Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cell ; 138(6): 1222-35, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19766573

RESUMEN

Plasticity related gene-1 (PRG-1) is a brain-specific membrane protein related to lipid phosphate phosphatases, which acts in the hippocampus specifically at the excitatory synapse terminating on glutamatergic neurons. Deletion of prg-1 in mice leads to epileptic seizures and augmentation of EPSCs, but not IPSCs. In utero electroporation of PRG-1 into deficient animals revealed that PRG-1 modulates excitation at the synaptic junction. Mutation of the extracellular domain of PRG-1 crucial for its interaction with lysophosphatidic acid (LPA) abolished the ability to prevent hyperexcitability. As LPA application in vitro induced hyperexcitability in wild-type but not in LPA(2) receptor-deficient animals, and uptake of phospholipids is reduced in PRG-1-deficient neurons, we assessed PRG-1/LPA(2) receptor-deficient animals, and found that the pathophysiology observed in the PRG-1-deficient mice was fully reverted. Thus, we propose PRG-1 as an important player in the modulatory control of hippocampal excitability dependent on presynaptic LPA(2) receptor signaling.


Asunto(s)
Proteoglicanos/metabolismo , Sinapsis/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Electroencefalografía , Hipocampo/química , Hipocampo/citología , Hipocampo/metabolismo , Lisofosfolípidos/metabolismo , Ratones , Ratones Noqueados , Proteoglicanos/análisis , Proteoglicanos/genética , Receptores AMPA/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal , Proteínas de Transporte Vesicular/análisis , Proteínas de Transporte Vesicular/genética
2.
J Neuroinflammation ; 17(1): 9, 2020 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-31915017

RESUMEN

BACKGROUND: The multi-drug resistance transporter ABCG2, a member of the ATP-binding cassette (ABC) transporter family, mediates the efflux of different immunotherapeutics used in multiple sclerosis (MS), e.g., teriflunomide (teri), cladribine, and mitoxantrone, across cell membranes and organelles. Hence, the modulation of ABCG2 activity could have potential therapeutic implications in MS. In this study, we aimed at investigating the functional impact of abcg2 modulation on teri-induced effects in vitro and in vivo. METHODS: T cells from C57BL/6 J wild-type (wt) and abcg2-knockout (KO) mice were treated with teri at different concentrations with/without specific abcg2-inhibitors (Ko143; Fumitremorgin C) and analyzed for intracellular teri concentration (HPLC; LS-MS/MS), T cell apoptosis (annexin V/PI), and proliferation (CSFE). Experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6J by active immunization with MOG35-55/CFA. Teri (10 mg/kg body weight) was given orally once daily after individual disease onset. abcg2-mRNA expression (spinal cord, splenic T cells) was analyzed using qRT-PCR. RESULTS: In vitro, intracellular teri concentration in T cells was 2.5-fold higher in abcg2-KO mice than in wt mice. Teri-induced inhibition of T cell proliferation was two fold increased in abcg2-KO cells compared to wt cells. T cell apoptosis demonstrated analogous results with 3.1-fold increased apoptosis after pharmacological abcg2-inhibition in wt cells. abcg2-mRNA was differentially regulated during different phases of EAE within the central nervous system and peripheral organs. In vivo, at a dosage not efficacious in wt animals, teri treatment ameliorated clinical EAE in abcg2-KO mice which was accompanied by higher spinal cord tissue concentrations of teri. CONCLUSION: Functional relevance of abcg2 modulation on teri effects in vitro and in vivo warrants further investigation as a potential determinant of interindividual treatment response in MS, with potential implications for other immunotherapies.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/fisiología , Crotonatos/uso terapéutico , Modelos Animales de Enfermedad , Inmunoterapia/métodos , Esclerosis Múltiple/inmunología , Linfocitos T/inmunología , Toluidinas/uso terapéutico , Animales , Crotonatos/farmacología , Femenino , Humanos , Hidroxibutiratos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Esclerosis Múltiple/tratamiento farmacológico , Nitrilos , Ratas , Linfocitos T/efectos de los fármacos , Toluidinas/farmacología
3.
Acta Neuropathol ; 130(3): 373-87, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26025657

RESUMEN

Diabetic neuropathy (DNP), afflicting sensory and motor nerve fibers, is a major complication in diabetes. The underlying cellular mechanisms of axon degeneration are poorly understood. IGFBP5, an inhibitory binding protein for insulin-like growth factor 1 (IGF1) is highly up-regulated in nerve biopsies of patients with DNP. We investigated the pathogenic relevance of this finding in transgenic mice overexpressing IGFBP5 in motor axons and sensory nerve fibers. These mice develop motor axonopathy and sensory deficits similar to those seen in DNP. Motor axon degeneration was also observed in mice in which the IGF1 receptor (IGF1R) was conditionally depleted in motoneurons, indicating that reduced activity of IGF1 on IGF1R in motoneurons is responsible for the observed effect. These data provide evidence that elevated expression of IGFBP5 in diabetic nerves reduces the availability of IGF1 for IGF1R on motor axons, thus leading to progressive neurodegeneration. Inhibition of IGFBP5 could thus offer novel treatment strategies for DNP.


Asunto(s)
Axones/fisiología , Proteínas Portadoras/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Neuropatías Diabéticas/fisiopatología , Neuronas Motoras/fisiología , Degeneración Nerviosa/fisiopatología , Animales , Axones/patología , Aumento de la Célula , Supervivencia Celular/fisiología , Células Cultivadas , Diabetes Mellitus Experimental/patología , Neuropatías Diabéticas/patología , Humanos , Ratones Transgénicos , Actividad Motora/fisiología , Neuronas Motoras/patología , Degeneración Nerviosa/patología , Nervio Frénico/patología , Nervio Frénico/fisiopatología , Receptor IGF Tipo 1/metabolismo , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Sensación/fisiología
4.
Development ; 138(24): 5321-31, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22071102

RESUMEN

The generation of astrocytes during the development of the mammalian spinal cord is poorly understood. Here, we demonstrate for the first time that the extracellular matrix glycoprotein tenascin C regulates the expression of key patterning genes during late embryonic spinal cord development, leading to a timely maturation of gliogenic neural precursor cells. We first show that tenascin C is expressed by gliogenic neural precursor cells during late embryonic development. The loss of tenascin C leads to a sustained generation and delayed migration of Fgfr3-expressing immature astrocytes in vivo. Consistent with an increased generation of astroglial cells, we documented an increased number of GFAP-positive astrocytes at later stages. Mechanistically, we could demonstrate an upregulation and domain shift of the patterning genes Nkx6.1 and Nkx2.2 in vivo. In addition, sulfatase 1, a known downstream target of Nkx2.2 in the ventral spinal cord, was also upregulated. Sulfatase 1 regulates growth factor signalling by cleaving sulphate residues from heparan sulphate proteoglycans. Consistent with this function, we observed changes in both FGF2 and EGF responsiveness of spinal cord neural precursor cells. Taken together, our data implicate Tnc in the regulation of proliferation and lineage progression of astroglial progenitors in specific domains of the developing spinal cord.


Asunto(s)
Astrocitos/citología , Tipificación del Cuerpo/genética , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/genética , Médula Espinal/crecimiento & desarrollo , Tenascina/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Sulfotransferasas/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Proteínas de Pez Cebra
5.
Mol Cell Neurosci ; 56: 18-28, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23500004

RESUMEN

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family and a ligand for the tropomyosin-receptor kinase B (TrkB), mediates neuronal survival, differentiation, and synaptic plasticity. However, BDNF is not used to treat neurodegenerative diseases because of its poor pharmacokinetic profile, side effects, and absence of survival properties in clinical trials. Consequently, alternative approaches such as TrkB receptor agonist application are gaining importance. 7,8-Dihydroxyflavone (7,8-DHF), a member of the flavonoid family, has been described as a robust TrkB receptor agonist in hippocampal neurons. Nevertheless, the influence of 7,8-DHF on motoneurons, one of the main targets of BDNF in vivo, is so far unknown. Therefore, we investigated the impact of 7,8-DHF treatment on primary cultured mouse motoneurons. Indeed, we found an activation of the TrkB receptor. Moreover, 7,8-DHF application promotes survival and neurite growth of cultured motoneurons and these effects appear dose-dependent. To investigate the PI3K/AKT and MAPK pathway activation in 7,8-DHF treated motoneurons, we developed a high-density culture system of primary mouse motoneurons. Analysis of both pathways demonstrated a PI3K/AKT but not MAPK pathway activation in cultured motoneurons. This is in contrast to previously published reports about BDNF-mediated activation of TrkB. The lack of MAPK pathway activation is also in contrast to what has been found for hippocampal neurons that indeed show MAPK activation after 7,8-DHF treatment. The ability of 7,8-DHF to imitate BDNF function in motoneurons by using Trk receptor signaling would provide a new approach for the treatment of motoneuron diseases, but needs a more detailed analysis of the activation profile of 7,8-DHF.


Asunto(s)
Flavonas/farmacología , Sistema de Señalización de MAP Quinasas , Neuronas Motoras/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Procesos de Crecimiento Celular , Supervivencia Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hipocampo/citología , Hipocampo/embriología , Ratones , Neuronas Motoras/metabolismo , Neuronas Motoras/fisiología , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuritas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor trkB/agonistas , Receptor trkB/metabolismo , Médula Espinal/citología , Médula Espinal/embriología
6.
J Am Chem Soc ; 135(16): 6033-46, 2013 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-23384205

RESUMEN

A series of Ni-based electrocatalysts, [Ni(7P(Ph)2N(C6H4X))2](BF4)2, featuring seven-membered cyclic diphosphine ligands incorporating a single amine base, 1-para-X-phenyl-3,6-triphenyl-1-aza-3,6-diphosphacycloheptane (7P(Ph)2N(C6H4X), where X = OMe, Me, Br, Cl, or CF3), have been synthesized and characterized. X-ray diffraction studies have established that the [Ni(7P(Ph)2N(C6H4X))2](2+) complexes have a square planar geometry, with bonds to four phosphorus atoms of the two bidentate diphosphine ligands. Each of the complexes is an efficient electrocatalyst for hydrogen production at the potential of the Ni(II/I) couple, with turnover frequencies ranging from 2400 to 27,000 s(-1) with [(DMF)H](+) in acetonitrile. Addition of water (up to 1.0 M) accelerates the catalysis, giving turnover frequencies ranging from 4100 to 96,000 s(-1). Computational studies carried out on the [Ni(7P(Ph)2N(C6H4X))2](2+) family indicate the catalytic rates reach a maximum when the electron-donating character of X results in the pKa of the Ni(I) protonated pendant amine matching that of the acid used for proton delivery. Additionally, the fast catalytic rates for hydrogen production by the [Ni(7P(Ph)2N(C6H4X))2](2+) family relative to the analogous [Ni(P(Ph)2N(C6H4X)2)2](2+) family are attributed to preferred formation of endo protonated isomers with respect to the metal center in the former, which is essential to attain suitable proximity to the reduced metal center to generate H2. The results of this work highlight the importance of precise pKa matching with the acid for proton delivery to obtain optimal rates of catalysis.

7.
Am J Pathol ; 180(1): 267-74, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22152994

RESUMEN

Laquinimod is a promising, orally available compound that has been successfully evaluated in placebo-controlled phase II/III studies of relapsing-remitting multiple sclerosis (MS). Studies are ongoing to further define laquinimod's modulatory mechanisms. Analyses in the animal model of experimental autoimmune encephalomyelitis (EAE) demonstrate that laquinimod reduces infiltration of leukocytes into the central nervous system, induces a Th1 to Th2/3 shift, and suppresses Th17 responses. To evaluate the potential neuroprotective capacity of laquinimod via modulation of brain-derived neurotrophic factor (BDNF), we analyzed the expression of BDNF in blood samples from 203 MS patients treated with laquinimod. Furthermore, we investigated the effect of laquinimod in EAE using a conditional BDNF knockout strain lacking BDNF expression in myeloid cells and T cells (LLF mice). Treatment with laquinimod resulted in a significant and persistent increase in BDNF serum levels of MS patients when compared to baseline and placebo-treated patients. LLF mice treated with laquinimod display a more severe EAE disease course in comparison to wild-type mice. Furthermore, laquinimod-treated wild-type monocytes secreted an anti-inflammatory cytokine pattern in comparison to untreated wild-type monocytes and treated LLF monocytes. Adoptive transfer of laquinimod stimulated monocytes into mice with EAE ameliorated the disease course. Consistent with immunomodulatory properties, laquinimod skewed monocytes toward a regulatory phenotype and also acted via modulation of BDNF, which may contribute to neuroprotection in MS patients.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/prevención & control , Esclerosis Múltiple/prevención & control , Fármacos Neuroprotectores/farmacología , Quinolonas/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Esclerosis Múltiple/metabolismo
8.
Neurochem Res ; 38(6): 1285-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23624942

RESUMEN

Complex glycan structures and their respective carrier molecules are often expressed in a cell type specific manner. Thus, glycans can be used for the enrichment of specific cell types such as neural precursor cells (NPCs). We have recently shown that the monoclonal antibodies 487(LeX) and 5750(LeX) differentially detect the LewisX (LeX) glycan on NPCs in the developing mouse forebrain. Here, we analysed the staining pattern of both antibodies during late embryonic mouse spinal cord development. At E13.5 both antibodies strongly label the central canal region. Along these lines they detect the LeX glycan primarily on Nestin-positive NPCs at that age. Moreover, we show that spinal cord NPCs cultured as free floating neurospheres display a high immunoreactivity to both antibodies. In that context, we also demonstrate that the 487(LeX) antibody can be used to deplete a subpopulation of neurosphere forming NPCs from a mixed E13.5 spinal cord cell suspension. Towards the end of embryogenesis the overall immunoreactivity to both antibodies increases and the staining appears very diffuse. However, the 5750(LeX) antibody still labels the central canal region. The increase in immunoreactivity correlates with an expression increase of the extracellular matrix molecules Tenascin C and Receptor Protein Tyrosine Phosphatase ß/ζ, two potential LeX carrier proteins. In line with this, immunoprecipitation analyses confirmed Tenascin C as a LeX carrier protein in the embryonic mouse spinal cord. However, the immunoreactivity to both antibodies appears only to be marginally affected in the absence of Tenascin C, arguing against Tenascin C being the major LeX carrier. In conclusion our study gives some novel insights into the complex expression of LeX glycans and potential carrier proteins during the development of the mouse spinal cord.


Asunto(s)
Antígeno Lewis X/biosíntesis , Células-Madre Neurales/metabolismo , Polisacáridos/biosíntesis , Médula Espinal/embriología , Animales , Células Cultivadas , Ratones , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/biosíntesis , Médula Espinal/metabolismo , Tenascina/biosíntesis
9.
J Am Chem Soc ; 134(24): 10114-21, 2012 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-22616768

RESUMEN

We report bifunctional reactivity of the ß-diketiminato Ni(III)-imide [Me(3)NN]Ni═NAd (1), which undergoes H-atom abstraction (HAA) reactions with benzylic substrates R-H (indane, ethylbenzene, toluene). Nickel-imide 1 competes with the nickel-amide HAA product [Me(3)NN]Ni-NHAd (2) for the resulting hydrocarbyl radical R(•) to give the nickel-amide [Me(3)NN]Ni-N(CHMePh)Ad (3) (R-H = ethylbenzene) or aminoalkyl tautomer [Me(3)NN]Ni(η(2)-CH(Ph)NHAd) (4) (R-H = toluene). A significant amount of functionalized amine R-NHAd is observed in the reaction of 1 with indane along with the dinickel imide {[Me(3)NN]Ni}(2)(µ-NAd) (5). Kinetic and DFT analyses point to rate-limiting HAA from R-H by 1 to give R(•), which may add to either imide 1 or amide 2, each featuring significant N-based radical character. Thus, these studies illustrate a fundamental competition possible in C-H amination systems that proceed via a HAA/radical rebound mechanism.

10.
Brain ; 134(Pt 3): 678-92, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21354971

RESUMEN

Inflammation and oxidative stress are thought to promote tissue damage in multiple sclerosis. Thus, novel therapeutics enhancing cellular resistance to free radicals could prove useful for multiple sclerosis treatment. BG00012 is an oral formulation of dimethylfumarate. In a phase II multiple sclerosis trial, BG00012 demonstrated beneficial effects on relapse rate and magnetic resonance imaging markers indicative of inflammation as well as axonal destruction. First we have studied effects of dimethylfumarate on the disease course, central nervous system, tissue integrity and the molecular mechanism of action in an animal model of chronic multiple sclerosis: myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis in C57BL/6 mice. In the chronic phase of experimental autoimmune encephalomyelitis, preventive or therapeutic application of dimethylfumarate ameliorated the disease course and improved preservation of myelin, axons and neurons. In vitro, the application of fumarates increased murine neuronal survival and protected human or rodent astrocytes against oxidative stress. Application of dimethylfumarate led to stabilization of the transcription factor nuclear factor (erythroid-derived 2)-related factor 2, activation of nuclear factor (erythroid-derived 2)-related factor 2-dependent transcriptional activity and accumulation of NADP(H) quinoline oxidoreductase-1 as a prototypical target gene. Furthermore, the immediate metabolite of dimethylfumarate, monomethylfumarate, leads to direct modification of the inhibitor of nuclear factor (erythroid-derived 2)-related factor 2, Kelch-like ECH-associated protein 1, at cysteine residue 151. In turn, increased levels of nuclear factor (erythroid-derived 2)-related factor 2 and reduced protein nitrosylation were detected in the central nervous sytem of dimethylfumarate-treated mice. Nuclear factor (erythroid-derived 2)-related factor 2 was also upregulated in the spinal cord of autopsy specimens from untreated patients with multiple sclerosis. In dimethylfumarate-treated mice suffering from experimental autoimmune encephalomyelitis, increased immunoreactivity for nuclear factor (erythroid-derived 2)-related factor 2 was detected by confocal microscopy in neurons of the motor cortex and the brainstem as well as in oligodendrocytes and astrocytes. In mice deficient for nuclear factor (erythroid-derived 2)-related factor 2 on the same genetic background, the dimethylfumarate mediated beneficial effects on clinical course, axon preservation and astrocyte activation were almost completely abolished thus proving the functional relevance of this transcription factor for the neuroprotective mechanism of action. We conclude that the ability of dimethylfumarate to activate nuclear factor (erythroid-derived 2)-related factor 2 may offer a novel cytoprotective modality that further augments the natural antioxidant responses in multiple sclerosis tissue and is not yet targeted by other multiple sclerosis therapies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/prevención & control , Fumaratos/uso terapéutico , Factor 2 Relacionado con NF-E2/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Aldehído Reductasa/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Antioxidantes/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Axones/metabolismo , Axones/patología , Complejo CD3/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromatografía Líquida de Alta Presión/métodos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Encefalomielitis Autoinmune Experimental/etiología , Femenino , Fumaratos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/efectos adversos , Proteínas Fluorescentes Verdes/genética , Humanos , Peróxido de Hidrógeno/farmacología , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/citología , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Proteínas de la Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Fármacos Neuroprotectores/farmacología , Proteínas Nogo , Estrés Oxidativo/efectos de los fármacos , Fragmentos de Péptidos/efectos adversos , ARN Interferente Pequeño/farmacología , Sueño/fisiología , Médula Espinal/citología , Estadísticas no Paramétricas , Espectrometría de Masas en Tándem/métodos , Factores de Tiempo , Transfección
11.
J Neurosci ; 30(5): 1739-49, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-20130183

RESUMEN

Although brain-derived neurotrophic factor (BDNF) is linked with an increasing number of conditions causing brain dysfunction, its role in the postnatal CNS has remained difficult to assess. This is because the bdnf-null mutation causes the death of the animals before BDNF levels have reached adult levels. In addition, the anterograde axonal transport of BDNF complicates the interpretation of area-specific gene deletion. The present study describes the generation of a new conditional mouse mutant essentially lacking BDNF throughout the CNS. It shows that BDNF is not essential for prolonged postnatal survival, but that the behavior of such mutant animals is markedly altered. It also reveals that BDNF is not a major survival factor for most CNS neurons and for myelination of their axons. However, it is required for the postnatal growth of the striatum, and single-cell analyses revealed a marked decreased in dendritic complexity and spine density. In contrast, BDNF is dispensable for the growth of the hippocampus and only minimal changes were observed in the dendrites of CA1 pyramidal neurons in mutant animals. Spine density remained unchanged, whereas the proportion of the mushroom-type spine was moderately decreased. In line with these in vivo observations, we found that BDNF markedly promotes the growth of cultured striatal neurons and of their dendrites, but not of those of hippocampal neurons, suggesting that the differential responsiveness to BDNF is part of a neuron-intrinsic program.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/crecimiento & desarrollo , Neostriado/crecimiento & desarrollo , Animales , Recuento de Células , Células Cultivadas , Dendritas/metabolismo , Dendritas/ultraestructura , Femenino , Hipocampo/citología , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Neostriado/ultraestructura , Neuronas/citología , Neuronas/ultraestructura , Oligodendroglía/citología , Oligodendroglía/ultraestructura , Nervio Óptico/crecimiento & desarrollo , Nervio Óptico/ultraestructura , Proteínas tau/metabolismo
12.
J Neurosci Res ; 89(2): 127-41, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21162121

RESUMEN

Mechanisms controlling neuronal survival and regeneration play an important role during development, after birth, and under lesion conditions. Isolated embryonic mouse motoneurons have been a useful tool for studying such basic mechanisms. These cultured motoneurons depend on extracellular matrix (ECM) molecules, which are potent mediators of survival and axonal growth and guidance in the CNS and in vitro, exhibiting either attractive or repellent guidance cues. Additionally, ECM proteoglycans and glycoproteins are components of the glial scar acting as a growth barrier for regenerating axons. Compared with CNS axon outgrowth, less is known about the cues that guide motoneurons toward their peripheral targets. Because we are interested in the effects of glial-derived chondroitin sulfate proteoglycans (CSPGs), we have worked out a model system for investigating the influences of glial-derived matrix molecules on motoneuron outgrowth and survival. We used cultured embryonic mouse motoneurons to investigate axon growth effects of matrix molecules produced by the glial-derived cell lines A7, Neu7, and Oli-neu primary astrocytes as well as the immortalized Schwann cell line IMS32. The results indicate that molecules of the ECM, especially chondroitin sulfates, play an important role as axon growth-promoting cues. We could demonstrate a modifying effect of the matrix components on motoneuron survival and caspase3-induced apoptosis.


Asunto(s)
Sulfatos de Condroitina/metabolismo , Matriz Extracelular/metabolismo , Neuronas Motoras/citología , Neuritas , Neuroglía/metabolismo , Animales , Diferenciación Celular/fisiología , Línea Celular , Supervivencia Celular , Embrión de Mamíferos , Técnica del Anticuerpo Fluorescente , Ratones , Neuronas Motoras/metabolismo , Neuritas/metabolismo
13.
Brain ; 133(Pt 8): 2248-63, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20826430

RESUMEN

Brain-derived neurotrophic factor plays a key role in neuronal and axonal survival. Brain-derived neurotrophic factor is expressed in the immune cells in lesions of experimental autoimmune encephalomyelitis and multiple sclerosis, thus potentially mediating neuroprotective effects. We investigated the functional role of brain-derived neurotrophic factor in myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Mice deficient for brain-derived neurotrophic factor in immune cells displayed an attenuated immune response in the acute phase of experimental autoimmune encephalomyelitis, but progressive disability with enhanced axonal loss in the chronic phase of the disease. In mice deficient for central nervous system-derived brain-derived neurotrophic factor via glial fibrillary acidic protein-crescentin-mediated deletion, a more severe course of experimental autoimmune encephalomyelitis and an overall increased axonal loss was observed. In a lentiviral approach, injection of brain-derived neurotrophic factor-overexpressing T cells led to a less severe course of experimental autoimmune encephalomyelitis and direct axonal protection. Our data imply a functional role of brain-derived neurotrophic factor in autoimmune demyelination by mediating axon protection.


Asunto(s)
Autoinmunidad/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Enfermedad Aguda , Animales , Axones/inmunología , Axones/patología , Encéfalo/inmunología , Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Factor Neurotrófico Derivado del Encéfalo/genética , Enfermedad Crónica , Evaluación de la Discapacidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Esclerosis Múltiple , Neuroinmunomodulación/fisiología , Receptor trkB/metabolismo , Médula Espinal/inmunología , Médula Espinal/patología , Linfocitos T/metabolismo
14.
J Neurosci ; 29(43): 13640-8, 2009 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-19864576

RESUMEN

Growth/differentiation factor-15 (GDF-15) is a widely expressed distant member of the TGF-beta superfamily with prominent neurotrophic effects on midbrain dopaminergic neurons. We show here that GDF-15-deficient mice exhibit progressive postnatal losses of spinal, facial, and trigeminal motoneurons. This deficit reaches a approximately 20% maximum at 6 months and is accompanied by losses of motor axons and significant impairment of rotarod skills. Similarly, sensory neurons in dorsal root ganglia (L4, L5) are reduced by 20%, whereas sympathetic neurons are not affected. GDF-15 is expressed and secreted by Schwann cells, retrogradely transported along adult sciatic nerve axons, and promotes survival of axotomized facial neurons as well as cultured motor, sensory, and sympathetic neurons. Despite striking similarities in the GDF-15 and CNTF knock-out phenotypes, expression levels of CNTF and other neurotrophic factors in the sciatic nerve were unaltered suggesting that GDF-15 is a genuine novel trophic factor for motor and sensory neurons.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/fisiología , Neuronas Motoras/fisiología , Neuronas/fisiología , Animales , Muerte Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Factor Neurotrófico Ciliar/metabolismo , Nervio Facial/crecimiento & desarrollo , Nervio Facial/fisiopatología , Ganglios Espinales/fisiopatología , Factor 15 de Diferenciación de Crecimiento/deficiencia , Factor 15 de Diferenciación de Crecimiento/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Destreza Motora/fisiología , Músculo Esquelético/fisiopatología , Células de Schwann/fisiología , Nervio Ciático/fisiopatología , Células Receptoras Sensoriales/fisiología , Médula Espinal/crecimiento & desarrollo , Médula Espinal/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Nervio Trigémino/crecimiento & desarrollo , Nervio Trigémino/fisiopatología
15.
J Am Chem Soc ; 131(50): 18105-11, 2009 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20000860

RESUMEN

Monovalent nickel and copper beta-diketiminato complexes react with ArN=O (Ar = 3,5-Me(2)C(6)H(3), Ph) to give C-nitroso adducts that exhibit three different modes of bonding with varying degrees of N-O bond activation. The addition of ArNO to 2 equiv of [Me(2)NN]Ni(2,4-lutidine) {[Me(2)NN](-) = 2,4-bis(2,6-dimethylphenylimido)pentyl} gives {[Me(2)NN]Ni}(2)(mu-eta(2):eta(2)-ONAr) (1a and 1b), which exhibit symmetrical bonding of the ArN=O moiety between two [Me(2)NN]Ni fragments, with a N-O bond distance of 1.440(4) A in 1a that is significantly longer than those in free C-organonitroso compounds (1.13-1.29 A). [Me(2)NN]Cu(NCMe) reacts with 0.5 equiv of ArNO in ether to give the dinuclear adducts {[Me(2)NN]Cu}(2)(mu-eta(2):eta(1)-ONAr) (2a and 2b), which exhibit eta(2) and eta(1) bonding of the ArN=O moiety with separate [Me(2)NN]Cu fragments possessing N-O distances of 1.375(6) A (2a) and 1.368(2) A (2b). In arene solvents, one beta-diketiminatocopper(I) fragment dissociates from 2 to give [Me(2)NN]Cu(eta(2)-ONAr) (3a and 3b), which may be isolated by the addition of 1 equiv of ArNO to [Me(2)NN]Cu(NCMe). The X-ray structures of 3a and 3b are similar to those of related Cu(I) alkene adducts, with N-O distances in the narrow range 1.333(4)-1.338(5) A. IR spectra of the nitrosobenzene adducts 1b, 2b, and 3b exhibit nu(NO) stretching frequencies at 915, 1040, and 1113 cm(-1), respectively, following the decreasing degree of N=O activation observed in the X-ray structures of species 1, 2, and 3. Both 1a and 3a react with anaerobic NO(g) to give the corresponding N-aryl-N-nitrosohydroxylaminato complexes [Me(2)NN]M(kappa(2)-O(2)N(2)Ar) [M = Ni (4), Cu (5)]. In the reaction of dinuclear 1a with NO, one [Me(2)NN]Ni fragment is trapped as the nickel nitrosyl [Me(2)NN]Ni(NO). Reaction of the monovalent complex [Me(2)NN]Cu(eta(2)-ONAr) with NO(g) to give divalent [Me(2)NN]Cu(kappa(2)-O(2)N(2)Ar) represents an example of oxidative nitrosylation.


Asunto(s)
Cobre/química , Níquel/química , Óxido Nítrico/química , Compuestos Nitrosos/síntesis química , Compuestos Organometálicos/síntesis química , Modelos Moleculares , Estructura Molecular , Compuestos Nitrosos/química , Compuestos Organometálicos/química , Oxidación-Reducción
16.
J Cell Biol ; 159(4): 563-9, 2002 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-12446740

RESUMEN

Progressive motor neuronopathy (pmn) mutant mice have been widely used as a model for human motoneuron disease. Mice that are homozygous for the pmn gene defect appear healthy at birth but develop progressive motoneuron disease, resulting in severe skeletal muscle weakness and respiratory failure by postnatal week 3. The disease starts at the motor endplates, and then leads to axonal loss and finally to apoptosis of the corresponding cell bodies. We localized the genetic defect in pmn mice to a missense mutation in the tubulin-specific chaperone E (Tbce) gene on mouse chromosome 13. The human orthologue maps to chromosome 1q42.3. The Tbce gene encodes a protein (cofactor E) that is essential for the formation of primary alpha-tubulin and beta-tubulin heterodimeric complexes. Isolated motoneurons from pmn mutant mice exhibit shorter axons and axonal swelling with irregularly structured beta-tubulin and tau immunoreactivity. Thus, the pmn gene mutation provides the first genetic evidence that alterations in tubulin assembly lead to retrograde degeneration of motor axons, ultimately resulting in motoneuron cell death.


Asunto(s)
Chaperonas Moleculares/genética , Enfermedad de la Neurona Motora/genética , Mutación Missense , Secuencia de Aminoácidos , Animales , Tamaño de la Célula , Supervivencia Celular , Cromosomas Humanos Par 1 , Modelos Animales de Enfermedad , Embrión de Mamíferos/anatomía & histología , Femenino , Genotipo , Humanos , Masculino , Ratones , Ratones Endogámicos , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Enfermedad de la Neurona Motora/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Linaje , Alineación de Secuencia
17.
J Cell Biol ; 156(2): 287-97, 2002 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-11807093

RESUMEN

Members of the ciliary neurotrophic factor (CNTF)/leukemia inhibitory factor (LIF)/cardiotrophin gene family are potent survival factors for embryonic and lesioned motoneurons. These factors act via receptor complexes involving gp130 and LIFR-beta and ligand binding leads to activation of various signaling pathways, including phosphorylation of Stat3. The role of Stat3 in neuronal survival was investigated in mice by Cre-mediated gene ablation in motoneurons. Cre is expressed under the neurofilament light chain (NF-L) promoter, starting around E12 when these neurons become dependent on neurotrophic support. Loss of motoneurons during the embryonic period of naturally occurring cell death is not enhanced in NF-L-Cre; Stat3(flox/KO) mice although motoneurons isolated from these mice need higher concentrations of CNTF for maximal survival in culture. In contrast, motoneuron survival is significantly reduced after facial nerve lesion in the adult. These neurons, however, can be rescued by the addition of neurotrophic factors, including CNTF. Stat3 is essential for upregulation of Reg-2 and Bcl-xl expression in lesioned motoneurons. Our data show that Stat3 activation plays an essential role for motoneuron survival after nerve lesion in postnatal life but not during embryonic development, indicating that signaling requirements for motoneuron survival change during maturation.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Traumatismos del Nervio Facial/patología , Neuronas Motoras/citología , Proteínas del Tejido Nervioso , Sistema Nervioso/embriología , Sistema Nervioso/patología , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo , Envejecimiento , Animales , Animales Recién Nacidos , Axotomía , Proteínas de Unión al Calcio/metabolismo , Muerte Celular/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Factor Neurotrófico Ciliar/farmacología , Traumatismos del Nervio Facial/genética , Traumatismos del Nervio Facial/metabolismo , Eliminación de Gen , Integrasas/genética , Integrasas/metabolismo , Litostatina , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Sistema Nervioso/crecimiento & desarrollo , Sistema Nervioso/metabolismo , Proteínas de Neurofilamentos/genética , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factor de Transcripción STAT3 , Proteínas Virales/genética , Proteínas Virales/metabolismo , Proteína bcl-X
18.
Mol Cell Biol ; 26(19): 7103-15, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16980614

RESUMEN

We have previously shown that mice lacking the protein kinase B-RAF have defects in both neural and endothelial cell lineages and die around embryonic day 12 (E12). To delineate the function of B-RAF in the brain, B-RAF KIN/KIN mice lacking B-RAF and expressing A-RAF under the control of the B-RAF locus were created. B-RAF KIN/KIN embryos displayed no vascular defects, no endothelial and neuronal apoptosis, or gross developmental abnormalities, and a significant proportion of these animals survived for up to 8 weeks. Cell proliferation in the neocortex was reduced from E14.5 onwards. Newborn cortical neurons were impaired in their migration toward the cortical plate, causing a depletion of Brn-2-expressing pyramidal neurons in layers II, III, and V of the postnatal cortex. Our data reveal that B-RAF is an important mediator of neuronal survival, migration, and dendrite formation and that A-RAF cannot fully compensate for these functions.


Asunto(s)
Movimiento Celular , Neocórtex/patología , Proteínas Proto-Oncogénicas A-raf/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Animales , Muerte Celular , Proliferación Celular , Supervivencia Celular/fisiología , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/patología , Dendritas/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Endoteliales/citología , Genotipo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neocórtex/citología , Proteínas Proto-Oncogénicas B-raf/deficiencia , Células Madre/citología
19.
Nat Neurosci ; 8(9): 1169-78, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116448

RESUMEN

Bag1 is a cochaperone for the heat-shock protein Hsp70 that interacts with C-Raf, B-Raf, Akt, Bcl-2, steroid hormone receptors and other proteins. Here we use targeted gene disruption in mice to show that Bag1 has an essential role in the survival of differentiating neurons and hematopoietic cells. Cells of the fetal liver and developing nervous system in Bag1-/- mice underwent massive apoptosis. Lack of Bag1 did not disturb the primary function of Akt or Raf, as phosphorylation of the forkhead transcription factor FKHR and activation of extracellular signal-regulated kinase (Erk)-1/2 were not affected. However, the defect was associated with the disturbance of a tripartite complex formed by Akt, B-Raf and Bag1, in addition to the absence of Bad phosphorylation at Ser136. We also observed reduced expression of members of the inhibitor of apoptosis (IAP) family. Our data show that Bag1 is a physiological mediator of extracellular survival signals linked to the cellular mechanisms that prevent apoptosis in hematopoietic and neuronal progenitor cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Proteínas de la Membrana/fisiología , Neuronas Motoras/fisiología , Médula Espinal/citología , Fosfatasa Alcalina/metabolismo , Análisis de Varianza , Animales , Southern Blotting/métodos , Western Blotting/métodos , Proteínas Portadoras/metabolismo , Recuento de Células/métodos , Supervivencia Celular/fisiología , Células Cultivadas , Proteínas de Unión al ADN , Complejo IV de Transporte de Electrones/metabolismo , Embrión de Mamíferos , Proteínas del Ojo/metabolismo , Fibroblastos/metabolismo , Proteínas HSP70 de Choque Térmico/deficiencia , Proteínas de Homeodominio/metabolismo , Inmunohistoquímica/métodos , Inmunoprecipitación/métodos , Etiquetado Corte-Fin in Situ/métodos , Proteínas de Filamentos Intermediarios/metabolismo , Isoenzimas/metabolismo , Hígado/citología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Proteínas de la Membrana/deficiencia , Ratones , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/metabolismo , Nestina , Proteínas de Neurofilamentos/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , Propidio , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-akt , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/metabolismo , Ratas , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Factores de Transcripción , Transfección/métodos , Proteína Letal Asociada a bcl
20.
PLoS One ; 12(3): e0171552, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28273090

RESUMEN

Fingolimod is an oral sphingosine-1-phosphate-receptor modulator which reduces the recirculation of immune cells and may also directly target glial cells. Here we investigate effects of fingolimod on expression of astroglial glutamate transporters under pro-inflammatory conditions. In astrocyte cell culture, the addition of pro-inflammatory cytokines led to a significant downregulation of glutamate transporters glutamate transporter-1 (slc1a2/SLC1A2) and glutamate aspartate transporter (slc1a3/SLC1A3) expression on the mRNA or protein level. In this setting, the direct application of fingolimod-1 phosphate (F1P) on astrocytes did not change expression levels of slc1a2 and slc1a3 mRNA. The analysis of both transporters on the protein level by Western Blot and immunocytochemistry did also not reveal any effect of F1P. On a functional level, the addition of conditioned supernatants from F1P treated astrocytes to neuronal cell culture did not result in increased neurite growth. In experimental autoimmune encephalomyelitis as a model of multiple sclerosis, fingolimod treatment reduced T cell and macrophages/microglia mediated inflammation and also diminished astrocyte activation. At the same time, fingolimod restored the reduced expression of slc1a2 and slc1a3 in the inflamed spinal cord on the mRNA level and of SLC1A2 and SLC1A3 on the protein level, presumably via indirect, anti-inflammatory mechanisms. These findings provide further evidence for a predominantly peripheral effect of the compound in neuroinflammation.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Transportador 1 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Clorhidrato de Fingolimod/farmacología , Inmunosupresores/farmacología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Células Cultivadas , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/genética , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Interleucina-1beta/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Células PC12 , ARN Mensajero/metabolismo , Ratas , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA