 61 right-handed patients with multiple sclerosis (MS) and 74 age-matched healthy subjects assessed with the use of functional magnetic resonance imaging (fMRI) and a simple motor task of their right dominant hand. FC was investigated by using: (i) voxel-wise correlations between the left sensorimotor cortex (SMC) and any other area in the brain; and (ii) bivariate correlations between time series extracted from several regions of interest (ROIs) belonging to the sensorimotor network. Both healthy controls and MS patients had significant FC between the left SMC and several areas of the sensorimotor network, including the bilateral postcentral and precentral gyri, supplementary motor area, middle frontal gyri, insulae, secondary somatosensory cortices, thalami, and right cerebellum. Voxel-wise assessment of FC revealed increased connectivity between the left SMC and the right precentral gyrus, right middle frontal gyrus (MFG) and bilateral postcentral gyri in MS patients as compared with controls. ROI analysis also showed a widespread pattern of altered connectivity, characterized by increased FC between the right MFG, the left insula and the right inferior frontal gyrus in comparison with many regions of the sensorimotor network. These results provide further evidence for increased bihemispheric contributions to motor control in patients with MS relative to healthy controls. They further suggest that multicentre fMRI studies of FC changes are possible, and provide a potential imaging biomarker for use in experimental therapeutic studies directed at enhancing adaptive plasticity in the disease.


Subject(s)
Motor Activity/physiology , Multiple Sclerosis/physiopathology , Nerve Net/physiology , Nerve Net/physiopathology , Neural Pathways/physiopathology , Adult , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis/pathology , Nerve Net/pathology , Neural Pathways/pathology , Neural Pathways/physiology
19.
Biochim Biophys Acta ; 1812(2): 275-82, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20637864

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system which responds to anti-inflammatory treatments in the early disease phase. However, the pathogenesis of the progressive disease phase is less well understood, and inflammatory as well as neurodegenerative mechanisms of tissue damage are currently being discussed. This review summarizes current knowledge on the interrelation between inflammation, demyelination, and neurodegeneration derived from the study of human autopsy and biopsy brain tissue and experimental models of MS.


Subject(s)
Multiple Sclerosis/pathology , Animals , Autoantigens , Autoimmunity , Axons/pathology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Multiple Sclerosis/etiology , Multiple Sclerosis/immunology , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Neurons/pathology
20.
J Neuroimmunol ; 227(1-2): 133-43, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20684995

ABSTRACT

We investigated the effect of laquinimod on inflammatory demyelination, axonal damage, cytokine profiles and migratory capacities of lymphocytes in C57BL/6 mice with active EAE induced with MOG(35-55) peptide. The mice were treated at disease induction and after disease onset. Spinal cords were assessed histologically. Cytokines and adhesive properties were analyzed in splenocytes. Preventive and therapeutic laquinimod treatment reduced clinical signs, inflammation, and demyelination. VLA-4-mediated adhesiveness and pro-inflammatory cytokines such as IL-17 were down-regulated in treated animals. Within lesions, treated mice showed similar axonal densities, but less acute axonal damage than controls. Laquinimod might thus protect myelin and axons by decreasing pro-inflammatory cytokines and impairing the migratory capacity of lymphocytes.


Subject(s)
Axons/pathology , Cell Movement/immunology , Demyelinating Diseases/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Inflammation Mediators/therapeutic use , Interleukin-17/antagonists & inhibitors , Quinolones/therapeutic use , T-Lymphocyte Subsets/pathology , Animals , Axons/drug effects , Axons/immunology , Cell Movement/drug effects , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Down-Regulation/drug effects , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...