Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 40(15): 3104-3118, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32127495

RESUMEN

Krabbe's disease is an infantile neurodegenerative disease, which is affected by mutations in the lysosomal enzyme galactocerebrosidase, leading to the accumulation of its metabolite psychosine. We have shown previously that the S1P receptor agonist fingolimod (FTY720) attenuates psychosine-induced glial cell death and demyelination both in vitro and ex vivo models. These data, together with a lack of therapies for Krabbe's disease, prompted the current preclinical study examining the effects of fingolimod in twitcher mice, a murine model of Krabbe's disease. Twitcher mice, both male and female, carrying a natural mutation in the galc gene were given fingolimod via drinking water (1 mg/kg/d). The direct impact of fingolimod administration was assessed via histochemical and biochemical analysis using markers of myelin, astrocytes, microglia, neurons, globoid cells, and immune cells. The effects of fingolimod on twitching behavior and life span were also demonstrated. Our results show that treatment of twitcher mice with fingolimod significantly rescued myelin levels compared with vehicle-treated animals and also regulated astrocyte and microglial reactivity. Furthermore, nonphosphorylated neurofilament levels were decreased, indicating neuroprotective and neurorestorative processes. These protective effects of fingolimod on twitcher mice brain pathology was reflected by an increased life span of fingolimod-treated twitcher mice. These in vivo findings corroborate initial in vitro studies and highlight the potential use of S1P receptors as drug targets for treatment of Krabbe's disease.SIGNIFICANCE STATEMENT This study demonstrates that the administration of the therapy known as fingolimod in a mouse model of Krabbe's disease (namely, the twitcher mouse model) significantly rescues myelin levels. Further, the drug fingolimod also regulates the reactivity of glial cells, astrocytes and microglia, in this mouse model. These protective effects of fingolimod result in an increased life span of twitcher mice.


Asunto(s)
Enfermedades Desmielinizantes/tratamiento farmacológico , Clorhidrato de Fingolimod/uso terapéutico , Leucodistrofia de Células Globoides/tratamiento farmacológico , Animales , Astrocitos/metabolismo , Conducta Animal , Femenino , Leucodistrofia de Células Globoides/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes Neurológicos , Microglía/metabolismo , Vaina de Mielina/metabolismo , Proteínas de Neurofilamentos/metabolismo , Neuronas/metabolismo , Fosforilación , Proproteína Convertasas/efectos de los fármacos , Células de Purkinje/metabolismo , Serina Endopeptidasas/efectos de los fármacos
2.
Int J Mol Sci ; 22(9)2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919387

RESUMEN

The EBI2 receptor regulates the immune system and is expressed in various immune cells including B and T lymphocytes. It is also expressed in astrocytes in the central nervous system (CNS) where it regulates pro-inflammatory cytokine release, cell migration and protects from chemically induced demyelination. Its signaling and expression are implicated in various diseases including multiple sclerosis, where its expression is increased in infiltrating immune cells in the white matter lesions. Here, for the first time, the EBI2 protein in the CNS cells in the human brain was examined. The function of the receptor in MO3.13 oligodendrocytes, as well as its role in remyelination in organotypic cerebellar slices, were investigated. Human brain sections were co-stained for EBI2 receptor and various markers of CNS-specific cells and the human oligodendrocyte cell line MO3.13 was used to investigate changes in EBI2 expression and cellular migration. Organotypic cerebellar slices prepared from wild-type and cholesterol 25-hydroxylase knock-out mice were used to study remyelination following lysophosphatidylcholine (LPC)-induced demyelination. The data showed that EBI2 receptor is present in OPCs but not in myelinating oligodendrocytes in the human brain and that EBI2 expression is temporarily upregulated in maturing MO3.13 oligodendrocytes. Moreover, we show that migration of MO3.13 cells is directly regulated by EBI2 and that its signaling is necessary for remyelination in cerebellar slices post-LPC-induced demyelination. The work reported here provides new information on the expression and role of EBI2 in oligodendrocytes and myelination and provides new tools for modulation of oligodendrocyte biology and therapeutic approaches for demyelinating diseases.


Asunto(s)
Encéfalo/citología , Cerebelo/citología , Oligodendroglía/citología , Receptores Acoplados a Proteínas G/metabolismo , Células Madre/citología , Animales , Encéfalo/metabolismo , Cerebelo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/genética , Remielinización , Células Madre/metabolismo
3.
Glia ; 68(1): 145-160, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31433095

RESUMEN

Astrocytes are important for information processing in the brain and they achieve this by fine-tuning neuronal communication via continuous uptake and release of biochemical modulators of neurotransmission and synaptic plasticity. Often overlooked are their important functions in mechanosensation. Indeed, astrocytes can detect pathophysiological changes in the mechanical properties of injured, ageing, or degenerating brain tissue. We have recently shown that astrocytes surrounding mechanically-stiff amyloid plaques upregulate the mechanosensitive ion channel, Piezo1. Moreover, ageing transgenic Alzheimer's rats harboring a chronic peripheral bacterial infection displayed enhanced Piezo1 expression in amyloid plaque-reactive astrocytes of the hippocampus and cerebral cortex. Here, we have shown that the bacterial endotoxin, lipopolysaccharide (LPS), also upregulates Piezo1 in primary mouse cortical astrocyte cultures in vitro. Activation of Piezo1, via the small molecule agonist Yoda1, enhanced Ca2+ influx in both control and LPS-stimulated astrocytes. Moreover, Yoda1 augmented intracellular Ca2+ oscillations but decreased subsequent Ca2+ influx in response to adenosine triphosphate (ATP) stimulation. Neither blocking nor activating Piezo1 affected cell viability. However, LPS-stimulated astrocyte cultures exposed to the Piezo1 activator, Yoda1, migrated significantly slower than reactive astrocytes treated with the mechanosensitive channel-blocking peptide, GsMTx4. Furthermore, our data show that activating Piezo1 channels inhibits the release of cytokines and chemokines, such as IL-1ß, TNFα, and fractalkine (CX3 CL1), from LPS-stimulated astrocyte cultures. Taken together, our results suggest that astrocytic Piezo1 upregulation may act to dampen neuroinflammation and could be a useful drug target for neuroinflammatory disorders of the brain.


Asunto(s)
Astrocitos/metabolismo , Señalización del Calcio/fisiología , Citocinas/metabolismo , Canales Iónicos/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Femenino , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL
4.
Glia ; 68(2): 356-375, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31596529

RESUMEN

Piezo1 is a mechanosensitive ion channel that facilitates the translation of extracellular mechanical cues to intracellular molecular signaling cascades through a process termed, mechanotransduction. In the central nervous system (CNS), mechanically gated ion channels are important regulators of neurodevelopmental processes such as axon guidance, neural stem cell differentiation, and myelination of axons by oligodendrocytes. Here, we present evidence that pharmacologically mediated overactivation of Piezo1 channels negatively regulates CNS myelination. Moreover, we found that the peptide GsMTx4, an antagonist of mechanosensitive cation channels such as Piezo1, is neuroprotective and prevents chemically induced demyelination. In contrast, the positive modulator of Piezo1 channel opening, Yoda-1, induces demyelination and neuronal damage. Using an ex vivo murine-derived organotypic cerebellar slice culture model, we demonstrate that GsMTx4 attenuates demyelination induced by the cytotoxic lipid, psychosine. Importantly, we confirmed the potential therapeutic effects of GsMTx4 peptide in vivo by co-administering it with lysophosphatidylcholine (LPC), via stereotactic injection, into the cerebral cortex of adult mice. GsMTx4 prevented both demyelination and neuronal damage usually caused by the intracortical injection of LPC in vivo; a well-characterized model of focal demyelination. GsMTx4 also attenuated both LPC-induced astrocyte toxicity and microglial reactivity within the lesion core. Overall, our data suggest that pharmacological activation of Piezo1 channels induces demyelination and that inhibition of mechanosensitive channels, using GsMTx4, may alleviate the secondary progressive neurodegeneration often present in the latter stages of demyelinating diseases.


Asunto(s)
Astrocitos/efectos de los fármacos , Enfermedades Desmielinizantes/tratamiento farmacológico , Canales Iónicos/efectos de los fármacos , Péptidos/farmacología , Animales , Astrocitos/metabolismo , Diferenciación Celular/fisiología , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Enfermedades Desmielinizantes/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Ratones , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos
5.
J Neurochem ; 144(6): 736-747, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29377126

RESUMEN

Sphingosine 1-phosphate receptors (S1PR) are G protein-coupled and compose a family with five subtypes, S1P1R-S1P5R. The drug Gilenya® (Novartis, Basel, Switzerland) (Fingolimod; FTY720) targets S1PRs and was the first oral therapy for patients with relapsing-remitting multiple sclerosis (MS). The phosphorylated form of FTY720 (pFTY720) binds S1PRs causing initial agonism, then subsequent receptor internalization and functional antagonism. Internalization of S1P1R attenuates sphingosine 1-phosphate (S1P)-mediated egress of lymphocytes from lymph nodes, limiting aberrant immune function in MS. pFTY720 also exerts direct actions on neurons and glial cells which express S1PRs. In this study, we investigated the regulation of pro-inflammatory chemokine release by S1PRs in enriched astrocytes and microglial cultures. Astrocytes and microglia were stimulated with lipopolysaccharide (LPS) and increases in C-X-C motif chemokine 5 (CXCL5), also known as LIX (lipopolysaccharide-induced CXC chemokine) expression were quantified. Results showed that pFTY720 attenuated LPS-induced CXCL5 (LIX) protein release from astrocytes, as did the S1P1R selective agonist, SEW2871. In addition, pFTY720 blocked messenger ribonucleic acid (mRNA) transcription of the chemokines, (i) CXCL5/LIX, (ii) C-X-C motif chemokine 10 (CXCL10) also known as interferon gamma-induced protein 10 (IP10) and (iii) chemokine (C-C motif) ligand 2 (CCL2) also known as monocyte chemoattractant protein 1 (MCP1). Interestingly, inhibition of sphingosine kinase attenuated LPS-induced increases in mRNA levels of all three chemokines, suggesting that LPS-TLR4 (Toll-like receptor 4) signalling may enhance chemokine expression via S1P-S1PR transactivation. Lastly, these observations were not limited to astrocytes since we also found that pFTY720 attenuated LPS-induced release of CXCL5 from microglia. These data highlight a role for S1PR signalling in regulating the levels of chemokines in glial cells and support the notion that pFTY720 efficacy in multiple sclerosis may involve the direct modulation of astrocytes and microglia.


Asunto(s)
Astrocitos/metabolismo , Quimiocina CXCL5/metabolismo , Inflamación/metabolismo , Microglía/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Astrocitos/efectos de los fármacos , Femenino , Clorhidrato de Fingolimod/administración & dosificación , Humanos , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas Wistar , Receptores de Lisoesfingolípidos/agonistas , Transducción de Señal
6.
Glia ; 65(7): 1119-1136, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28375547

RESUMEN

Oxidative stress is a pathological condition defined as an imbalance between production and removal of reactive oxygen species. This process causes structural cell damage, disrupts DNA repair and induces mitochondrial dysfunction. Many in vitro studies have used direct bolus application of H2 O2 to investigate the role of oxidative stress in cell culture. In this study, using mouse organotypic cerebellar slice cultures, the effects of H2 O2 -induced oxidative stress on myelination state were examined, using bolus concentrations of H2 O2 (0.1-1 mM) and low-continuous H2 O2 (∼20 µM) generated from glucose oxidase and catalase (GOX-CAT). Using these models, the potential therapeutic effects of pFTY720, an oral therapy used in multiple sclerosis, was also examined. We found bolus treatment of H2 O2 (0.5 mM) and, for the first time, low-continuous H2 O2 (GOX-CAT) to induce demyelination in organotypic slices. Both bolus H2 O2 and GOX-CAT treatments significantly decreased vimentin expression in these slice cultures as well as increased cell death in isolated astrocyte cultures. Importantly, pre-treatment with pFTY720 significantly attenuated both bolus H2 O2 and GOX-CAT-induced demyelination and the GOX-CAT-induced decrease in vimentin in cerebellar slices, without altering levels of the proinflammatory cytokines such as IL-6 and CX3CL1. We also observed increased SMI-32 immunoreactivity in the white matter tract induced by GOX-CAT indicating axonal damage, which was remarkably attenuated by pFTY720. Taken together, this data establishes a novel GOX-CAT model of demyelination and demonstrates that pFTY720 can act independently of inflammatory cytokines to attenuate decreases in vimentin, as well as axonal damage and demyelination induced by oxidative stress.


Asunto(s)
Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/metabolismo , Estrés Oxidativo/fisiología , Receptores de Lisoesfingolípidos/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/efectos de los fármacos , Astrocitos/fisiología , Encéfalo/citología , Proteínas de Unión al Calcio , Células Cultivadas , Cerebelo/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Enfermedades Desmielinizantes/tratamiento farmacológico , Femenino , Feto , Clorhidrato de Fingolimod/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/toxicidad , Inmunosupresores/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos , Glicoproteína Mielina-Oligodendrócito/metabolismo , Proteínas de Neurofilamentos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Receptores de Lisoesfingolípidos/genética , Proteínas Represoras/metabolismo , Proteínas de Pez Cebra/metabolismo
7.
J Cell Sci ; 128(21): 3878-87, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26359302

RESUMEN

Globoid cell leukodystrophy (Krabbe disease) is a rare infantile neurodegenerative disorder. Krabbe disease is caused by deficiency in the lysosomal enzyme galactocerebrosidase (GALC) resulting in accumulation, in the micromolar range, of the toxic metabolite galactosylsphingosine (psychosine) in the brain. Here we find that psychosine induces human astrocyte cell death probably via an apoptotic process in a concentration- and time-dependent manner (EC50 ∼ 15 µM at 4 h). We show these effects of psychosine are attenuated by pre-treatment with the sphingosine 1-phosphate receptor agonist pFTY720 (fingolimod) (IC50 ∼ 100 nM). Psychosine (1 µM, 10 µM) also enhances LPS-induced (EC50 ∼ 100 ng/ml) production of pro-inflammatory cytokines in mouse astrocytes, which is also attenuated by pFTY720 (1 µM). Most notably, for the first time, we show that psychosine, at a concentration found in the brains of patients with Krabbe disease (EC50 ∼ 100 nM), directly induces demyelination in mouse organotypic cerebellar slices in a manner that is independent of pro-inflammatory cytokine response and that pFTY720 (0.1 nM) significantly inhibits. These results support the idea that psychosine is a pathogenic agent in Krabbe disease and suggest that sphingosine 1-phosphate signalling could be a potential drug target for this disorder.


Asunto(s)
Enfermedades Desmielinizantes/inducido químicamente , Psicosina/farmacología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/metabolismo , Clorhidrato de Fingolimod/farmacología , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Leucodistrofia de Células Globoides/tratamiento farmacológico , Leucodistrofia de Células Globoides/metabolismo , Lipopolisacáridos/farmacología , Lisofosfolípidos , Ratones , Microscopía Fluorescente , Psicosina/uso terapéutico , Ratas , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados
8.
J Neuroinflammation ; 14(1): 159, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28810923

RESUMEN

BACKGROUND: Fractalkine/CX3CR1 signalling has been implicated in many neurodegenerative and neurological diseases of the central nervous system (CNS). This signalling pathway plays an important role in regulating reactive oxygen species (ROS), as well as itself being altered in conditions of oxidative stress. Here, we investigated the effects of recombinant fractalkine (rCX3CL1) in models of hydrogen peroxide (H2O2)-induced demyelination and astrocyte toxicity, within organotypic cerebellar slice cultures. METHODS: Organotypic cerebellar slice cultures were generated from postnatal day 10 C57BL/6J mice to assess myelination. Immunohistochemistry was used to measure the degree of myelination. Fluorescent images were obtained using a leica SP8 confocal microscope and data analysed using ImageJ software. RESULTS: We show here, for the first time, that rCX3CL1 significantly attenuated bolus H2O2-induced demyelination as measured by expression of myelin basic protein (MBP) and attenuated reduced vimentin expression. Using the GOX-CAT system to continuously generate low levels of H2O2 and induce demyelination, we observed similar protective effects of rCX3CL1 on MBP and MOG fluorescence, although in this model, the decrease in vimentin expression was not altered. CONCLUSIONS: This data indicates possible protective effects of fractalkine signalling in oxidative stress-induced demyelination in the central nervous system. This opens up the possibility of fractalkine receptor (CX3CR1) modulation as a potential new target for protecting against oxidative stress-induced demyelination in both inflammatory and non-inflammatory nervous system disorders.


Asunto(s)
Cerebelo/efectos de los fármacos , Cerebelo/patología , Quimiocina CX3CL1/uso terapéutico , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/prevención & control , Peróxido de Hidrógeno/toxicidad , Animales , Animales Recién Nacidos , Receptor 1 de Quimiocinas CX3C/metabolismo , Catalasa/toxicidad , Muerte Celular/efectos de los fármacos , Femenino , Gliosis/inducido químicamente , Gliosis/prevención & control , Glucosa Oxidasa/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/metabolismo , Proteínas de Neurofilamentos/metabolismo , Técnicas de Cultivo de Órganos , Especies Reactivas de Oxígeno/metabolismo
9.
J Neuroinflammation ; 14(1): 250, 2017 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-29246262

RESUMEN

BACKGROUND: The G protein-coupled receptor EBI2 (Epstein-Barr virus-induced gene 2) is activated by 7α, 25-dihydroxycholesterol (7α25HC) and plays a role in T cell-dependant antibody response and B cell migration. Abnormal EBI2 signaling is implicated in a range of autoimmune disorders; however, its role in the CNS remains poorly understood. METHODS: Here we characterize the role of EBI2 in myelination under normal and pathophysiological conditions using organotypic cerebellar slice cultures and EBI2 knock-out (KO) animals. RESULTS: We find that MBP expression in brains taken from EBI2 KO mice is delayed compared to those taken from wild type (WT) mice. In agreement with these in vivo findings, we show that antagonism of EBI2 reduces MBP expression in vitro. Importantly, we demonstrate that EBI2 activation attenuates lysolecithin (LPC)-induced demyelination in mouse organotypic slice cultures. Moreover, EBI2 activation also inhibits LPC-mediated release of pro-inflammatory cytokines such as IL6 and IL1ß in cerebellar slices. CONCLUSIONS: These results, for the first time, display a role for EBI2 in myelin development and protection from demyelination under pathophysiological conditions and suggest that modulation of this receptor may be beneficial in neuroinflammatory and demyelinating disorders such as multiple sclerosis.


Asunto(s)
Cerebelo/metabolismo , Enfermedades Desmielinizantes/metabolismo , Vaina de Mielina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Enfermedades Desmielinizantes/inducido químicamente , Lisofosfatidilcolinas/toxicidad , Ratones , Ratones Noqueados , Proteína Básica de Mielina/biosíntesis , Técnicas de Cultivo de Órganos
10.
J Neuroinflammation ; 13: 31, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26856814

RESUMEN

BACKGROUND: BAF312 (Siponimod) is a dual agonist at the sphingosine-1 phosphate receptors, S1PR1 and S1PR5. This drug is currently undergoing clinical trials for the treatment of secondary progressive multiple sclerosis (MS). Here, we investigated the effects of BAF312 on isolated astrocyte and microglia cultures as well as in slice culture models of demyelination. METHODS: Mouse and human astrocytes were treated with S1PR modulators and changes in the levels of pERK, pAkt, and calcium signalling as well as S1PR1 internalization and cytokine levels was investigated using Western blotting, immunochemistry, ELISA and confocal microscopy. Organotypic slice cultures were prepared from the cerebellum of 10-day-old mice and treated with lysophosphatidylcholine (LPC), psychosine and/or S1PR modulators, and changes in myelination states were measured by fluorescence of myelin basic protein and neurofilament H. RESULTS: BAF312 treatment of human and mouse astrocytes activated pERK, pAKT and Ca(2+) signalling as well as inducing S1PR1 internalization. Notably, activation of S1PR1 increased pERK and pAKT in mouse astrocytes while both S1PR1 and S1PR3 equally increased pERK and pAKT in human astrocytes, suggesting that the coupling of S1PR1 and S1PR3 to pERK and pAKT differ in mouse and human astrocytes. We also observed that BAF312 moderately attenuated lipopolysaccharide (LPS)- or TNFα/IL17-induced levels of IL6 in both astrocyte and microglia cell cultures. In organotypic slice cultures, BAF312 reduced LPC-induced levels of IL6 and attenuated LPC-mediated demyelination. We have shown previously that the toxic lipid metabolite psychosine induces demyelination in organotypic slice cultures, without altering the levels of cytokines, such as IL6. Importantly, psychosine-induced demyelination was also attenuated by BAF312. CONCLUSIONS: Overall, this study suggests that BAF312 can modulate glial cell function and attenuate demyelination, highlighting this drug as a further potential therapy in demyelinating disorders, beyond MS.


Asunto(s)
Antiinflamatorios/farmacología , Astrocitos/efectos de los fármacos , Azetidinas/farmacología , Compuestos de Bencilo/farmacología , Cerebelo/citología , Enfermedades Desmielinizantes/tratamiento farmacológico , Animales , Animales Recién Nacidos , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Humanos , Inmunosupresores/farmacología , Técnicas In Vitro , Indanos/farmacología , Interleucina-6/metabolismo , Lisofosfatidilcolinas/farmacología , Ratones , Proteína Básica de Mielina/metabolismo , Técnicas de Cultivo de Órganos , Oxadiazoles/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Tiofenos/farmacología , Factores de Tiempo , beta-Alanina/análogos & derivados , beta-Alanina/farmacología , eIF-2 Quinasa/metabolismo
11.
J Neuroinflammation ; 13(1): 189, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27549131

RESUMEN

BACKGROUND: The fractalkine (CX3CR1) ligand is expressed in astrocytes and reported to be neuroprotective. When cleaved from the membrane, soluble fractalkine (sCX3CL1) activates the receptor CX3CR1. Although somewhat controversial, CX3CR1 is reported to be expressed in neurons and microglia. The membrane-bound form of CX3CL1 additionally acts as an adhesion molecule for microglia and infiltrating white blood cells. Much research has been done on the role of fractalkine in neuronal cells; however, little is known about the regulation of the CX3CL1 ligand in astrocytes. METHODS: The mechanisms involved in the up-regulation and cleavage of CX3CL1 from human astrocytes were investigated using immunocytochemistry, Q-PCR and ELISA. All statistical analysis was performed using GraphPad Prism 5. RESULTS: A combination of ADAM17 (TACE) and ADAM10 protease inhibitors was found to attenuate IL-1ß-, TNF-α- and IFN-γ-induced sCX3CL1 levels in astrocytes. A specific ADAM10 (but not ADAM17) inhibitor also attenuated these effects, suggesting ADAM10 proteases induce release of sCX3CL1 from stimulated human astrocytes. A p38 MAPK inhibitor also attenuated the levels of sCX3CL1 upon treatment with IL-1ß, TNF-α or IFN-γ. In addition, an IKKß inhibitor significantly reduced the levels of sCX3CL1 induced by IL-1ß or TNF-α in a concentration-dependent manner, suggesting a role for the NF-kB pathway. CONCLUSIONS: In conclusion, this study shows that the release of soluble astrocytic fractalkine is regulated by ADAM10 proteases with p38 MAPK also playing a role in the fractalkine shedding event. These findings are important for understanding the role of CX3CL1 in healthy and stimulated astrocytes and may benefit our understanding of this pathway in neuro-inflammatory and neurodegenerative diseases.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Quimiocina CX3CL1/metabolismo , Citocinas/farmacología , Proteínas de la Membrana/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Cultivadas , Quimiocina CX3CL1/genética , Medio de Cultivo Libre de Suero/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Metaloproteinasas de la Matriz/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo
12.
Glia ; 63(2): 341-51, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25297897

RESUMEN

The G protein-coupled receptor EBI2 (Epstein-Barr virus-induced gene 2) is activated by 7α, 25-dihydroxycholesterol (7α25HC) and plays a role in T cell-dependant antibody response and B cell migration. Aberrant EBI2 signaling is implicated in a range of autoimmune disorders however its role in the CNS remains unknown. Here we characterize the functional role of EBI2 in GLIA cells using primary human astrocytes and EBI2 knockout animals. We find human and mouse astrocytes express EBI2 and the enzymes necessary for synthesis and degradation of 7α25HC. In astrocytes, EBI2 activation stimulates ERK phosphorylation, Ca(2+) signaling and induces cellular migration. These results, for the first time, demonstrate a role for EBI2 in astrocyte function and suggest that modulation of this receptor may be beneficial in neuroinflammatory disorders.


Asunto(s)
Astrocitos/metabolismo , Movimiento Celular/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Análisis de Varianza , Animales , Animales Recién Nacidos , Área Bajo la Curva , Señalización del Calcio/fisiología , Diferenciación Celular , Movimiento Celular/genética , Corteza Cerebral/citología , Colesterol/farmacocinética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , Transfección
13.
Cytokine ; 73(1): 196-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25769808

RESUMEN

BACKGROUND: Schizophrenia is a mental disorder, where genetic and environmental factors contribute to disease onset and progression. The immune system appears to play a role in schizophrenia, where altered cytokines levels and autoantibodies have been described. Notably however, to our knowledge, IL23 levels have not before been measured in schizophrenia patients treated with depot medication. METHODS: We examined IL23 levels in serum samples obtained from patients with schizophrenia, treated with depot medication (n=35) compared with healthy controls (n=38) and correlated these levels with treatment time, patient age and illness severity. RESULTS: IL23 levels were raised in depot treated groups compared with healthy controls. No correlation was observed, however, between IL23 levels and treatment time, patient age or illness severity. CONCLUSIONS: IL23 levels are raised in schizophrenia patients prescribed with depot medication, supporting the role of aberrant cytokine signalling in schizophrenia.


Asunto(s)
Antipsicóticos/uso terapéutico , Subunidad p19 de la Interleucina-23/sangre , Esquizofrenia/sangre , Esquizofrenia/tratamiento farmacológico , Adulto , Preparaciones de Acción Retardada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo , Adulto Joven
14.
Glia ; 62(5): 725-35, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24677511

RESUMEN

The family of interleukin 17 receptors (IL17Rs), subtypes IL17RA-IL17RE, is targeted by the group of pro-inflammatory IL17 cytokines (IL17A-F) and moreover the newly developed anti-IL17A antibody secukinumab (AIN457) has shown promise in Phase II trials in multiple sclerosis. Here, we show that human astrocytes, isolated from a fetal cerebral cortex, express IL17RA and IL17RC and in vitro treatment with IL17A increases protein levels of IL6 in human astrocytes, which is enhanced in the presence of TNFα, as determined by homogeneous time resolved fluorescence. Studies on acutely isolated mouse astrocytes are comparable to human astrocytes although the protein levels of IL6 are lower in mouse astrocytes, which also show a lower response to IL17F and IL1ß in promoting IL6 levels. In human astrocytes, IL17A and TNFα also induce mRNA expression of IL6, IL8 and the Th17 cytokines CXCL1, CXCL2, and CCL20, with little effect on Th1 cytokines CXCL9, CXCL10, and CXCL11. The effects of IL17A are associated with nuclear translocation of the NF-κB transcription factor, as determined by immunocytochemistry, where treatment of human astrocytes with the inhibitors of the NF-κB pathway and with secukinumab inhibits the IL17A and IL17A/TNFα-induced increase in nuclear translocation of NF-κB and levels of IL6. Taken together the data shows that IL17A signaling plays a key role in regulating the levels of cytokines, such as IL6, in human astrocytes via a mechanism that involves NF-κB signaling and that selective inhibition of IL17A signaling attenuates levels of pro-inflammatory molecules in astrocytes.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Interleucina-17/antagonistas & inhibidores , Interleucina-17/farmacología , Interleucina-6/biosíntesis , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales Humanizados , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
15.
J Neurochem ; 130(3): 360-73, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24749734

RESUMEN

The parkin-associated endothelial-like receptor (PAELR, GPR37) is an orphan G protein-coupled receptor that interacts with and is degraded by parkin-mediated ubiquitination. Mutations in parkin are thought to result in PAELR accumulation and increase neuronal cell death in Parkinson's disease. In this study, we find that the protein interacting with C-kinase (PICK1) interacts with PAELR. Specifically, the Postsynaptic density protein-95/Discs large/ZO-1 (PDZ) domain of PICK1 interacted with the last three residues of the c-terminal (ct) located PDZ motif of PAELR. Pull-down assays indicated that recombinant and native PICK1, obtained from heterologous cells and rat brain tissue, respectively, were retained by a glutathione S-transferase fusion of ct-PAELR. Furthermore, coimmunoprecipitation studies isolated a PAELR-PICK1 complex from transiently transfected cells. PICK1 interacts with parkin and our data showed that PICK1 reduces PAELR expression levels in transiently transfected heterologous cells compared to a PICK1 mutant that does not interact with PAELR. Finally, PICK1 over-expression in HEK293 cells reduced cell death induced by PAEALR over-expression during rotenone treatment and these effects of PICK1 were attenuated during inhibition of the proteasome. These results suggest a role for PICK1 in preventing PAELR-induced cell toxicity.


Asunto(s)
Proteínas Portadoras/fisiología , Muerte Celular/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteínas Nucleares/fisiología , Receptores Acoplados a Proteínas G/fisiología , Secuencia de Aminoácidos , Animales , Western Blotting , Proteínas Portadoras/metabolismo , Células Cultivadas , Proteínas del Citoesqueleto , ADN Complementario/biosíntesis , ADN Complementario/genética , Femenino , Glutatión Transferasa/metabolismo , Células HEK293 , Humanos , Inmunohistoquímica , Inmunoprecipitación , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Dominios PDZ , Reacción en Cadena de la Polimerasa , Conformación Proteica , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo
16.
Biomedicines ; 12(3)2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38540207

RESUMEN

Krabbe's disease (KD) is caused by mutations in the lysosomal enzyme galactocerebrosidase and is associated with psychosine toxicity. The sphingosine 1-phosphate receptor (S1PR) agonist fingolimod (FTY720) attenuates psychosine-induced cell death of human astrocytes, demyelination in cerebellar slices, as well as demyelination in the central nervous system of twitcher mice. Psychosine also accumulates in the peripheral nervous system in twitcher mice; however, effects of fingolimod on this peripheral myelin have not been examined. The aim of this study was to investigate the effects of fingolimod administration on peripheral and central markers of myelination. Here, we report that fingolimod administration (1 mg/kg/day) from postnatal day 5 (PND) onwards did not alter peripheral demyelination in the sciatic nerve of twitcher mice, despite significantly reducing myelin debris, glial reactivity, and neuronal damage in the cerebellum. We also find fingolimod administration improves twitching and mobility scores in twitcher mice. Importantly, we find that fingolimod significantly increases the lifespan of twitcher mice by approximately 5 days. These findings suggest differential effects of fingolimod on peripheral and central neuropathy in twitcher mice, which may explain its modest efficacy on behavior and lifespan.

17.
Mol Neurobiol ; 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38558359

RESUMEN

Krabbe disease (KD) is a rare demyelinating disorder characterized by demyelination caused by mutations in the GALC gene, resulting in toxic accumulation of psychosine. Psychosine has been identified as detrimental to oligodendrocytes, leading to demyelination through diverse hypothesized pathways. Reducing demyelination is essential to maintain neurological function in KD; however, therapeutic interventions are currently limited. Acetylcholinesterase inhibitors (AChEi) are commonly used for symptomatic management of Alzheimer's Disease and are suggested to have potential disease-modifying effects, including regulating myelin state. In particular, donepezil, an AChEi, has demonstrated promising effects in cellular and animal models, including promotion of the expression of myelin-related genes and reduction of glial cell reactivity. This drug also acts as an agonist for sigma-1 receptors (Sig-1R), which are implicated in demyelination diseases. In the context of drug repurposing, here, we demonstrate that administration of donepezil has protective effects in the twitcher mouse model of KD. We provide data showing that donepezil preserves myelin and reduces glial cell reactivity in the brains of twitcher mice. Moreover, donepezil also improves behavioral phenotypes and increases lifespan in twitcher animals. These findings suggest that donepezil, with its dual activity as an AChE inhibitor and Sig-1R agonist, may hold promise as a therapeutic candidate for demyelinating diseases, including KD.

18.
BMC Psychiatry ; 13: 146, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23705829

RESUMEN

BACKGROUND: The neurotrophic factor, S100B, is released primarily from astrocytes, with serum and CSF levels of S100B reported as altered in schizophrenia. However, many of these reports are contradictory. Here, serum levels of S100B in schizophrenia and influence of age, gender, medication and illness severity were examined. METHODS: Serum S100B levels were measured in patients with schizophrenia treated with clozapine. Lifestyle, metabolic and illness severity parameters were correlated with S100B concentrations. RESULTS: Data showed raised serum levels of S100B in schizophrenia female patients, but not male patients, compared to controls. Correlation analysis demonstrated a positive association between S100B serum concentrations and BMI. CONCLUSIONS: This study supports previous findings that adipocytes may contribute to S100B serum concentrations in females, in addition to astrocytes. This study also supports the hypothesis that metabolic effects of medication, lifestyle choices and the illness itself, may be contributing factors to altered levels of S100B.


Asunto(s)
Factores de Crecimiento Nervioso/sangre , Proteínas S100/sangre , Esquizofrenia/sangre , Caracteres Sexuales , Adulto , Anciano , Antipsicóticos , Clozapina/uso terapéutico , Femenino , Humanos , Estilo de Vida , Masculino , Persona de Mediana Edad , Subunidad beta de la Proteína de Unión al Calcio S100 , Esquizofrenia/tratamiento farmacológico , Índice de Severidad de la Enfermedad
19.
Biomedicines ; 11(5)2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37238985

RESUMEN

The role of altered myelin in the onset and development of schizophrenia and changes in myelin due to antipsychotics remains unclear. Antipsychotics are D2 receptor antagonists, yet D2 receptor agonists increase oligodendrocyte progenitor numbers and limit oligodendrocyte injury. Conflicting studies suggest these drugs promote the differentiation of neural progenitors to oligodendrocyte lineage, while others report antipsychotics inhibit the proliferation and differentiation of oligodendrocyte precursors. Here, we utilised in-vitro (human astrocytes), ex-vivo (organotypic slice cultures) and in-vivo (twitcher mouse model) experimental study designs of psychosine-induced demyelination, a toxin that accumulates in Krabbe disease (KD), to investigate direct effects of antipsychotics on glial cell dysfunction and demyelination. Typical and atypical antipsychotics, and selective D2 and 5HT2A receptor antagonists, attenuated psychosine-induced cell viability, toxicity, and morphological aberrations in human astrocyte cultures. Haloperidol and clozapine reduced psychosine-induced demyelination in mouse organotypic cerebellar slices. These drugs also attenuated the effects of psychosine on astrocytes and microglia and restored non-phosphorylated neurofilament levels, indicating neuroprotective effects. In the demyelinating twitcher mouse model of KD, haloperidol improved mobility and significantly increased the survival of these animals. Overall, this study suggests that antipsychotics directly regulate glial cell dysfunction and exert a protective effect on myelin loss. This work also points toward the potential use of these pharmacological agents in KD.

20.
Pharmaceutics ; 15(2)2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36839682

RESUMEN

Glioblastoma multiforme (GBM) is the most common adult primary brain malignancy, with dismal survival rates of ~14.6 months. The current standard-of-care consists of surgical resection and chemoradiotherapy, however the treatment response is limited by factors such as tumour heterogeneity, treatment resistance, the blood-brain barrier, and immunosuppression. Several immunotherapies have undergone clinical development for GBM but demonstrated inadequate efficacy, yet future combinatorial approaches are likely to hold more promise. Olaparib is FDA-approved for BRCA-mutated advanced ovarian and breast cancer, and clinical studies have revealed its utility as a safe and efficacious radio- and chemo-sensitiser in GBM. The ability of Olaparib to enhance natural killer (NK) cell-mediated responses has been reported in prostate, breast, and lung cancer. This study examined its potential combination with NK cell therapies in GBM by firstly investigating the susceptibility of the GBM cell line T98G to NK cells and, secondly, examining whether Olaparib can sensitise T98G cells to NK cell-mediated responses. Here, we characterise the NK receptor ligand profile of T98G cells and demonstrate that Olaparib does not dampen T98G susceptibility to NK cells or elicit immunomodulatory effects on the function of NK cells. This study provides novel insights into the potential combination of Olaparib with NK cell therapies for GBM.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA