Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Neurobiol Dis ; 156: 105418, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34102276

RESUMEN

Cystatin B (CSTB) acts as an inhibitor of cysteine proteases of the cathepsin family and loss-of-function mutations result in human brain diseases with a genotype-phenotype correlation. In the most severe case, CSTB-deficiency disrupts brain development, and yet the molecular basis of this mechanism is missing. Here, we establish CSTB as a regulator of chromatin structure during neural stem cell renewal and differentiation. Murine neural precursor cells (NPCs) undergo transient proteolytic cleavage of the N-terminal histone H3 tail by cathepsins B and L upon induction of differentiation into neurons and glia. In contrast, CSTB-deficiency triggers premature H3 tail cleavage in undifferentiated self-renewing NPCs and sustained H3 tail proteolysis in differentiating neural cells. This leads to significant transcriptional changes in NPCs, particularly of nuclear-encoded mitochondrial genes. In turn, these transcriptional alterations impair the enhanced mitochondrial respiration that is induced upon neural stem cell differentiation. Collectively, our findings reveal the basis of epigenetic regulation in the molecular pathogenesis of CSTB deficiency.


Asunto(s)
Cistatina B/deficiencia , Histonas/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Células Cultivadas , Cistatina B/genética , Epigénesis Genética/fisiología , Histonas/genética , Ratones , Ratones de la Cepa 129 , Ratones Noqueados
2.
Epilepsia ; 61(11): 2593-2608, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32940364

RESUMEN

OBJECTIVE: Microglial phagocytosis of apoptotic cells is an essential component of the brain regenerative response during neurodegeneration. Whereas it is very efficient in physiological conditions, it is impaired in mouse and human mesial temporal lobe epilepsy, and now we extend our studies to a model of progressive myoclonus epilepsy type 1 in mice lacking cystatin B (CSTB). METHODS: We used confocal imaging and stereology-based quantification of apoptosis and phagocytosis of the hippocampus of Cstb knockout (KO) mice, an in vitro model of phagocytosis and siRNAs to acutely reduce Cstb expression, and a virtual three-dimensional (3D) model to analyze the physical relationship between apoptosis, phagocytosis, and active hippocampal neurons. RESULTS: Microglial phagocytosis was impaired in the hippocampus of Cstb KO mice at 1 month of age, when seizures arise and hippocampal atrophy begins. This impairment was not related to the lack of Cstb in microglia alone, as shown by in vitro experiments with microglial Cstb depletion. The phagocytosis impairment was also unrelated to seizures, as it was also present in Cstb KO mice at postnatal day 14, before seizures begin. Importantly, phagocytosis impairment was restricted to the granule cell layer and spared the subgranular zone, where there are no active neurons. Furthermore, apoptotic cells (both phagocytosed and not phagocytosed) in Cstb-deficient mice were at close proximity to active cFos+ neurons, and a virtual 3D model demonstrated that the physical relationship between apoptotic cells and cFos+ neurons was specific for Cstb KO mice. SIGNIFICANCE: These results suggest a complex crosstalk between apoptosis, phagocytosis, and neuronal activity, hinting that local neuronal activity could be related to phagocytosis dysfunction in Cstb KO mice. Overall, these data suggest that phagocytosis impairment is an early feature of hippocampal damage in epilepsy and opens novel therapeutic approaches for epileptic patients based on targeting microglial phagocytosis.


Asunto(s)
Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Microglía/metabolismo , Neuronas/metabolismo , Fagocitosis/fisiología , Síndrome de Unverricht-Lundborg/metabolismo , Animales , Cistatina B/deficiencia , Cistatina B/genética , Giro Dentado/patología , Ratones , Ratones Noqueados , Microglía/patología , Neuronas/patología , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/patología
3.
J Neuroinflammation ; 13(1): 298, 2016 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-27894304

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal recessively inherited childhood-onset neurodegenerative disorder, characterized by myoclonus, seizures, and ataxia. Mutations in the cystatin B gene (CSTB) underlie EPM1. The CSTB-deficient (Cstb -/- ) mouse model recapitulates key features of EPM1, including myoclonic seizures. The mice show early microglial activation that precedes seizure onset and neuronal loss and leads to neuroinflammation. We here characterized the inflammatory phenotype of Cstb -/- mice in more detail. We found higher concentrations of chemokines and pro-inflammatory cytokines in the serum of Cstb -/- mice and higher CXCL13 expression in activated microglia in Cstb -/- compared to control mouse brains. The elevated chemokine levels were not accompanied by blood-brain barrier disruption, despite increased brain vascularization. Macrophages in the spleen and brain of Cstb -/- mice were predominantly pro-inflammatory. Taken together, these data show that CXCL13 expression is a hallmark of microglial activation in Cstb -/- mice and that the brain inflammation is linked to peripheral inflammatory changes, which might contribute to the disease pathology of EPM1.


Asunto(s)
Cistatina B/deficiencia , Encefalitis/etiología , Regulación de la Expresión Génica/genética , Inflamación/etiología , Epilepsias Mioclónicas Progresivas/complicaciones , Epilepsias Mioclónicas Progresivas/genética , Animales , Encéfalo/patología , Cistatina B/genética , Citocinas/sangre , Modelos Animales de Enfermedad , Encefalitis/sangre , Inflamación/sangre , Ratones , Ratones Noqueados , Microglía/metabolismo
4.
Glia ; 63(3): 400-11, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25327891

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal-recessively inherited neurodegenerative disorder characterized by severely incapacitating myoclonus, seizures, and ataxia, and caused by loss-of-function mutations in the cystatin B gene (CSTB). A central neuropathological finding in the Cstb(-/-) mouse, an animal model for EPM1, is early microglial activation, which precedes astroglial activation, neuronal loss, and onset of myoclonus, thus implying a critical role for microglia in EPM1 pathogenesis. Here, we characterized phenotypic and functional properties of microglia from Cstb(-/-) mice utilizing brain tissue, microglia directly isolated from the brain, and primary microglial cultures. Our results show significantly higher Cstb mRNA expression in microglia than in neurons and astrocytes. In Cstb(-/-) mouse brain, expression of the inflammatory marker p-p38 MAPK and the proportion of both pro-inflammatory M1 and anti-inflammatory M2 microglia is higher than in control mice. Moreover, M1/M2 polarization of microglia in presymptomatic Cstb(-/-) mice is, compared to control mice, skewed towards M2 type at postnatal day 14 (P14), but towards M1 type at P30, a time point associated with onset of myoclonus. At this age, the high expression of both pro-inflammatory inducible nitric oxide synthase (iNOS) and anti-inflammatory arginase 1 (ARG1) in Cstb(-/-) mouse cortex is accompanied by the presence of peripheral immune cells. Consistently, activated Cstb(-/-) microglia show elevated chemokine release and chemotaxis. However, their MHCII surface expression is suppressed. Taken together, our results link CSTB deficiency to neuroinflammation with early activation and dysfunction of microglia and will open new avenues for therapeutic interventions for EPM1.


Asunto(s)
Encéfalo/inmunología , Cistatina B/deficiencia , Microglía/fisiología , Síndrome de Unverricht-Lundborg/inmunología , Animales , Arginasa/metabolismo , Astrocitos/metabolismo , Células Cultivadas , Cistatina B/genética , Modelos Animales de Enfermedad , Genes MHC Clase II/fisiología , Granulocitos/fisiología , Macrófagos/fisiología , Ratones de la Cepa 129 , Neuroinmunomodulación/fisiología , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Mensajero/metabolismo , Linfocitos T/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
PLoS One ; 9(6): e90709, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24603771

RESUMEN

Unverricht-Lundborg type progressive myoclonus epilepsy (EPM1, OMIM 254800) is an autosomal recessive disorder characterized by onset at the age of 6 to 16 years, incapacitating stimulus-sensitive myoclonus and tonic-clonic epileptic seizures. It is caused by mutations in the gene encoding cystatin B. Previously, widespread white matter changes and atrophy has been detected both in adult EPM1 patients and in 6-month-old cystatin B-deficient mice, a mouse model for the EPM1 disease. In order to elucidate the spatiotemporal dynamics of the brain atrophy and white matter changes in EPM1, we conducted longitudinal in vivo magnetic resonance imaging and ex vivo diffusion tensor imaging accompanied with tract-based spatial statistics analysis to compare volumetric changes and fractional anisotropy in the brains of 1 to 6 months of age cystatin B-deficient and control mice. The results reveal progressive but non-uniform volume loss of the cystatin B-deficient mouse brains, indicating that different neuronal populations possess distinct sensitivity to the damage caused by cystatin B deficiency. The diffusion tensor imaging data reveal early and progressive white matter alterations in cystatin B-deficient mice affecting all major tracts. The results also indicate that the white matter damage in the cystatin B-deficient brain is most likely secondary to glial activation and neurodegenerative events rather than a primary result of CSTB deficiency. The data also show that diffusion tensor imaging combined with TBSS analysis provides a feasible approach not only to follow white matter damage in neurodegenerative mouse models but also to detect fractional anisotropy changes related to normal white matter maturation and reorganisation.


Asunto(s)
Cerebelo/patología , Cistatina B/deficiencia , Tálamo/patología , Síndrome de Unverricht-Lundborg/patología , Animales , Imagen de Difusión Tensora , Progresión de la Enfermedad , Femenino , Humanos , Imagen por Resonancia Magnética , Ratones Noqueados , Tamaño de los Órganos
6.
FEBS Lett ; 588(5): 720-6, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24462687

RESUMEN

Innate immune responses are tightly regulated to avoid excessive activation and subsequent inflammatory damage to the host, and interleukin-10 (IL-10) plays a crucial role in preventing inflammation. Stefin B (cystatin B) is an endogenous inhibitor of cysteine proteinases. In stefin B-deficient bone marrow-derived macrophages (BMDMs), we detected an increase in the induction of the LPS-induced pro-inflammatory signal nitric oxide (NO) but decreased IL-10 expression. The phosphorylation of ERK and p38 MAP-kinases was significantly decreased in stefin B-deficient macrophages, as was STAT-3 phosphorylation. These findings show that stefin B influences the expression of anti-inflammatory IL-10 in response to the TLR4 agonist LPS.


Asunto(s)
Cistatina B/deficiencia , Interleucina-10/metabolismo , Macrófagos/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Células Cultivadas , Cistatina B/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interferón gamma/genética , Interleucina-10/genética , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas , Activación de Macrófagos , Macrófagos/inmunología , Ratones Noqueados , Óxido Nítrico/metabolismo , Activación Transcripcional/inmunología
7.
Radiology ; 269(1): 232-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23788720

RESUMEN

PURPOSE: To study white matter (WM) changes in patients with Unverricht-Lundborg progressive myoclonus epilepsy (EPM1) caused by mutations in the cystatin B gene and in the cystatin B-deficient (Cstb-/-) mouse model and to validate imaging findings with histopathologic analysis of mice. MATERIALS AND METHODS: Informed consent was obtained and the study was approved by an institutional ethics committee. Animal work was approved by the Animal Experiment Board of Finland. Diffusion-tensor imaging and tract-based spatial statistics (TBSS) were used to compare fractional anisotropic (FA) results and axial, radial, and mean diffusion among patients with EPM1 (n = 19) and control subjects (n = 18). Ex vivo diffusion-tensor imaging and TBSS were used to compare Cstb-/- mice (n = 9) with wild controls (n = 4). Areas of FA decrease in mice were characterized by means of immunohistochemical analysis and transmission electron microscopy. Student t test statistics were applied to report significant findings (threshold-free cluster enhancement, P < .05). RESULTS: Patients with EPM1 showed significantly (P < .05) reduced FA and increased radial and mean diffusion in all major WM tracts compared with those of control subjects, shown as global FA decrease along the TBSS skeleton (0.41 ± 0.03 vs 0.45 ± 0.02, respectively; P < 5 × 10(-6)). Cstb-/- mice exhibited significantly reduced FA (P < .05) and antimyelin basic protein staining. Transmission electron microscopy revealed degenerating axons in Cstb-/- mice vs controls (979 axons counted, 51 degenerating axons; 2.09 ± 0.29 per field vs 1072 axons counted, nine degenerating axons; 0.48 ± 0.19 per field; P = .002). CONCLUSION: EPM1 is characterized by widespread alterations in subcortical WM, the thalamocortical system, and the cerebellum, which result in axonal degeneration and WM loss. These data suggest that motor disturbances and other symptoms in patients with EPM1 involve not only the cortical system but also the thalamocortical system and cerebellum.


Asunto(s)
Cistatina B/deficiencia , Imagen por Resonancia Magnética/métodos , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/patología , Síndrome de Unverricht-Lundborg/metabolismo , Síndrome de Unverricht-Lundborg/patología , Adolescente , Adulto , Animales , Niño , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Investigación Biomédica Traslacional , Adulto Joven
8.
J Neuropathol Exp Neurol ; 71(1): 40-53, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22157618

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is a hereditary neurodegenerative disorder caused by mutations in the cystatin B (CSTB) gene encoding an inhibitor of cysteine proteases. Here, we provide the first detailed description of the onset and progression of pathologic changes in the CNS of Cstb-deficient (Cstb) mice. Our data reveal early and localized glial activation in brain regions where neuron loss subsequently occurs. These changes are most pronounced in the thalamocortical system, with neuron loss occurring first within the cortex and only subsequently in the corresponding thalamic relay nucleus. Microglial activation precedes the emergence of myoclonia and is followed by successive astrocytosis and selective neuron loss. Neuron loss was not detected in thalamic relay nuclei that displayed no glial activation. Microglia showed morphologic changes during disease progression from that of phagocytic brain macrophages in young animals to having thickened branched processes in older animals. These novel data on the timing of pathologic events in the CSTB-deficient brain highlight the potential role of glial activation at the initial stages of the disease. Determining the precise sequence of the neurodegenerative events in Cstb mouse brains will lay the basis for understanding the pathophysiology of EPM1.


Asunto(s)
Cistatina B/deficiencia , Modelos Animales de Enfermedad , Microglía/metabolismo , Microglía/patología , Neuronas/patología , Síndrome de Unverricht-Lundborg/patología , Animales , Encéfalo , Muerte Celular/genética , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cistatina B/genética , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Factores de Tiempo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo
9.
Cancer Res ; 69(10): 4537-44, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19435903

RESUMEN

Tumors initiate angiogenesis primarily by secreting vascular endothelial growth factor (VEGF-A(164)). The first new vessels to form are greatly enlarged, pericyte-poor sinusoids, called mother vessels (MV), that originate from preexisting venules. We postulated that the venular enlargement necessary to form MV would require a selective degradation of their basement membranes, rigid structures that resist vascular expansion. To identify the specific proteases responsible for MV formation, we induced angiogenesis in mouse tissues with an adenoviral vector expressing VEGF-A(164) (Ad-VEGF-A(164)) or with VEGF-A-secreting TA3/St mammary tumors. We found that MV formation resulted from greatly increased activity of cathepsins (B>S>L) in venules transitioning into MV, as well as from a reciprocal decrease in the expression of several cysteine protease inhibitors (CPI), stefin A and cystatins B and C, by these same venules. Using a fluorescence probe that selectively binds cellular sites of cathepsin protease activity in vivo, we showed that increased cathepsin activity was localized exclusively to perivenular cells, not to venule endothelial cells. CPI strikingly inhibited angiogenesis in the Matrigel assay, and Ad-VEGF-A(164)-induced angiogenesis was reduced by approximately 50% in cathepsin B-null mice. Thus, VEGF-A, whether expressed by interstitial cells infected with an adenoviral vector or by tumor cells, upsets the normal cathepsin-CPI balance in nearby venules, leading to degradation of their basement membranes, an important first step in angiogenesis.


Asunto(s)
Catepsinas/genética , Inhibidores de Cisteína Proteinasa/farmacología , Neoplasias/irrigación sanguínea , Neovascularización Patológica/fisiopatología , Factor A de Crecimiento Endotelial Vascular/farmacología , Vénulas/fisiología , Animales , Catepsinas/antagonistas & inhibidores , Cistatina A/genética , Cistatina A/farmacología , Cistatina B/deficiencia , Cistatina B/farmacología , Ratones , Ratones Noqueados , Ratones Desnudos , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Reacción en Cadena de la Polimerasa , Vénulas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...