Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Neuropathol Appl Neurobiol ; 49(5): e12935, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37705188

RESUMEN

AIMS: Fibroblast growth factor (FGF) signalling is dysregulated in multiple sclerosis (MS) and other neurological and psychiatric conditions, but there is little or no consensus as to how individual FGF family members contribute to disease pathogenesis. Lesion development in MS is associated with increased expression of FGF1, FGF2 and FGF9, all of which modulate remyelination in a variety of experimental settings. However, FGF9 is also selectively upregulated in major depressive disorder (MDD), prompting us to speculate it may also have a direct effect on neuronal function and survival. METHODS: Transcriptional profiling of myelinating cultures treated with FGF1, FGF2 or FGF9 was performed, and the effects of FGF9 on cortical neurons investigated using a combination of transcriptional, electrophysiological and immunofluorescence microscopic techniques. The in vivo effects of FGF9 were explored by stereotactic injection of adeno-associated viral (AAV) vectors encoding either FGF9 or EGFP into the rat motor cortex. RESULTS: Transcriptional profiling of myelinating cultures after FGF9 treatment revealed a distinct neuronal response with a pronounced downregulation of gene networks associated with axonal transport and synaptic function. In cortical neuronal cultures, FGF9 also rapidly downregulated expression of genes associated with synaptic function. This was associated with a complete block in the development of photo-inducible spiking activity, as demonstrated using multi-electrode recordings of channel rhodopsin-transfected rat cortical neurons in vitro and, ultimately, neuronal cell death. Overexpression of FGF9 in vivo resulted in rapid loss of neurons and subsequent development of chronic grey matter lesions with neuroaxonal reduction and ensuing myelin loss. CONCLUSIONS: These observations identify overexpression of FGF9 as a mechanism by which neuroaxonal pathology could develop independently of immune-mediated demyelination in MS. We suggest targeting neuronal FGF9-dependent pathways may provide a novel strategy to slow if not halt neuroaxonal atrophy and loss in MS, MDD and potentially other neurodegenerative diseases.


Asunto(s)
Trastorno Depresivo Mayor , Esclerosis Múltiple , Animales , Ratas , Factor 1 de Crecimiento de Fibroblastos , Factor 2 de Crecimiento de Fibroblastos , Factor 9 de Crecimiento de Fibroblastos
2.
Int J Mol Sci ; 24(18)2023 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-37762198

RESUMEN

Modeling chronic cortical demyelination allows the study of long-lasting pathological changes observed in multiple sclerosis such as failure of remyelination, chronically disturbed functions of oligodendrocytes, neurons and astrocytes, brain atrophy and cognitive impairments. We aimed at generating an animal model for studying the consequences of chronic cortical demyelination and meningeal inflammation. To induce long-lasting cortical demyelination and chronic meningeal inflammation, we immunized female Lewis rats against myelin oligodendrocyte glycoprotein (MOG) and injected lentiviruses for continuing overexpression of the cytokines TNFα and IFNγ in the cortical brain parenchyma. Immunization with MOG and overexpression of TNFα and IFNγ led to widespread subpial demyelination and meningeal inflammation that were stable for at least 10 weeks. We demonstrate here that immunization with MOG is necessary for acute as well as chronic cortical demyelination. In addition, long-lasting overexpression of TNFα and IFNγ in the brain parenchyma is sufficient to induce chronic meningeal inflammation. Our model simulates key features of chronic cortical demyelination and inflammation, reminiscent of human multiple sclerosis pathology. This will allow molecular, cellular and functional investigations for a better understanding of the adaptation mechanisms of the cerebral cortex in multiple sclerosis.


Asunto(s)
Esclerosis Múltiple , Factor de Necrosis Tumoral alfa , Ratas , Animales , Humanos , Femenino , Ratas Endogámicas Lew , Factor de Necrosis Tumoral alfa/genética , Modelos Animales , Glicoproteína Mielina-Oligodendrócito , Corteza Cerebral , Inflamación
3.
Glia ; 67(6): 1196-1209, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30980503

RESUMEN

X-linked adrenoleukodystrophy (X-ALD) and metachromatic leukodystrophy (MLD) are two relatively common examples of hereditary demyelinating diseases caused by a dysfunction of peroxisomal or lysosomal lipid degradation. In both conditions, accumulation of nondegraded lipids leads to the destruction of cerebral white matter. Because of their high lipid content, oligodendrocytes are considered key to the pathophysiology of these leukodystrophies. However, the response to allogeneic stem cell transplantation points to the relevance of cells related to the hematopoietic lineage. In the present study, we aimed to better characterize the pathogenetic role of microglia in the above-mentioned diseases. Applying recently established microglia markers to human autopsy cases of X-ALD and MLD we were able to delineate distinct lesion stages in evolving demyelinating lesions. The immune-phenotype of microglia was altered already early in lesion evolution, and microglia loss preceded full-blown myelin degeneration both in X-ALD and MLD. DNA fragmentation indicating phagocyte death was observed in areas showing microglia loss. The morphology and dynamics of phagocyte decay differed between the diseases and between lesion stages, hinting at distinct pathways of programmed cell death. In summary, the present study shows an early and severe damage to microglia in the pathogenesis of X-ALD and MLD. This hints at a central pathophysiologic role of these cells in the diseases and provides evidence for an ongoing transfer of toxic substrates primarily enriched in myelinating cells to microglia.


Asunto(s)
Adrenoleucodistrofia/patología , Leucodistrofia Metacromática/patología , Microglía/patología , Vaina de Mielina/patología , Adolescente , Adrenoleucodistrofia/genética , Adrenoleucodistrofia/metabolismo , Adulto , Anciano , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Masculino , Microglía/metabolismo , Persona de Mediana Edad , Vaina de Mielina/genética , Vaina de Mielina/metabolismo
4.
Glia ; 65(8): 1350-1360, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28560740

RESUMEN

Remyelination is in the center of new therapies for the treatment of multiple sclerosis to resolve and improve disease symptoms and protect axons from further damage. Although remyelination is considered beneficial in the long term, it is not known, whether this is also the case early in lesion formation. Additionally, the precise timing of acute axonal damage and remyelination has not been assessed so far. To shed light onto the interrelation between axons and the myelin sheath during de- and remyelination, we employed cuprizone- and focal lysolecithin-induced demyelination and performed time course experiments assessing the evolution of early and late stage remyelination and axonal damage. We observed damaged axons with signs of remyelination after cuprizone diet cessation and lysolecithin injection. Similar observations were made in early multiple sclerosis lesions. To assess the correlation of remyelination and axonal damage in multiple sclerosis lesions, we took advantage of a cohort of patients with early and late stage remyelinated lesions and assessed the number of APP- and SMI32- positive damaged axons and the density of SMI31-positive and silver impregnated preserved axons. Early de- and remyelinating lesions did not differ with respect to axonal density and axonal damage, but we observed a lower axonal density in late stage demyelinated multiple sclerosis lesions than in remyelinated multiple sclerosis lesions. Our findings suggest that remyelination may not only be protective over a long period of time, but may play an important role in the immediate axonal recuperation after a demyelinating insult.


Asunto(s)
Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Microglía/patología , Esclerosis Múltiple/patología , Vaina de Mielina/patología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Axones/patología , Cuprizona/toxicidad , Enfermedades Desmielinizantes/inducido químicamente , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Lisofosfatidilcolinas/toxicidad , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Inhibidores de la Monoaminooxidasa/toxicidad , Esclerosis Múltiple/inducido químicamente , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Ratas , Ratas Endogámicas Lew , Estadísticas no Paramétricas , Factores de Tiempo
5.
Acta Neuropathol ; 134(1): 15-34, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28386765

RESUMEN

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.


Asunto(s)
Corteza Cerebral/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Monocitos/inmunología , Esclerosis Múltiple/inmunología , Adulto , Animales , Callithrix , Corteza Cerebral/patología , Estudios de Cohortes , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Masculino , Meninges/inmunología , Meninges/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Monocitos/patología , Esclerosis Múltiple/patología , Distribución Aleatoria , Receptores CCR2/metabolismo , Linfocitos T/inmunología , Linfocitos T/patología
6.
Glia ; 64(4): 635-49, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26683584

RESUMEN

Microglia, innate immune cells of the CNS, sense infection and damage through overlapping receptor sets. Toll-like receptor (TLR) 4 recognizes bacterial lipopolysaccharide (LPS) and multiple injury-associated factors. We show that its co-receptor CD14 serves three non-redundant functions in microglia. First, it confers an up to 100-fold higher LPS sensitivity compared to peripheral macrophages to enable efficient proinflammatory cytokine induction. Second, CD14 prevents excessive responses to massive LPS challenges via an interferon ß-mediated feedback. Third, CD14 is mandatory for microglial reactions to tissue damage-associated signals. In mice, these functions are essential for balanced CNS responses to bacterial infection, traumatic and ischemic injuries, since CD14 deficiency causes either hypo- or hyperinflammation, insufficient or exaggerated immune cell recruitment or worsened stroke outcomes. While CD14 orchestrates functions of TLR4 and related immune receptors, it is itself regulated by TLR and non-TLR systems to thereby fine-tune microglial damage-sensing capacity upon infectious and non-infectious CNS challenges.


Asunto(s)
Lesiones Encefálicas/inmunología , Isquemia Encefálica/inmunología , Infecciones por Escherichia coli/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Microglía/inmunología , Accidente Cerebrovascular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Encéfalo/inmunología , Encéfalo/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Isquemia Encefálica/patología , Células Cultivadas , Modelos Animales de Enfermedad , Escherichia coli , Infecciones por Escherichia coli/complicaciones , Infecciones por Escherichia coli/patología , Retroalimentación Fisiológica/fisiología , Infarto de la Arteria Cerebral Media , Interferón beta/metabolismo , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación , Accidente Cerebrovascular/patología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
7.
Acta Neuropathol ; 127(4): 523-38, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24292009

RESUMEN

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.


Asunto(s)
Encéfalo/patología , Neuromielitis Óptica/patología , Oligodendroglía/patología , Médula Espinal/patología , Adulto , Anciano , Animales , Acuaporina 4/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteína Básica de Mielina/metabolismo , Proteínas de la Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nogo , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Ratas Endogámicas Lew
8.
Neuroimage ; 59(3): 2678-88, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21914485

RESUMEN

Magnetic resonance imaging (MRI) is the gold standard for the detection of multiple sclerosis (MS) lesions. However, current MRI techniques provide little information about the structural features of a brain lesion with inflammatory cell infiltration, demyelination, gliosis, acute axonal damage and axonal loss. To identify methods for a differentiation of demyelination, inflammation, and axonal damage we developed a novel mouse model combining cuprizone-induced demyelination and experimental autoimmune encephalomyelitis. MS-like brain lesions were assessed by T1-weighted, T2-weighted, and magnetization transfer MRI as well as by diffusion tensor imaging (DTI). T2-weighted MRI differentiated control and diseased mice, while T1-weighted MRI better reflected the extent of inflammation and axonal damage. In DTI, axonal damage and cellular infiltration led to a reduction of the axial diffusivity, whereas primary demyelination after cuprizone treatment was reflected by changes in radial but not axial diffusivity. Importantly, alterations in radial diffusivity were less pronounced in mice with demyelination, inflammation, and acute axonal damage, indicating that radial diffusivity may underestimate demyelination in acute MS lesions. In conclusion, the combined information from different DTI parameters allows for a more precise identification of solely demyelinated lesions versus demyelinated and acutely inflamed lesions. These findings are of relevance for offering individualized, stage-adapted therapies for MS patients.


Asunto(s)
Encéfalo/patología , Imagen de Difusión Tensora/métodos , Encefalomielitis Autoinmune Experimental/patología , Imagen por Resonancia Magnética/métodos , Esclerosis Múltiple/patología , Animales , Axones/patología , Barrera Hematoencefálica/patología , Cuprizona , Enfermedades Desmielinizantes/patología , Análisis Discriminante , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Gliosis/patología , Procesamiento de Imagen Asistido por Computador/métodos , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Inhibidores de la Monoaminooxidasa , Esclerosis Múltiple/inducido químicamente , Toxina del Pertussis/toxicidad
9.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33645550

RESUMEN

Approximately 80% of neuromyelitis optica spectrum disorder (NMOSD) patients harbor serum anti-aquaporin-4 autoantibodies targeting astrocytes in the CNS. Crucial for NMOSD lesion initiation is disruption of the blood-brain barrier (BBB), which allows the entrance of Abs and serum complement into the CNS and which is a target for new NMOSD therapies. Astrocytes have important functions in BBB maintenance; however, the influence of their loss and the role of immune cell infiltration on BBB permeability in NMOSD have not yet been investigated. Using an experimental model of targeted NMOSD lesions in rats, we demonstrate that astrocyte destruction coincides with a transient disruption of the BBB and a selective loss of occludin from tight junctions. It is noteworthy that BBB integrity is reestablished before astrocytes repopulate. Rather than persistent astrocyte loss, polymorphonuclear leukocytes (PMNs) are the main mediators of BBB disruption, and their depletion preserves BBB integrity and prevents astrocyte loss. Inhibition of PMN chemoattraction, activation, and proteolytic function reduces lesion size. In summary, our data support a crucial role for PMNs in BBB disruption and NMOSD lesion development, rendering their recruitment and activation promising therapeutic targets.


Asunto(s)
Astrocitos/inmunología , Barrera Hematoencefálica/inmunología , Leucocitos Mononucleares/inmunología , Neuromielitis Óptica/inmunología , Animales , Astrocitos/patología , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Leucocitos Mononucleares/patología , Neuromielitis Óptica/patología , Ratas , Ratas Endogámicas Lew
10.
Sci Transl Med ; 9(419)2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29212715

RESUMEN

Investigations into brain function and disease depend on the precise classification of neural cell types. Cells of the oligodendrocyte lineage differ greatly in their morphology, but accurate identification has thus far only been possible for oligodendrocyte progenitor cells and mature oligodendrocytes in humans. We find that breast carcinoma amplified sequence 1 (BCAS1) expression identifies an oligodendroglial subpopulation in the mouse and human brain. These cells are newly formed, myelinating oligodendrocytes that segregate from oligodendrocyte progenitor cells and mature oligodendrocytes and mark regions of active myelin formation in development and in the adult. We find that BCAS1+ oligodendrocytes are restricted to the fetal and early postnatal human white matter but remain in the cortical gray matter until old age. BCAS1+ oligodendrocytes are reformed after experimental demyelination and found in a proportion of chronic white matter lesions of patients with multiple sclerosis (MS) even in a subset of patients with advanced disease. Our work identifies a means to map ongoing myelin formation in health and disease and presents a potential cellular target for remyelination therapies in MS.


Asunto(s)
Esclerosis Múltiple/metabolismo , Proteínas de Neoplasias/metabolismo , Oligodendroglía/metabolismo , Animales , Enfermedades Desmielinizantes , Humanos , Ratones , Esclerosis Múltiple/patología , Vaina de Mielina/metabolismo
11.
Ann Clin Transl Neurol ; 4(11): 768-783, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29159189

RESUMEN

Objective: Autoimmune encephalitis is most frequently associated with anti-NMDAR autoantibodies. Their pathogenic relevance has been suggested by passive transfer of patients' cerebrospinal fluid (CSF) in mice in vivo. We aimed to analyze the intrathecal plasma cell repertoire, identify autoantibody-producing clones, and characterize their antibody signatures in recombinant form. Methods: Patients with recent onset typical anti-NMDAR encephalitis were subjected to flow cytometry analysis of the peripheral and intrathecal immune response before, during, and after immunotherapy. Recombinant human monoclonal antibodies (rhuMab) were cloned and expressed from matching immunoglobulin heavy- (IgH) and light-chain (IgL) amplicons of clonally expanded intrathecal plasma cells (cePc) and tested for their pathogenic relevance. Results: Intrathecal accumulation of B and plasma cells corresponded to the clinical course. The presence of cePc with hypermutated antigen receptors indicated an antigen-driven intrathecal immune response. Consistently, a single recombinant human GluN1-specific monoclonal antibody, rebuilt from intrathecal cePc, was sufficient to reproduce NMDAR epitope specificity in vitro. After intraventricular infusion in mice, it accumulated in the hippocampus, decreased synaptic NMDAR density, and caused severe reversible memory impairment, a key pathogenic feature of the human disease, in vivo. Interpretation: A CNS-specific humoral immune response is present in anti-NMDAR encephalitis specifically targeting the GluN1 subunit of the NMDAR. Using reverse genetics, we recovered the typical intrathecal antibody signature in recombinant form, and proved its pathogenic relevance by passive transfer of disease symptoms from man to mouse, providing the critical link between intrathecal immune response and the pathogenesis of anti-NMDAR encephalitis as a humorally mediated autoimmune disease.

12.
Cell Rep ; 16(2): 314-322, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27346352

RESUMEN

Breakdown of myelin sheaths is a pathological hallmark of several autoimmune diseases of the nervous system. We employed autoantibody-mediated animal models of demyelinating diseases, including a rat model of neuromyelitis optica (NMO), to target myelin and found that myelin lamellae are broken down into vesicular structures at the innermost region of the myelin sheath. We demonstrated that myelin basic proteins (MBP), which form a polymer in between the myelin membrane layers, are targeted in these models. Elevation of intracellular Ca(2+) levels resulted in MBP network disassembly and myelin vesiculation. We propose that the aberrant phase transition of MBP molecules from their cohesive to soluble and non-adhesive state is a mechanism triggering myelin breakdown in NMO and possibly in other demyelinating diseases.


Asunto(s)
Proteína Básica de Mielina/metabolismo , Vaina de Mielina/patología , Neuromielitis Óptica/metabolismo , Animales , Señalización del Calcio , Modelos Animales de Enfermedad , Neuromielitis Óptica/patología , Ratas Endogámicas Lew
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA