Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
FASEB J ; 30(5): 2040-57, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26917739

RESUMEN

An important barrier for axon regeneration and recovery after traumatic spinal cord injury (SCI) is attributed to the scar that is formed at the lesion site. Here, we investigated the effect of mouse mast cell protease (mMCP) 6, a mast cell (MC)-specific tryptase, on scarring and functional recovery after a spinal cord hemisection injury. Functional recovery was significantly impaired in both MC-deficient and mMCP6-knockout (mMCP6(-/-)) mice after SCI compared with wild-type control mice. This decrease in locomotor performance was associated with an increased lesion size and excessive scarring at the injury site. Axon growth-inhibitory chondroitin sulfate proteoglycans and the extracellular matrix components fibronectin, laminin, and collagen IV were significantly up-regulated in MC-deficient and mMCP6(-/-) mice, with an increase in scar volume between 23 and 32%. A degradation assay revealed that mMCP6 directly cleaves fibronectin and collagen IV in vitro In addition, gene expression levels of the scar components fibronectin, aggrecan, and collagen IV were increased up to 6.8-fold in mMCP6(-/-) mice in the subacute phase after injury. These data indicate that endogenous mMCP6 has scar-suppressing properties after SCI via indirect cleavage of axon growth-inhibitory scar components and alteration of the gene expression profile of these factors.-Vangansewinkel, T., Geurts, N., Quanten, K., Nelissen, S., Lemmens, S., Geboes, L., Dooley, D., Vidal, P. M., Pejler, G., Hendrix, S. Mast cells promote scar remodeling and functional recovery after spinal cord injury via mouse mast cell protease 6.


Asunto(s)
Cicatriz/metabolismo , Mastocitos/fisiología , Traumatismos de la Médula Espinal/metabolismo , Triptasas/metabolismo , Cicatrización de Heridas/fisiología , Animales , Citocinas/genética , Citocinas/metabolismo , Matriz Extracelular , Regulación Enzimológica de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Triptasas/genética
2.
Epilepsia ; 57(5): 717-26, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27020476

RESUMEN

OBJECTIVE: Febrile seizures (FS) are fever-associated convulsions, being the most common seizure disorder in early childhood. A subgroup of these children later develops epilepsy characterized by a hyperexcitable neuronal network in the hippocampus. Hippocampal excitability is regulated by the hippocampal dentate gyrus (DG) where postnatal neurogenesis occurs. Experimental FS increase the survival of newborn hippocampal dentate granule cells (DGCs), yet the significance of this neuronal subpopulation to the hippocampal network remains unclear. In the current study, we characterized the temporal maturation and structural integration of these post-FS born DGCs in the DG. METHODS: Experimental FS were induced in 10-day-old rat pups. The next day, retroviral particles coding for enhanced green fluorescent protein (eGFP) were stereotactically injected in the DG to label newborn cells. Histochemical analyses of eGFP expressing DGCs were performed one, 4, and 8 weeks later and consisted of the following: (1) colocalization with neurodevelopmental markers doublecortin, calretinin, and the mature neuronal marker NeuN; (2) quantification of dendritic complexity; and (3) quantification of spine density and morphology. RESULTS: At neither time point were neurodevelopmental markers differently expressed between FS animals and normothermia (NT) controls. One week after treatment, DGCs from FS animals showed dendrites that were 66% longer than those from NT controls. At 4 and 8 weeks, Sholl analysis of the outer 83% of the molecular layer showed 20-25% more intersections in FS animals than in NT controls (p < 0.01). Although overall spine density was not affected, an increase in mushroom-type spines was observed after 8 weeks. SIGNIFICANCE: Experimental FS increase dendritic complexity and the number of mushroom-type spines in post-FS born DGCs, demonstrating a more mature phenotype and suggesting increased incoming excitatory information. The consequences of this hyperconnectivity to signal processing in the DG and the output of the hippocampus remain to be studied.


Asunto(s)
Dendritas/fisiología , Giro Dentado/patología , Neuronas/ultraestructura , Convulsiones Febriles/patología , Factores de Edad , Animales , Animales Recién Nacidos , Calbindina 2/metabolismo , Convulsivantes/toxicidad , Giro Dentado/crecimiento & desarrollo , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Neuronas/patología , Neuropéptidos/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Polimetil Metacrilato/toxicidad , Ratas , Ratas Sprague-Dawley , Convulsiones Febriles/inducido químicamente , Transducción Genética , Transfección
3.
Neurobiol Dis ; 62: 260-72, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24075853

RESUMEN

Mast cells (MCs) are found abundantly in the central nervous system and play a complex role in neuroinflammatory diseases such as multiple sclerosis and stroke. In the present study, we show that MC-deficient Kit(W-sh/W-sh) mice display significantly increased astrogliosis and T cell infiltration as well as significantly reduced functional recovery after spinal cord injury compared to wildtype mice. In addition, MC-deficient mice show significantly increased levels of MCP-1, TNF-α, IL-10 and IL-13 protein levels in the spinal cord. Mice deficient in mouse mast cell protease 4 (mMCP4), an MC-specific chymase, also showed increased MCP-1, IL-6 and IL-13 protein levels in spinal cord samples and a decreased functional outcome after spinal cord injury. A degradation assay using supernatant from MCs derived from either mMCP4(-/-) mice or controls revealed that mMCP4 cleaves MCP-1, IL-6, and IL-13 suggesting a protective role for MC proteases in neuroinflammation. These data show for the first time that MCs may be protective after spinal cord injury and that they may reduce CNS damage by degrading inflammation-associated cytokines via the MC-specific chymase mMCP4.


Asunto(s)
Citocinas/metabolismo , Mastocitos/metabolismo , Serina Endopeptidasas/metabolismo , Traumatismos de la Médula Espinal/metabolismo , Animales , Astrocitos/patología , Femenino , Mediadores de Inflamación/metabolismo , Locomoción/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Traumatismos de la Médula Espinal/patología , Linfocitos T/metabolismo , Vértebras Torácicas/lesiones
4.
FASEB J ; 27(3): 920-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23193170

RESUMEN

Mast cells (MCs) are found abundantly in the brain and the meninges and play a complex role in neuroinflammatory diseases, such as stroke and multiple sclerosis. Here, we show that MC-deficient Kit/Kit mice display increased neurodegeneration in the lesion area after brain trauma. Furthermore, MC-deficient mice display significantly more brain inflammation, namely an increased presence of macrophages/microglia, as well as dramatically increased T-cell infiltration at days 4 and 14 after injury, combined with increased astrogliosis at day 14 following injury. The number of proliferating Ki67 macrophages/microglia and astrocytes around the lesion area is more than doubled in these MC-deficient mice. In parallel, MC-deficient Kit mice display increased presence of macrophages/microglia at day 4, and persistent astrogliosis at day 4 and 14 after brain trauma. Further analysis of mice deficient in one of the most relevant MC proteases, i.e., mouse mast cell protease 4 (mMCP-4), revealed that astrogliosis and T-cell infiltration are significantly increased in mMCP-4-knockout mice. Finally, treatment with an inhibitor of mMCP-4 significantly increased macrophage/microglia numbers and astrogliosis. These data suggest that MCs exert protective functions after trauma, at least in part via mMCP-4, by suppressing exacerbated inflammation via their proteases.


Asunto(s)
Lesiones Encefálicas/enzimología , Quimasas/metabolismo , Mastocitos/enzimología , Proteínas del Tejido Nervioso/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Quimasas/antagonistas & inhibidores , Quimasas/genética , Inflamación/enzimología , Inflamación/genética , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Mastocitos/patología , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Serina Endopeptidasas/genética , Linfocitos T/metabolismo , Linfocitos T/patología , Factores de Tiempo
5.
J Neuroinflammation ; 10: 6, 2013 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-23317037

RESUMEN

Precise crosstalk between the nervous and immune systems is important for neuroprotection and axon plasticity after injury. Recently, we demonstrated that IL-1ß acts as a potent inducer of neurite outgrowth from organotypic brain slices in vitro, suggesting a potential function of IL-1ß in axonal plasticity. Here, we have investigated the effects of IL-1ß on axon plasticity during glial scar formation and on functional recovery in a mouse model of spinal cord compression injury (SCI). We used an IL-1ß deficiency model (IL-1ßKO mice) and administered recombinant IL-1ß. In contrast to our hypothesis, the histological analysis revealed a significantly increased lesion width and a reduced number of corticospinal tract fibers caudal to the lesion center after local application of recombinant IL-1ß. Consistently, the treatment significantly worsened the neurological outcome after SCI in mice compared with PBS controls. In contrast, the absence of IL-1ß in IL-1ßKO mice significantly improved recovery from SCI compared with wildtype mice. Histological analysis revealed a smaller lesion size, reduced lesion width and greatly decreased astrogliosis in the white matter, while the number of corticospinal tract fibers increased significantly 5 mm caudal to the lesion in IL-1ßKO mice relative to controls. Our study for the first time characterizes the detrimental effects of IL-1ß not only on lesion development (in terms of size and glia activation), but also on the plasticity of central nervous system axons after injury.


Asunto(s)
Axones/fisiología , Interleucina-1beta/deficiencia , Regeneración Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Animales , Axones/efectos de los fármacos , Axones/patología , Femenino , Gliosis/inducido químicamente , Gliosis/metabolismo , Gliosis/patología , Interleucina-1beta/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración Nerviosa/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Proteínas Recombinantes/toxicidad , Traumatismos de la Médula Espinal/inducido químicamente , Resultado del Tratamiento
6.
Acta Neuropathol ; 125(5): 637-50, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23404369

RESUMEN

Mast cells (MCs) are densely granulated perivascular resident cells of hematopoietic origin and well known for their pathogenetic role in allergic and anaphylactic reactions. In addition, they are also involved in processes of innate and adaptive immunity. MCs can be activated in response to a wide range of stimuli, resulting in the release of not only pro-inflammatory, but also anti-inflammatory mediators. The patterns of secreted mediators depend upon the given stimuli and microenvironmental conditions, accordingly MCs have the ability to promote or attenuate inflammatory processes. Their presence in the central nervous system (CNS) has been recognized for more than a century. Since then a participation of MCs in various pathological processes in the CNS has been well documented. They can aggravate CNS damage in models of brain ischemia and hemorrhage, namely through increased blood-brain barrier damage, brain edema and hemorrhage formation and promotion of inflammatory responses to such events. In contrast, recent evidence suggests that MCs may have a protective role following traumatic brain injury by degrading pro-inflammatory cytokines via specific proteases. In neuroinflammatory diseases such as multiple sclerosis, the role of MCs seems to be ambiguous. MCs have been shown to be damaging, neuroprotective, or even dispensable, depending on the experimental protocols used. The role of MCs in the formation and progression of CNS tumors such as gliomas is complex and both positive and negative relationships between MC activity and tumor progression have been reported. In summary, MCs and their secreted mediators modulate inflammatory processes in multiple CNS pathologies and can thereby either contribute to neurological damage or confer neuroprotection. This review intends to give a concise overview of the regulatory roles of MCs in brain disease.


Asunto(s)
Degranulación de la Célula/fisiología , Encefalitis/etiología , Encefalitis/patología , Mastocitos/fisiología , Animales , Encefalitis/terapia , Humanos , Ratones , Ratas
7.
Eur J Immunol ; 39(3): 843-57, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19224633

RESUMEN

Chemokines mediate the inflammatory response by attracting various leukocyte types. MCP-2/CC chemokine ligand 8 (CCL8) was induced at only suboptimal levels in fibroblasts and endothelial cells by IL-1beta or IFN-gamma, unless these cytokines were combined. IFN-gamma also synergized with the TLR ligands peptidoglycan (TLR2), dsRNA (TLR3) or LPS (TLR4). Under these conditions, intact MCP-2/CCL8(1-76) produced by fibroblasts was found to be processed into MCP-2/CCL8(6-75), which lacked chemotactic activity for monocytic cells. Furthermore, the capacity of MCP-2/CCL8(6-75) to increase intracellular calcium levels through CCR1, CCR2, CCR3 and CCR5 was severely reduced. However, the truncated isoform still blocked these receptors for other ligands. MCP-2/CCL8(6-75) induced internalization of CCR2, inhibited MCP-1/CCL2 and MCP-2/CCL8 ERK signaling and antagonized the chemotactic activity of several CCR2 ligands (MCP-1/CCL2, MCP-2/CCL8, MCP-3/CCL7). In contrast to MCP-3/CCL7, parvoviral delivery of MCP-2/CCL8 into B78/H1 melanoma failed to inhibit tumor growth, partially due to proteolytic cleavage into inactive MCP-2/CCL8 missing five NH(2)-terminal residues. However, in an alternative tumor model, using HeLa cells, MCP-2/CCL8 retarded tumor development. These data indicate that optimal induction and delivery of MCP-2/CCL8 is counteracted by converting this chemokine into a receptor antagonist, thereby losing its anti-tumoral potential.


Asunto(s)
Quimiocina CCL8/metabolismo , Fibroblastos/inmunología , Neoplasias/inmunología , Receptores de Quimiocina/inmunología , Receptores Toll-Like/inmunología , Animales , Calcio/análisis , Calcio/inmunología , Calcio/metabolismo , Línea Celular , Línea Celular Tumoral , Quimiocina CCL2/inmunología , Quimiocina CCL2/metabolismo , Quimiocina CCL8/inmunología , Medios de Cultivo Condicionados/análisis , Fibroblastos/metabolismo , Humanos , Interferón gamma/farmacología , Interleucina-1beta/farmacología , Lipopolisacáridos/farmacología , Ratones , Peptidoglicano/farmacología , Receptores de Quimiocina/metabolismo , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
8.
J Neurotrauma ; 37(3): 564-571, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31210094

RESUMEN

A mounting body of evidence suggests that stress plays a major role in the injury progression after spinal cord injury (SCI). Injury activates the stress systems; this in turn may augment the generation of pro-inflammatory cytokines, stimulate pro-inflammatory immune cells, and alter the balance between the pro- and anti-inflammatory immune response. As a result, it is suggested that stress pathways may augment neuronal damage and loss after SCI. Considering these potential detrimental effects of stress after SCI, we hypothesized that inhibition of stress pathways immediately after SCI may offer protection from damage and improve recovery. To investigate the relevance of stress responses in SCI recovery, we investigated the effects of blocking three well-studied stress response axes in a mouse model of SCI. Propranolol, RU-486, and CP-99994 were administered to inhibit the sympathetic axis, the hypothalamus-pituitary-adrenal axis, and the neuropeptide axis, respectively. Surprisingly, assessing functional recovery by the Basso Mouse Scale revealed that RU-486 and CP-99994 did not affect functional outcome, indicating that these pathways are dispensable for neuroprotection or repair after SCI. Moreover, the beta-blocker propranolol worsened functional outcome in the mouse SCI model. In conclusion, immediate inhibition of three major stress axes has no beneficial effects on functional recovery after SCI in mice. These results suggest that injury-induced stress responses do not interfere with the healing process and hence, pharmacological targeting of stress responses is not a recommended treatment option for SCI. These findings are of great importance for other researchers to avoid unnecessary and potentially futile animal experiments.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Neuropéptidos/metabolismo , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Estrés Fisiológico/fisiología , Sistema Nervioso Simpático/metabolismo , Antagonistas Adrenérgicos beta/farmacología , Animales , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Femenino , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/antagonistas & inhibidores , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/fisiopatología , Estrés Fisiológico/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos , Vértebras Torácicas/lesiones
9.
J Leukoc Biol ; 99(4): 579-82, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26578647

RESUMEN

Basophils are the smallest population of granulocytes found in the circulation. They have crucial and nonredundant roles in allergic disorders, in protection from parasite infections, in autoimmunity, and in the regulation of type 2 immunity. They share phenotypic and functional properties with mast cells, which exert substantial protective effects after traumatic brain injury and spinal cord injury, although they are considered one of the most proinflammatory cell types in the body. In contrast, the in vivo functions of basophils in central nervous system trauma are still obscure and not well studied. In this study, we show that by comparing spinal cord injury in wild type vs. basophil-deficient Mcpt8Cre transgenic mice, the locomotor recovery is not affected in mice depleted in basophils. In addition, no substantial differences were observed in the lesion size and in the astrocytic and macrophage/microglia reaction between both mouse strains. Hence, despite the multiple properties shared with mast cells, these data show, for the first time, to our knowledge, that basophils are dispensable for the functional recovery process after hemisection injury to the spinal cord in mice.


Asunto(s)
Basófilos/inmunología , Locomoción , Recuperación de la Función/inmunología , Traumatismos de la Médula Espinal/inmunología , Animales , Basófilos/patología , Ratones , Ratones Transgénicos , Traumatismos de la Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología
10.
Mol Neurobiol ; 50(3): 1142-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24996996

RESUMEN

The family of interleukin (IL)-6 like cytokines plays an important role in the neuroinflammatory response to injury by regulating both neural as well as immune responses. Here, we show that expression of the IL-6 family member oncostatin M (OSM) and its receptor is upregulated after spinal cord injury (SCI). To reveal the relevance of increased OSM signaling in the pathophysiology of SCI, OSM was applied locally after spinal cord hemisection in mice. OSM treatment significantly improved locomotor recovery after mild and severe SCI. Improved recovery in OSM-treated mice was associated with a reduced lesion size. OSM significantly diminished astrogliosis and immune cell infiltration. Thus, OSM limits secondary damage after CNS trauma. In vitro viability assays demonstrated that OSM protects primary neurons in culture from cell death, suggesting that the underlying mechanism involves direct neuroprotective effects of OSM. Furthermore, OSM dose-dependently promoted neurite outgrowth in cultured neurons, indicating that the cytokine plays an additional role in CNS repair. Indeed, our in vivo experiments demonstrate that OSM treatment increases plasticity of serotonergic fibers after SCI. Together, our data show that OSM is produced at the lesion site, where it protects the CNS from further damage and promotes recovery.


Asunto(s)
Neuritas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oncostatina M/farmacología , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Relación Dosis-Respuesta a Droga , Ratones , Neuritas/fisiología , Fármacos Neuroprotectores/uso terapéutico , Oncostatina M/metabolismo , Oncostatina M/uso terapéutico , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Regulación hacia Arriba
11.
Immunobiology ; 218(2): 281-4, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22749984

RESUMEN

Spinal cord injury (SCI) is characterized by different phases of inflammatory responses. Increasing evidence indicates that the early chronic phase (two to three weeks after SCI) is characterized by a dramatic invasion of immune cells and a peak of pro-inflammatory cytokine levels, such as tumor necrosis factor-α (TNF-α) derived from the injured spinal cord as well as from injured skin, muscles and bones. However, there is substantial controversy whether these inflammatory processes in later phases lead to pro-regenerative or detrimental effects. In the present study, we investigated whether the inhibition of peripheral TNF-α in the early chronic phase after injury promotes functional recovery in a dorsal hemisection model of SCI. Three different approaches were used to continuously block peripheral TNF-α in vivo, starting 14 days after injury. We administered the TNF-α blocker etanercept intraperitoneally (every second day or daily) as well as continuously via osmotic minipumps. None of these administration routes for the TNF-α inhibitor influenced locomotor restoration as assessed by the Basso mouse scale (BMS), nor did they affect coordination and strength as evaluated by the Rotarod test. These data suggest that peripheral TNF-α inhibition may not be an effective therapeutic strategy in the early chronic phase after SCI.


Asunto(s)
Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/terapia , Factor de Necrosis Tumoral alfa/inmunología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Etanercept , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/administración & dosificación , Recuperación de la Función , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA