Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
F1000Res ; 9: 1336, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-34745570

RESUMEN

The COVID-19 pandemic has posed and is continuously posing enormous societal and health challenges worldwide. The research community has mobilized to develop novel projects to find a cure or a vaccine, as well as to contribute to mass testing, which has been a critical measure to contain the infection in several countries. Through this article, we share our experiences and learnings as a group of volunteers at the Centre for Genomic Regulation (CRG) in Barcelona, Spain. As members of the ORFEU project, an initiative by the Government of Catalonia to achieve mass testing of people at risk and contain the epidemic in Spain, we share our motivations, challenges and the key lessons learnt, which we feel will help better prepare the global society to address similar situations in the future.


Asunto(s)
COVID-19 , Prueba de COVID-19 , Genómica , Humanos , Pandemias , SARS-CoV-2 , Voluntarios
2.
J Neuroimmune Pharmacol ; 13(1): 6-23, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28776122

RESUMEN

Remyelination occurs in demyelinated lesions in multiple sclerosis (MS) and pharmacological treatments that enhance this process will critically impact the long term functional outcome in the disease. Sildenafil, a cyclic GMP (cGMP)-specific phosphodiesterase 5 inhibitor (PDE5-I), is an oral vasodilator drug extensively used in humans for treatment of erectile dysfunction and pulmonary arterial hypertension. PDE5 is expressed in central nervous system (CNS) neuronal and glial populations and in endothelial cells and numerous studies in rodent models of neurological disease have evidenced the neuroprotective potential of PDE5-Is. Using myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) as a MS model, we previously showed that daily administration of sildenafil starting at peak disease rapidly ameliorates clinical symptoms while administration at symptoms onset prevents disease progression. These beneficial effects of the drug involved down-regulation of adaptive and innate immune responses, protection of axons and oligodendrocytes (OLs) and promotion of remyelination. In this work we have investigated mechanisms involved in the remyelinating effect of sildenafil. Using demyelinated organotypic cerebellar slice cultures we demonstrate that sildenafil stimulates remyelination by direct effects on CNS cells in a nitric oxide (NO)-cGMP-protein kinase G (PKG)-dependent manner. We also show that sildenafil treatment enhances OL maturation and induces expression of the promyelinating factor ciliary neurotrophic factor (CNTF) in spinal cord of EAE mice and in cerebellar slice cultures. Furthermore, we demonstrate that sildenafil promotes a M2 phenotype in bone marrow derived macrophages (BMDM) and increases myelin phagocytosis in these cells and in M2 microglia/macrophages in the spinal cord of EAE mice. Taken together these data indicate that promotion of OL maturation directly or through induction of growth factor expression, regulation of microglia/macrophage inflammatory phenotype and clearance of myelin debris may be relevant mechanisms involved in sildenafil enhancement of remyelination in demyelinated tissue and further support the contention that this well tolerated drug could be useful for ameliorating MS pathology.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Oligodendroglía/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Remielinización/efectos de los fármacos , Citrato de Sildenafil/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Cerebelo/efectos de los fármacos , Cerebelo/patología , Femenino , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Médula Espinal/efectos de los fármacos , Médula Espinal/patología
3.
Exp Neurol ; 251: 58-71, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24211383

RESUMEN

In addition to detrimental inflammation, widespread axon degeneration is an important feature of multiple sclerosis (MS) pathology and a major correlate for permanent clinical deficits. Thus, treatments that combine immunomodulatory and neuroprotective effects are beneficial for MS. Using myelin oligodendrocyte glycoprotein peptide 35-55 (MOG)-induced experimental autoimmune encephalomyelitis (EAE) as a model of MS, we recently showed that daily treatment with the phosphodiesterase 5 (PDE5) inhibitor sildenafil at peak disease rapidly ameliorates clinical symptoms and neuropathology (Pifarre et al., 2011). We have now investigated the immunomodulatory and neuroprotective actions of sildenafil treatment from the onset of EAE when the immune response prevails and show that early administration of the drug prevents disease progression. Ultrastructural analysis of spinal cord evidenced that sildenafil treatment preserves axons and myelin and increases the number of remyelinating axons. Immunostaining of oligodendrocytes at different stages of differentiation showed that sildenafil protects immature and mature myelinating oligodendrocytes. Brain-derived neurotrophic factor (BDNF), a recognized neuroprotectant in EAE, was up-regulated by sildenafil in immune and neural cells suggesting its implication in the beneficial effects of the drug. RNA microarray analysis of spinal cord revealed that sildenafil up-regulates YM-1, a marker of the alternative macrophage/microglial M2 phenotype that has neuroprotective and regenerative properties. Immunostaining confirmed up-regulation of YM-1 while the classical macrophage/microglial activation marker Iba-1 was down-regulated. Microarray analysis also showed a notable up-regulation of several members of the granzyme B cluster (GrBs). Immunostaining revealed expression of GrBs in Foxp3+-T regulatory cells (Tregs) suggesting a role for these proteases in sildenafil-induced suppression of T effector cells (Teffs). In vitro analysis of splenocytes from sildenafil-treated animals showed down-regulation of Th1/Th2/Th17 responses while Tregs were up-regulated. Additionally, sildenafil treatment prevented MOG-specific IgG2b accumulation in serum. Taken together these data demonstrates that daily sildenafil treatment from the initiation of EAE symptoms prevents further clinical deterioration by stimulating immunomodulatory and neuroprotective mechanisms. Importantly, we also show here that sildenafil enhances the ability of human Tregs from healthy donors to down-regulate the proliferation of Teffs in vitro, strongly supporting the potential of sildenafil for therapeutic intervention in MS.


Asunto(s)
Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Regulación de la Expresión Génica/inmunología , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Piperazinas/uso terapéutico , Sulfonas/uso terapéutico , Linfocitos T/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/patología , Axones/ultraestructura , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Femenino , Adyuvante de Freund/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/metabolismo , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Oligodendroglía/efectos de los fármacos , Purinas/uso terapéutico , Índice de Severidad de la Enfermedad , Citrato de Sildenafil , Linfocitos T/metabolismo , Factores de Tiempo
4.
Glia ; 61(4): 587-600, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23322593

RESUMEN

Interleukin (IL)-6 is crucial for the induction of many murine models of autoimmunity including experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. While IL-6-deficient mice (IL-6 KO) are resistant to EAE, we showed previously that in transgenic mice with astrocyte-targeted production of IL-6-restricted to the cerebellum (GFAP-IL6), EAE induced with MOG(35-55) was redirected away from the spinal cord to the cerebellum. To further establish the importance of IL-6 produced in the central nervous system, we have generated mice producing IL-6 essentially only in the brain by crossing the GFAP-IL6 mice with IL-6 KO mice. Interestingly, GFAP-IL6-IL-6 KO mice showed a milder but almost identical phenotype as the GFAP-IL6 mice, which correlated with a lower load of inflammatory cells and decreased microglial reactivity. These results indicate that not only is cerebellar IL-6 production and eventual leakage into the peripheral compartment the dominating factor controlling this type of EAE but that it can also facilitate induction of autoimmunity in the absence of normal systemic IL-6 production.


Asunto(s)
Astrocitos/patología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Interleucina-6/biosíntesis , Animales , Astrocitos/metabolismo , Células Cultivadas , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Interleucina-6/deficiencia , Interleucina-6/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
5.
Neurochem Int ; 62(1): 70-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23178801

RESUMEN

We recently reported that administration of the non-selective cyclic GMP-phosphodiesterase (cGMP-PDE) inhibitor zaprinast to cortically cryoinjured rats results three days post-lesion in reduced neuronal cell death that was associated to decreased macrophage/microglial activation and oxidative stress and increased astrogliosis and angiogenesis. Similar effects have been observed in cryoinjured animals overexpressing metallothioneins I/II (MT-I/II), metal-binding cysteine-rich proteins that are up-regulated in response to injury. In this work we have examined the effect of administration of the selective PDE5 inhibitor sildenafil (10mg/kg, sc) 2h before and 24 and 48h after induction of cortical cryolesion in wild-type and MT-I/II-deficient mice. Our results show that in wild-type animals sildenafil induces similar changes in glial reactivity, angiogenesis and antioxidant and antiapoptotic effects in the cryolesioned cortex as those observed in rats with zaprinast, indicating that inhibition of PDE5 is responsible for the neuroprotective actions. However, these effects were not observed in mice deficient in MT-I/II. We further show that sildenafil significantly increases MT-I/II protein levels in homogenates of lesioned cortex and MT-I/II immunostaining in glial cells around the lesion. Taken together these results indicate that cGMP-mediated pathways regulate expression of MT-I/II and support the involvement of these proteins in the neuroprotective effects of sildenafil in focal brain lesion.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Metalotioneína/fisiología , Fármacos Neuroprotectores , Inhibidores de Fosfodiesterasa/farmacología , Piperazinas/farmacología , Sulfonas/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Lesiones Encefálicas/patología , Corteza Cerebral/lesiones , Corteza Cerebral/patología , Frío , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Ensayo de Inmunoadsorción Enzimática , Gliosis/patología , Inmunohistoquímica , Activación de Macrófagos , Metalotioneína/genética , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Purinas/farmacología , Purinonas/farmacología , Citrato de Sildenafil , Regulación hacia Arriba/efectos de los fármacos
6.
Acta Neuropathol ; 121(4): 499-508, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21234581

RESUMEN

Cyclic GMP (cGMP)-mediated pathways regulate inflammatory responses in immune and CNS cells. Recently, cGMP phosphodiesterase inhibitors such as sildenafil, commonly used to treat sexual dysfunction in humans including multiple sclerosis (MS) patients, have been reported to be neuroprotective in animal models of stroke, Alzheimer's disease, and focal brain lesion. In this work, we have examined if sildenafil ameliorates myelin oligodendrocyte glycoprotein peptide (MOG35₋55)-induced experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. We show for the first time that treatment with sildenafil after disease onset markedly reduces the clinical signs of EAE by preventing axonal loss and promoting remyelination. Furthermore, sildenafil decreases CD3+ leukocyte infiltration and microglial/macrophage activation in the spinal cord, while increasing forkhead box transcription factor 3-expressing T regulatory cells (Foxp3 Tregs). However, sildenafil treatment did not significantly affect MOG35₋55-stimulated proliferation or release of Th1/Th2 cytokines in splenocytes but decreased ICAM-1 in spinal cord infiltrated cells. The presence of reactive astrocytes forming scar-like structures around infiltrates was enhanced by sildenafil suggesting a possible mechanism for restriction of leukocyte spread into healthy parenchyma. These results highlight novel actions of sildenafil that may contribute to its beneficial effects in EAE and suggest that treatment with this widely used and well-tolerated drug may be a useful therapeutic intervention to ameliorate MS neuropathology.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Piperazinas/uso terapéutico , Médula Espinal/patología , Sulfonas/uso terapéutico , Animales , Complejo CD3/metabolismo , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/tratamiento farmacológico , Gliosis/etiología , Glicoproteínas/efectos adversos , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito , Proteínas de Neurofilamentos/metabolismo , Fragmentos de Péptidos/efectos adversos , Purinas/uso terapéutico , Citrato de Sildenafil , Médula Espinal/efectos de los fármacos , Factores de Tiempo
7.
Neurochem Int ; 57(4): 367-74, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20302900

RESUMEN

Natriuretic peptides and their receptors are widely expressed in mammalian CNS and increasing evidence implicates them in the regulation of neural development, synaptic transmission and processing of information, and neuroprotection. Although the peptides have been mainly localized in neuronal populations they are also produced in glial cells. Astroglia and microglia also express functional natriuretic peptide receptors that can regulate important physiological responses. In this article we review evidence on the localization of natriuretic peptides and their receptors in astroglial and microglial cells and summarize data supporting the participation of this signalling system in neuron-glia and glia-brain blood vessel communication relevant to CNS function.


Asunto(s)
Péptidos Natriuréticos/metabolismo , Péptidos Natriuréticos/fisiología , Neuroglía/metabolismo , Neuroglía/fisiología , Animales , Sistema Nervioso Central/metabolismo , Humanos , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/fisiología , Transducción de Señal/fisiología
8.
J Neurochem ; 112(3): 807-17, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20002517

RESUMEN

Recent evidence obtained in cultured glial cells indicates that cGMP-mediated pathways regulate cytoskeleton dynamics, glial fibrillary acidic protein expression and motility in astrocytes, as well as inflammatory gene expression in microglia, suggesting a role in the regulation of the glial reactive phenotype. The aim of this work was to examine if cGMP regulates the glial inflammatory response in vivo following CNS damage caused by a focal cryolesion onto the cortex in rats. Results show that treatment with the cGMP phosphodiesterase inhibitor zaprinast (10 mg/kg i.p.) 2 h before and 24 and 48 h after the lesion results 3 days post-lesion in notably enhanced astrogliosis manifested by increased glial fibrillary acidic protein immunoreactivity and protein levels around the lesion. In contrast, zaprinast decreased the number of round/ameboid lectin-positive cells and the expression of the activated microglia/macrophage markers Iba-1 and CD11b indicating decreased recruitment and activation of these cells. This altered inflammatory response is accompanied by a decrease in protein oxidative stress, apoptotic cell death and neuronal degeneration. In addition, zaprinast enhanced angiogenesis in the lesioned cortex probably as a result of vascular endothelial growth factor expression in reactive astrocytes. These results suggest that regulation of the glial inflammatory response may contribute to the reported neuroprotective effects of cGMP-phosphodiesterase inhibitors in brain injury.


Asunto(s)
Lesiones Encefálicas , Neovascularización Fisiológica/efectos de los fármacos , Neuroglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Fosfodiesterasa/farmacología , Purinonas/farmacología , Animales , Lesiones Encefálicas/patología , Lesiones Encefálicas/fisiopatología , Lesiones Encefálicas/prevención & control , Antígeno CD11b/metabolismo , Proteínas de Unión al Calcio/metabolismo , Recuento de Células/métodos , Muerte Celular/efectos de los fármacos , Corteza Cerebral/patología , Criocirugía/efectos adversos , Modelos Animales de Enfermedad , Esquema de Medicación , Etiquetado Corte-Fin in Situ/métodos , Lectinas/metabolismo , Masculino , Proteínas de Microfilamentos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
Int J Biochem Cell Biol ; 41(8-9): 1719-30, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19433313

RESUMEN

NO-sensitive guanylyl cyclase (GC(NO)), the major NO target, is involved in important regulatory functions in the cardiovascular, gastrointestinal and central nervous systems. GC(NO) exists as heterodimers of alpha(1/2) and beta1 subunits. Deletion of the obligate beta1 dimerizing partner abrogates NO/cGMP signaling and shortens the life span of KO mice. Localization studies in the CNS have shown that beta1 is more widespread than alpha subunits and in some areas is the only GC(NO) subunit expressed, suggesting that beta1 may have GC(NO)-independent functions. GC(NO) is predominantly cytosolic, but association to membranes and other intracellular structures has been described. Here, we show localization of beta1 in cytoplasm and nucleus of cells expressing alpha subunits and GC(NO) activity (astrocytes, C6 cells), as well as in cells devoid of alpha subunits and GC(NO) activity (microglia). In both cell types beta1 associates peripherally to chromosomes in all phases of mitosis. Immunodepletion of beta1 in C6 cells enhances chromatin condensation in an in vitro assay. Moreover, silencing beta1 by siRNA induces cell cycle re-entry as determined by flow cytometry, and increases proliferation rate in a MTT-assay, whereas infection with beta1-containing adenovirus has the opposite effect. These actions are independent of cGMP formation. We postulate that beta1 is a multifunctional protein that regulates chromatin condensation and cell cycle progression, in addition to being an obligate monomer in functional GC(NO) heterodimers.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Cromatina/metabolismo , Guanilato Ciclasa/metabolismo , Mitosis/efectos de los fármacos , Óxido Nítrico/farmacología , Subunidades de Proteína/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/enzimología , Proliferación Celular/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Guanilato Ciclasa/genética , Microglía/citología , Microglía/efectos de los fármacos , Microglía/enzimología , Unión Proteica/efectos de los fármacos , Subunidades de Proteína/genética , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/genética , Guanilil Ciclasa Soluble , Transfección
10.
Neurochem Res ; 33(12): 2427-35, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18379874

RESUMEN

A large body of evidence supports a role for the NO-cGMP-protein kinase G pathway in the regulation of synaptic transmission and plasticity, brain development and neuroprotection. Circumstantial evidence implicates natriuretic peptide-stimulated cGMP formation in the same CNS functions. In addition to neurons, both cGMP-mediated pathways are functional in glial cells and an increasing number of reports indicate that they may control important aspects of glial cell physiology relevant to neuronal function. In this article we briefly review the regulation of cGMP formation in glial cells and summarize recent evidence indicating that cGMP-mediated pathways can play important roles in astroglial and microglial function in normal and diseased brain.


Asunto(s)
GMP Cíclico/fisiología , Neuroglía/fisiología , Animales , GMP Cíclico/biosíntesis , Humanos , Péptido Natriurético Encefálico/fisiología , Plasticidad Neuronal , Transmisión Sináptica
11.
Mol Cell Neurosci ; 37(3): 494-506, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18083532

RESUMEN

We previously showed that treatment with bacterial lipopolysaccharide (LPS) or pro-inflammatory cytokines decreases NO-sensitive guanylyl cyclase (GC(NO)) activity in astrocytes by decreasing the half-life of the obligate GC(NO) beta1 subunit in a NO-independent but transcription- and translation-dependent process. Here we show that LPS-induced beta1 degradation requires proteasome activity and is independent of NFkappaB activation or beta1 interaction with HSP90. Immunocytochemistry and confocal microscopy analysis revealed that LPS promotes colocalization of the predominantly soluble beta1 protein with ubiquitin and the 20S proteasome in nuclear aggregates that present characteristics of clastosomes, nuclear bodies involved in proteolysis via the ubiquitin-proteasome system. Proteasome and protein synthesis inhibitors prevented LPS-induced clastosome assembly and nuclear colocalization of beta1 with ubiquitin and 20S proteasome, strongly supporting a role for these transient nuclear structures in GC(NO) down-regulation during neuroinflammation.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Regulación hacia Abajo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Guanilato Ciclasa/metabolismo , Lipopolisacáridos/farmacología , Nitritos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/ultraestructura , Células Cultivadas , Cerebelo/citología , GMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Microscopía Electrónica de Transmisión/métodos , Radioinmunoensayo/métodos , Ratas , Ubiquitina/metabolismo
12.
Neurobiol Dis ; 17(3): 462-72, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15571982

RESUMEN

In Alzheimer's disease (AD) brains increased NO synthase (NOS) expression is found in reactive astrocytes surrounding amyloid plaques. We have recently shown that treatment with beta-amyloid peptides or IL-1beta down-regulates NO-sensitive soluble guanylyl cyclase (sGC) in cultured astrocytes and in adult rat brain. In this work, we have examined sGC activity and expression in postmortem brain tissue of AD patients and matched controls. No significant alteration was observed in basal or NO-stimulated sGC activity, nor in sGC beta1 and alpha1 subunit levels in cortical extracts of AD brains. Immunohistochemistry showed intense and widespread labeling of sGC beta1 in cortical and hippocampal neurons and white matter fibrillar astrocytes, while grey matter astrocytes were faintly stained. In AD, expression of sGC in neurons and fibrillar astrocytes is not altered but is markedly reduced in reactive astrocytes surrounding amyloid plaques. Immunostaining for sGC beta1 was also lacking in reactive astrocytes in cortex and subcortical white matter in Creutzfeldt-Jakob disease brains and in subacute and chronic plaques in multiple sclerosis (MS) brains. Thus, induction of astrocyte reactivity is associated with decreased capacity to generate cGMP in response to NO both in vitro and in vivo. This effect may be related to the development of the astroglial inflammatory response.


Asunto(s)
Astrocitos/enzimología , Encéfalo/enzimología , Síndrome de Creutzfeldt-Jakob/enzimología , Guanilato Ciclasa/metabolismo , Esclerosis Múltiple/enzimología , Óxido Nítrico/fisiología , Anciano , Anciano de 80 o más Años , Astrocitos/patología , Encéfalo/patología , Femenino , Humanos , Cinética , Masculino , Persona de Mediana Edad , Cambios Post Mortem
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...