Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 18(9)2017 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-28832497

RESUMEN

During surgery or infection, peripheral inflammation can lead to neuroinflammation, which is associated with cognitive impairment, neurodegeneration, and several neurodegenerative diseases. Dexmedetomidine, an α-2-adrenoceptor agonist, is known to exert anti-inflammatory and neuroprotective properties and reduces the incidence of postoperative cognitive impairments. However, on the whole the molecular mechanisms are poorly understood. This study aims to explore whether dexmedetomidine influences microRNAs (miRNAs) in a rat model of lipopolysaccharide (LPS)-induced neuroinflammation. Adult Wistar rats were injected with 1 mg/kg LPS intraperitoneal (i.p.) in the presence or absence of 5 µg/kg dexmedetomidine. After 6 h, 24 h, and 7 days, gene expressions of interleukin 1-ß (IL1-ß), tumor necrosis factor-α (TNF-α), and microRNA expressions of miR 124, 132, 134, and 155 were measured in the hippocampus, cortex, and plasma. Dexmedetomidine decreased the LPS-induced neuroinflammation in the hippocampus and cortex via significant reduction of the IL1-ß and TNF-α gene expressions after 24 h. Moreover, the LPS-mediated increased expressions of miR 124, 132, 134, and 155 were significantly decreased after dexmedetomidine treatment in both brain regions. In plasma, dexmedetomidine significantly reduced LPS-induced miR 155 after 6 h. Furthermore, there is evidence that miR 132 and 134 may be suitable as potential biomarkers for the detection of neuroinflammation.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Antiinflamatorios/farmacología , Encéfalo/metabolismo , Dexmedetomidina/farmacología , MicroARNs/genética , Animales , Encéfalo/efectos de los fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Masculino , MicroARNs/metabolismo , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
2.
Int J Mol Sci ; 18(1)2017 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-28106777

RESUMEN

Sequelae of prematurity triggered by oxidative stress and free radical-mediated tissue damage have coined the term "oxygen radical disease of prematurity". Caffeine, a potent free radical scavenger and adenosine receptor antagonist, reduces rates of brain damage in preterm infants. In the present study, we investigated the effects of caffeine on oxidative stress markers, anti-oxidative response, inflammation, redox-sensitive transcription factors, apoptosis, and extracellular matrix following the induction of hyperoxia in neonatal rats. The brain of a rat pups at postnatal Day 6 (P6) corresponds to that of a human fetal brain at 28-32 weeks gestation and the neonatal rat is an ideal model in which to investigate effects of oxidative stress and neuroprotection of caffeine on the developing brain. Six-day-old Wistar rats were pre-treated with caffeine and exposed to 80% oxygen for 24 and 48 h. Caffeine reduced oxidative stress marker (heme oxygenase-1, lipid peroxidation, hydrogen peroxide, and glutamate-cysteine ligase catalytic subunit (GCLC)), promoted anti-oxidative response (superoxide dismutase, peroxiredoxin 1, and sulfiredoxin 1), down-regulated pro-inflammatory cytokines, modulated redox-sensitive transcription factor expression (Nrf2/Keap1, and NFκB), reduced pro-apoptotic effectors (poly (ADP-ribose) polymerase-1 (PARP-1), apoptosis inducing factor (AIF), and caspase-3), and diminished extracellular matrix degeneration (matrix metalloproteinases (MMP) 2, and inhibitor of metalloproteinase (TIMP) 1/2). Our study affirms that caffeine is a pleiotropic neuroprotective drug in the developing brain due to its anti-oxidant, anti-inflammatory, and anti-apoptotic properties.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Cafeína/uso terapéutico , Hiperoxia/complicaciones , Hiperoxia/tratamiento farmacológico , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Lesiones Encefálicas/patología , Cafeína/administración & dosificación , Cafeína/farmacología , Citocinas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Metaloproteinasas de la Matriz/metabolismo , FN-kappa B/metabolismo , Fármacos Neuroprotectores/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Peroxirredoxinas/metabolismo , Plasminógeno/metabolismo , Ratas Wistar , Activador de Tejido Plasminógeno/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Brain Behav Immun ; 52: 106-119, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26456693

RESUMEN

Cerebral white matter injury is a leading cause of adverse neurodevelopmental outcome in prematurely born infants involving cognitive deficits in later life. Despite increasing knowledge about the pathophysiology of perinatal brain injury, therapeutic options are limited. In the adult demyelinating disease multiple sclerosis the sphingosine-1-phosphate (S1P) receptor modulating substance fingolimod (FTY720) has beneficial effects. Herein, we evaluated the neuroprotective potential of FTY720 in a neonatal model of oxygen-toxicity, which is associated with hypomyelination and impaired neuro-cognitive outcome. A single dose of FTY720 (1mg/kg) at the onset of neonatal hyperoxia (24h 80% oxygen on postnatal day 6) resulted in improvement of neuro-cognitive development persisting into adulthood. This was associated with reduced microstructural white matter abnormalities 4 months after the insult. In search of the underlying mechanisms potential non-classical (i.e. lymphocyte-independent) pathways were analysed shortly after the insult, comprising modulation of oxidative stress and local inflammatory responses as well as myelination, oligodendrocyte degeneration and maturation. Treatment with FTY720 reduced hyperoxia-induced oxidative stress, microglia activation and associated pro-inflammatory cytokine expression. In vivo and in vitro analyses further revealed that oxygen-induced hypomyelination is restored to control levels, which was accompanied by reduced oligodendrocyte degeneration and enhanced maturation. Furthermore, hyperoxia-induced elevation of S1P receptor 1 (S1P1) protein expression on in vitro cultured oligodendrocyte precursor cells was reduced by activated FTY720 and protection from degeneration is abrogated after selective S1P1 blockade. Finally, FTY720s' classical mode of action (i.e. retention of immune cells within peripheral lymphoid organs) was analysed demonstrating that FTY720 diminished circulating lymphocyte counts independent from hyperoxia. Cerebral immune cell counts remained unchanged by hyperoxia and by FTY720 treatment. Taken together, these results suggest that beneficial effects of FTY720 in neonatal oxygen-induced brain injury may be rather attributed to its anti-oxidative and anti-inflammatory capacity acting in concert with a direct protection of developing oligodendrocytes than to a modulation of peripheral lymphocyte trafficking. Thus, FTY720 might be a potential new therapeutic option for the treatment of neonatal brain injury through reduction of white matter damage.


Asunto(s)
Trastornos del Conocimiento/prevención & control , Clorhidrato de Fingolimod/uso terapéutico , Hiperoxia/tratamiento farmacológico , Sustancia Blanca/efectos de los fármacos , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Imagen de Difusión por Resonancia Magnética , Femenino , Hiperoxia/patología , Lisofosfolípidos/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Fibras Nerviosas Mielínicas/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Oxígeno/administración & dosificación , Embarazo , Distribución Aleatoria , Ratas , Ratas Wistar , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
4.
Exp Lung Res ; 41(8): 466-76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26381719

RESUMEN

AIM OF THE STUDY: Repeated pulmonary lavage allows to reliably reproduce failure of gas exchange and major histological findings of acute lung injury (ALI). However, because the capacity of pulmonary lavage to induce pulmonary inflammation is not well established in rodents, this study aims to characterize the induction of pulmonary inflammation in a rat model of ALI. MATERIALS AND METHODS: Male adult rats were divided into a treatment group (n = 9) that received pulmonary lavage with consecutive mechanical ventilation, and a control group that received mechanical ventilation only (n = 9). Arterial blood gas analyses were performed every 30 min throughout the study. Pressure-volume curves, and lung tissue and plasma samples, were obtained at 240 min after the start of mechanical ventilation. Protein content and surface activity of bronchoalveolar lavage fluid was assessed. Transcriptional and translational regulation of pro- and anti-inflammatory cytokines IL-1ß, TNF-α, IL-6, and IL-10 was determined in lungs and plasma. Markers of cellular stress were measured in lung tissue. RESULTS: Pulmonary lavage significantly decreased lung compliance, induced hypoxia and hypercapnia, and mediated respiratory acidosis. Protein content of lavage fluid was significantly increased and contained washed out surfactant. Expression of IL-1ß, TNF-α, and IL-6 mRNA and protein expression of IL-1ß and TNF-α was significantly induced in lavaged lungs, without spillover into the systemic circulation. Markers of cellular stress were significantly upregulated in lavaged lungs. CONCLUSIONS: This model of ALI applied in rats can induce pulmonary inflammation. The model might be used to develop therapeutic strategies that target pulmonary inflammation in ALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Pulmón/metabolismo , Pulmón/patología , Neumonía/metabolismo , Neumonía/patología , Animales , Lavado Broncoalveolar/métodos , Líquido del Lavado Bronquioalveolar/química , Modelos Animales de Enfermedad , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Surfactantes Pulmonares/metabolismo , Ratas , Ratas Sprague-Dawley , Respiración Artificial/métodos , Factor de Necrosis Tumoral alfa/metabolismo
5.
Int J Med Sci ; 12(12): 995-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26664262

RESUMEN

Uncoupling proteins (UCPs) are anion channels that can decouple the mitochondrial respiratory chain. "Mild uncoupling" of internal respiration reduces free radical production and oxidative cell stress. Chronic alcohol consumption is a potent inducer of oxidative stress in multiple tissues and regulates UCP-2 and -4 expression in the brain. To analyse the impact of chronic alcohol intake on UCP-2 expression in tissues with high endogenous UCP-2 contents, male Wistar rats (n=34) were treated with a 12-week 5% alcohol diet. In the lungs and the spleen of rats with a chronic alcohol diet cytochrome c release from mitochondria was significantly increased. Both organs did not show any altered gene and protein expression of UCP-2. Different to cerebral tissue chronic alcohol consumption has no regulatory effect on UCP-2 gene and protein expression in organs with a high endogenous UCP-2 content. Therefore, chronic alcohol consumption leads to a tissue specific expression of UCP-2.


Asunto(s)
Alcoholismo/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Adenosina Trifosfato/metabolismo , Alcoholismo/genética , Animales , Encéfalo/metabolismo , Citocromos c/metabolismo , Expresión Génica , Canales Iónicos/genética , Pulmón/metabolismo , Masculino , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Estrés Oxidativo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Bazo/metabolismo , Distribución Tisular , Proteína Desacopladora 2
6.
Cereb Cortex ; 23(9): 2245-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22806269

RESUMEN

Homozygous mutations in the cyclin-dependent kinase-5 regulatory subunit-associated protein 2 gene CDK5RAP2 cause primary autosomal recessive microcephaly (MCPH). MCPH is characterized by a pronounced reduction of brain volume, particularly of the cerebral cortex, and mental retardation. Though it is a rare developmental disorder, MCPH has moved into the spotlight of neuroscience because of its proposed central role in stem-cell biology and brain development. Investigation of the neural basis of genetically defined MCPH has been limited to animal studies and neuroimaging of affected patients as no neuropathological studies have been published. In the present study, we depict the spatiotemporal expression of CDK5RAP2 in the developing brain of mouse and human. We found intriguing concordance between regions of high CDK5RAP2 expression in the mouse and sites of pathology suggested by neuroimaging studies in humans and mouse. Our findings in human tissue confirm those in mouse tissues, underlining the function of CDK5RAP2 in cell proliferation and arguing for a conserved role of this protein in the development of the mammalian cerebral cortex.


Asunto(s)
Encéfalo/embriología , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Animales , Feto/metabolismo , Humanos , Ratones , Microcefalia/embriología , Microcefalia/metabolismo
7.
Int J Mol Sci ; 15(3): 3784-98, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24595240

RESUMEN

Exposure to N-methyl-d-aspartate (NMDA) receptor antagonists has been demonstrated to induce neurodegeneration in newborn rats. However, in clinical practice the use of NMDA receptor antagonists as anesthetics and sedatives cannot always be avoided. The present study investigated the effect of the indirect cholinergic agonist physostigmine on neurotrophin expression and the extracellular matrix during NMDA receptor antagonist induced injury to the immature rat brain. The aim was to investigate matrix metalloproteinase (MMP)-2 activity, as well as expression of tissue inhibitor of metalloproteinase (TIMP)-2 and brain-derived neurotrophic factor (BDNF) after co-administration of the non-competitive NMDA receptor antagonist MK801 (dizocilpine) and the acetylcholinesterase (AChE) inhibitor physostigmine. The AChE inhibitor physostigmine ameliorated the MK801-induced reduction of BDNF mRNA and protein levels, reduced MK801-triggered MMP-2 activity and prevented decreased TIMP-2 mRNA expression. Our results indicate that AChE inhibition may prevent newborn rats from MK801-mediated brain damage by enhancing neurotrophin-associated signaling pathways and by modulating the extracellular matrix.


Asunto(s)
Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Acetilcolinesterasa/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Maleato de Dizocilpina/farmacología , Expresión Génica/efectos de los fármacos , Immunoblotting , Metaloproteinasa 2 de la Matriz/metabolismo , Fisostigmina/farmacología , Ratas Wistar , Receptores de N-Metil-D-Aspartato/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
8.
Dev Neurosci ; 35(2-3): 255-64, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23445753

RESUMEN

The cholinergic anti-inflammatory pathway is a neural mechanism that suppresses the innate inflammatory response and controls inflammation employing acetylcholine as the key endogenous mediator. In this study, we investigated the effects of the cholinergic agonists, physostigmine and donepezil, on neurodegeneration, inflammation and oxidative stress during oxygen toxicity in the developing rat brain. The aim of this study was to investigate the level of neurodegeneration, expression of proinflammatory cytokines, glutathione and lipid peroxidation after hyperoxia and treatment with the acetylcholinesterase (AChE) inhibitors, physostigmine and donepezil in the brain of neonatal rats. Six-day-old Wistar rats were exposed to 80% oxygen for 12-24 h and received 100 µg/kg physostigmine or 200 µg/kg donepezil intraperitoneally. Sex-matched littermates kept in room air and injected with normal saline, physostigmine or donepezil served as controls. Treatment with both inhibitors significantly reduced hyperoxia-triggered activity of AChE, neural cell death and the upregulation of the proinflammatory cytokines IL-1ß and TNF-α in the immature rat brain on the mRNA and protein level. In parallel, hyperoxia-induced oxidative stress was reduced by concomitant physostigmine and donepezil administration, as shown by an increased reduced/oxidized glutathione ratio and attenuated malondialdehyde levels, as a sign of lipid peroxidation. Our results suggest that a single treatment with AChE inhibitors at the beginning of hyperoxia attenuated the detrimental effects of oxygen toxicity in the developing brain and may pave the way for AChE inhibitors, which are currently used for the treatment of Alzheimer's disease, as potential candidates for adjunctive neuroprotective therapies to the immature brain.


Asunto(s)
Encéfalo/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Hiperoxia/prevención & control , Estrés Oxidativo/efectos de los fármacos , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Modelos Animales de Enfermedad , Donepezilo , Femenino , Hiperoxia/patología , Inmunohistoquímica , Indanos/farmacología , Masculino , Degeneración Nerviosa/patología , Degeneración Nerviosa/prevención & control , Oxígeno/toxicidad , Fisostigmina/farmacología , Piperidinas/farmacología , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa
9.
Eur Respir J ; 41(4): 966-73, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22878872

RESUMEN

In preterm human infants, briefly elevated concentrations of oxygen are associated with a prolonged increase in blood chemokine concentrations and the development of bronchopulmonary dysplasia (BPD). Caffeine given to preterm infants for the prevention or treatment of apnoea has been shown to reduce the rate of BPD. We tested the hypotheses that infant rats exposed to a combination of caffeine and hyperoxia would be less susceptible to lung injury than those exposed to hyperoxia alone and that caffeine decreases the pulmonary tissue expression of chemokines and leukocyte influx following hyperoxia. Using 6-day-old rat pups, we demonstrated that 24 h of 80% oxygen exposure caused pulmonary recruitment of neutrophils and macrophages. High levels of oxygen upregulated the expression of: the CXC chemokines, cytokine-induced neutrophil chemoattractant-1 and macrophage inflammatory protein-2; the CC-chemokine monocyte chemoattractant protein-1; the pro-inflammatory cytokines tumour necrosis factor-α and interleukin-6, as measured by realtime PCR after the administration of caffeine (10 mg · kg(-1) body weight); and attenuated chemokine and cytokine upregulation, as well as the influx of CD11b(+), ED-1(+) and myeloperoxidase(+) leukocytes. These experiments suggest that protective effects of caffeine in the neonatal lung are mediated, at least in part, by reduction of pulmonary inflammation.


Asunto(s)
Cafeína/farmacología , Hiperoxia/patología , Neumonía/patología , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/patología , Estimulantes del Sistema Nervioso Central/farmacología , Quimiocinas CXC/metabolismo , Citocinas/metabolismo , Humanos , Recién Nacido , Leucocitos/citología , Pulmón/patología , Oxígeno/metabolismo , Neumonía/metabolismo , Alveolos Pulmonares/metabolismo , Ratas , Factores de Tiempo , Xantinas/farmacología
10.
Alcohol Clin Exp Res ; 37(10): 1650-6, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23800309

RESUMEN

BACKGROUND: Chronic alcohol consumption leads to oxidative stress in a variety of cells, especially in brain cells because they have a reduced oxidative metabolism of alcohol. Uncoupling proteins (UCPs) are anion channels of the inner mitochondrial membrane, which can decouple internal respiration. "Mild uncoupling" of the mitochondrial respiratory chain leads to a reduced production of free radicals (reactive oxygen species) and a reduction in oxidative cell stress. The extent to which chronic alcohol consumption regulates UCP-2 and -4 in the brain is still unknown. METHODS: We examined the effects of a 12-week 5% alcohol diet in the brain of male Wistar rats (n = 34). Cerebral gene and protein expression of UCP-2, -4, as well as Bcl-2, and the release of cytochrome c out of the mitochondria were detected by real-time polymerase chain reaction and Western blot analysis. The percentage of degenerated cells was determined by Fluoro-Jade B staining of brain slices. RESULTS: Brains of rats with a chronic alcohol diet showed an increased gene and protein expression of UCP-2 and -4. The expression of the antiapoptotic protein Bcl-2 in the brain of the alcohol-treated animals was decreased significantly, whereas cytochrome c release from mitochondria was increased. In addition increased neurodegeneration could be demonstrated in the alcohol-treated animals. CONCLUSIONS: Chronic alcohol consumption leads to a cerebral induction of UCP-2 and -4 with a simultaneous decrease in the antiapoptotic protein Bcl-2, cytochrome c release from mitochondria and increased neurodegeneration. This study reveals a compensatory effect of UCP-2 and -4 in the brain during chronic alcohol consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Encéfalo/metabolismo , Etanol/administración & dosificación , Regulación de la Expresión Génica , Canales Iónicos/biosíntesis , Proteínas Mitocondriales/biosíntesis , Animales , Encéfalo/efectos de los fármacos , Masculino , Proteínas Desacopladoras Mitocondriales , Ratas , Ratas Wistar , Proteína Desacopladora 2
11.
Nat Neurosci ; 11(4): 476-87, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18344994

RESUMEN

Intrinsic antioxidant defenses are important for neuronal longevity. We found that in rat neurons, synaptic activity, acting via NMDA receptor (NMDAR) signaling, boosted antioxidant defenses by making changes to the thioredoxin-peroxiredoxin (Prx) system. Synaptic activity enhanced thioredoxin activity, facilitated the reduction of overoxidized Prxs and promoted resistance to oxidative stress. Resistance was mediated by coordinated transcriptional changes; synaptic NMDAR activity inactivated a previously unknown Forkhead box O target gene, the thioredoxin inhibitor Txnip. Conversely, NMDAR blockade upregulated Txnip in vivo and in vitro, where it bound thioredoxin and promoted vulnerability to oxidative damage. Synaptic activity also upregulated the Prx reactivating genes Sesn2 (sestrin 2) and Srxn1 (sulfiredoxin), via C/EBPbeta and AP-1, respectively. Mimicking these expression changes was sufficient to strengthen antioxidant defenses. Trans-synaptic stimulation of synaptic NMDARs was crucial for boosting antioxidant defenses; chronic bath activation of all (synaptic and extrasynaptic) NMDARs induced no antioxidative effects. Thus, synaptic NMDAR activity may influence the progression of pathological processes associated with oxidative damage.


Asunto(s)
Antioxidantes/metabolismo , Estrés Oxidativo/fisiología , Peroxirredoxinas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Tiorredoxinas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Regulación de la Expresión Génica/fisiología , Ratones , Neuronas/metabolismo , Proteínas Nucleares , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Peroxidasas , Proteínas/metabolismo , Ratas , Transducción de Señal/fisiología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Transcripción Genética/fisiología
12.
Int J Mol Sci ; 13(10): 12939-51, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23202931

RESUMEN

Autophagy is a self-degradative process that involves turnover and recycling of cytoplasmic components in healthy and diseased tissue. Autophagy has been shown to be protective at the early stages of programmed cell death but it can also promote apoptosis under certain conditions. Earlier we demonstrated that oxygen contributes to the pathogenesis of neonatal brain damage, which can be ameliorated by intervention with recombinant human erythropoietin (rhEpo). Extrinsic- and intrinsic apoptotic pathways are involved in oxygen induced neurotoxicity but the role of autophagy in this model is unclear. We analyzed the expression of autophagy activity markers in the immature rodent brain after exposure to elevated oxygen concentrations. We observed a hyperoxia-exposure dependent regulation of autophagy-related gene (Atg) proteins Atg3, 5, 12, Beclin-1, microtubule-associated protein 1 light chain 3 (LC3), LC3A-II, and LC3B-II which are all key autophagy activity proteins. Interestingly, a single injection with rhEpo at the onset of hyperoxia counteracted these oxygen-mediated effects. Our results indicate that rhEpo generates its protective effect by modifying the key autophagy activity proteins.


Asunto(s)
Autofagia/efectos de los fármacos , Encéfalo/metabolismo , Eritropoyetina/farmacología , Hipoxia , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Beclina-1 , Encéfalo/efectos de los fármacos , Epoetina alfa , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Animales , Péptido Sintasas/genética , Péptido Sintasas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos
13.
J Neuroimmune Pharmacol ; 17(1-2): 261-276, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34357471

RESUMEN

Infections and perioperative stress can lead to neuroinflammation, which in turn is linked to cognitive impairments such as postoperative delirium or postoperative cognitive dysfunctions. The α2-adrenoceptor agonist dexmedetomidine (DEX) prevents cognitive impairments and has organo-protective and anti-inflammatory properties. Macroautophagy (autophagy) regulates many biological processes, but its role in DEX-mediated anti-inflammation and the underlying mechanism of DEX remains largely unclear. We were interested how a pretreatment with DEX protects against lipopolysaccharide (LPS)-induced inflammation in adult male Wistar rats. We used Western blot and activity assays to study how DEX modulated autophagy- and apoptosis-associated proteins as well as molecules of the cholinergic anti-inflammatory pathway, and qPCR to analyse the expression of autophagy and inflammation-associated microRNAs (miRNA) in the spleen, cortex and hippocampus at different time points (6 h, 24 h, 7 d). We showed that a DEX pretreatment prevents LPS-induced impairments in autophagic flux and attenuates the LPS-induced increase in the apoptosis-associated protein cleaved poly(ADP-ribose)-polymerase (PARP) in the spleen. Both, DEX and LPS altered miRNA expression and molecules of the cholinergic anti-inflammatory pathway in the spleen and brain. While only a certain set of miRNAs was up- and/or downregulated by LPS in each tissue, which was prevented or attenuated by a DEX pretreatment in the spleen and hippocampus, all miRNAs were up- and/or downregulated by DEX itself - independent of whether or not they were altered by LPS. Our results indicate that the organo-protective effect of DEX may be mediated by autophagy, possibly by acting on associated miRNAs, and the cholinergic anti-inflammatory pathway. Preventive effects of DEX on LPS-induced inflammation. DEX restores the LPS-induced impairments in autophagic flux, attenuates PARP cleavage and alters molecules of the cholinergic system in the spleen. Furthermore, DEX alters and prevents LPS-induced miRNA expression changes in the spleen and brain along with LPS.


Asunto(s)
MicroARNs , Neuroinmunomodulación , Masculino , Animales , Ratas , Lipopolisacáridos/toxicidad , Ratas Wistar , Autofagia
14.
Apoptosis ; 16(6): 636-51, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21437721

RESUMEN

Apoptosis is modulated by extrinsic and intrinsic signaling pathways through the formation of the death receptor-mediated death-inducing signaling complex (DISC) and the mitochondrial-derived apoptosome, respectively. Ino-C2-PAF, a novel synthetic phospholipid shows impressive antiproliferative and apoptosis-inducing activity. Little is known about the signaling pathway through which it stimulates apoptosis. Here, we show that this drug induces apoptosis through proteins of the death receptor pathway, which leads to an activation of the intrinsic apoptotic pathway. Apoptosis induced by Ino-C2-PAF and its glucosidated derivate, Glc-PAF, was dependent on the DISC components FADD and caspase-8. This can be inhibited in FADD--/-- and caspase-8--/-- cells, in which the breakdown of the mitochondrial membrane potential, release of cytochrome c and activation of caspase-9, -8 and -3 do not occur. In addition, the overexpression of crmA, c-Flip or dominant negative FADD as well as treatment with the caspase-8 inhibitor z-IETD-fmk protected against Ino-C2-PAF-induced apoptosis. Apoptosis proceeds in the absence of CD95/Fas-ligand expression and is independent of blockade of a putative death-ligand/receptor interaction. Furthermore, apoptosis cannot be inhibited in CD95/Fas--/-- Jurkat cells. Expression of Bcl-2 in either the mitochondria or the endoplasmic reticulum (ER) strongly inhibited Ino-C2-PAF- and Glc-PAF-induced apoptosis. In conclusion, Ino-C2-PAF and Glc-PAF trigger a CD95/Fas ligand- and receptor-independent atypical DISC that relies on the intrinsic apoptotic pathway via the ER and the mitochondria.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Mitocondrias/metabolismo , Neoplasias/fisiopatología , Fosfolípidos/farmacología , Transducción de Señal , Caspasa 8/genética , Activación Enzimática/efectos de los fármacos , Proteína de Dominio de Muerte Asociada a Fas/genética , Glicosilación , Humanos , Células Jurkat , Mitocondrias/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Fosfolípidos/síntesis química , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
15.
Alcohol Clin Exp Res ; 35(8): 1381-91, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21410490

RESUMEN

BACKGROUND: Alcohol induces cellular stress and promotes cell death in immune cells. Molecular mechanisms by which ethanol impairs the function of immune cells are largely unknown. Autophagy is a degradation pathway, acting either as a pro-survival or pro-death mechanism activated during stress conditions. We examined whether ethanol influences autophagy in monocytic human U937, CD4 Jurkat, and MCF-7 cells. METHODS: Effects of ethanol during starvation-induced autophagy were investigated, treating cells with ethanol alone and in combination with activation of autophagy by rapamycin or inhibition by wortmannin. Apoptotic and necrotic cell death features such as the breakdown of the mitochondrial membrane potential, DNA fragmentation, and cell permeability were assessed using FACS analyses. Expression level of Beclin-1, LC3-II, Bcl-2, and the activation of caspase-3, and PARP-1 were determined using Western blot analyses. Influence of ethanol on formation of LC3-II complexes was assessed using fluorescence microscopy in MCF-7 cells stable transfected with a GFP-LC3-II-expression vector. RESULTS: Ethanol down regulated autophagy proteins such as Beclin-1 and LC3-II. Apoptosis was enhanced as shown by breakdown of mitochondrial potential, up-regulation of cleaved caspase-3 and PARP-1 and down-regulation of anti-apoptotic protein Bcl-2. Formation of LC3-II complexes was inhibited by ethanol in caspase-3 deficient MCF-7 cells. Stimulation of autophagy by rapamycin prevented ethanol-induced apoptotic cell death. Inhibition of autophagy by wortmannin aggravated ethanol-mediated necrotic cell death. CONCLUSION: Inhibition of autophagy via ethanol enhances susceptibility to cell death.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular , Línea Celular Tumoral , Depresores del Sistema Nervioso Central/farmacología , Regulación hacia Abajo , Etanol/farmacología , Humanos , Mitocondrias/efectos de los fármacos , Necrosis/metabolismo , Células U937 , Regulación hacia Arriba/efectos de los fármacos
16.
J Perinat Med ; 39(1): 83-8, 2011 01.
Artículo en Inglés | MEDLINE | ID: mdl-20954855

RESUMEN

Perinatal brain damage may result in impaired neurological development in extremely preterm infants. The underlying pathophysiological mechanisms are complex, and biomarkers of prognostic value are not available. The aim of this study was to analyze soluble Fas (sFas) concentrations in the cerebrospinal fluid (CSF) representative for involvement of apoptotic processes in preterm infants developing posthemorrhagic hydrocephalus (PHHC) and to link them to white matter damage (WMD) diagnosed by cranial ultrasound. A total of 29 preterm infants with PHHC were included in the study; 17 of them had signs of cystic WMD (cWMD) on ultrasound examinations. CSF samples were obtained at first ventriculostomy, and results were compared to those of a reference group of 24 preterm and term infants without neurologic diseases. sFas concentrations were elevated in CSF samples of PHHC patients compared to the reference group. In patients with cWMD, sFas concentrations were significantly higher than in patients without cWMD. These results indicate that apoptosis via the Fas pathway is involved in the pathogenesis of cWMD in the context of PHHC, and that sFas in the CSF may serve as a marker of cWMD development.


Asunto(s)
Daño Encefálico Crónico/líquido cefalorraquídeo , Hidrocefalia/líquido cefalorraquídeo , Enfermedades del Prematuro/líquido cefalorraquídeo , Hemorragias Intracraneales/complicaciones , Receptor fas/líquido cefalorraquídeo , Apoptosis , Biomarcadores/líquido cefalorraquídeo , Daño Encefálico Crónico/diagnóstico por imagen , Daño Encefálico Crónico/etiología , Humanos , Hidrocefalia/complicaciones , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/diagnóstico por imagen , Enfermedades del Prematuro/etiología , Estudios Prospectivos , Ultrasonografía
17.
Brain Behav Immun ; 24(5): 792-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19729061

RESUMEN

Oxygen toxicity contributes to the pathogenesis of adverse neurological outcome in survivors of preterm birth in clinical studies. In infant rodent brains, hyperoxia triggers widespread apoptotic neurodegeneration, induces pro-inflammatory cytokines and inhibits growth factor signaling cascades. Since a tissue-protective effect has been observed for recombinant erythropoietin (rEpo), we hypothesized that rEpo would influence hyperoxia-induced oxidative stress in the developing rat brain. The aim of this study was to investigate the level of glutathione (reduced and oxidized), lipid peroxidation and the expression of heme oxygenase-1 (HO-1) and acetylcholinesterase (AChE) after hyperoxia and rEpo treatment. Six-day-old Wistar rats were exposed to 80% oxygen for 2-48 h and received 20,000 IU/kg rEpo intraperitoneally (i.p.). Sex-matched littermates kept under room air and injected with normal saline or rEpo served as controls. Treatment with rEpo significantly reduced hyperoxia-induced upregulation of oxidized glutathione (GSSG) and malondialdehyde, a product of lipid breakdown, whereas reduced glutathione (GSH) was upregulated by rEpo. In parallel, hyperoxia-treated immature rat brains revealed rEpo-suppressible upregulation of synaptic AChE-S as well as of the stress-inducible AChE-R variant, together predicting rEpo-protected cholinergic signaling and restrained inflammatory reactions. Furthermore, treatment with rEpo induced upregulation of HO-1 on mRNA, protein and activity level in the developing rat brain. Our results suggest that rEpo generates its protective effect against oxygen toxicity by a reduction of diverse oxidative stress parameters and by limiting the stressor-inducible changes in both HO-1 and cholinergic functions.


Asunto(s)
Encéfalo/patología , Eritropoyetina/farmacología , Hiperoxia/patología , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Eritropoyetina/uso terapéutico , Femenino , Glutatión/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hiperoxia/tratamiento farmacológico , Hiperoxia/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
18.
Mol Cell Proteomics ; 7(12): 2293-310, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18587059

RESUMEN

Neurotransmitter signaling is essential for physiologic brain development. Sedative and anticonvulsant agents that reduce neuronal excitability via antagonism at N-methyl-D-aspartate receptors (NMDARs) and/or agonism at gamma-aminobutyric acid subtype A receptors (GABA(A)Rs) are applied frequently in obstetric and pediatric medicine. We demonstrated that a 1-day treatment of infant mice at postnatal day 6 (P6) with the NMDAR antagonist dizocilpine or the GABA(A)R agonist phenobarbital not only has acute but also long term effects on the cerebral cortex. Changes of the cerebral cortex proteome 1 day (P7), 1 week (P14), and 4 weeks (P35) following treatment at P6 suggest that a suppression of synaptic neurotransmission during brain development dysregulates proteins associated with apoptosis, oxidative stress, inflammation, cell proliferation, and neuronal circuit formation. These effects appear to be age-dependent as most protein changes did not occur in mice subjected to such pharmacological treatment in adulthood. Previously performed histological evaluations of the brains revealed widespread apoptosis and decreased cell proliferation following such a drug treatment in infancy and are thus consistent with brain protein changes reported in this study. Our results point toward several pathways modulated by a reduction of neuronal excitability that might interfere with critical developmental events and thus affirm concerns about the impact of NMDAR- and/or GABA(A)R-modulating drugs on human brain development.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Receptores de GABA-A/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica/fisiología , Animales , Animales Recién Nacidos , Movimiento Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Maleato de Dizocilpina/farmacología , Electroforesis en Gel Bidimensional , Agonistas de Receptores de GABA-A , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/efectos de los fármacos , Fenobarbital/farmacología , Proteoma/química , Proteoma/metabolismo , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Transmisión Sináptica/efectos de los fármacos , Factores de Tiempo
19.
Dev Neurosci ; 31(5): 394-402, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19672068

RESUMEN

Oxygen toxicity appears to contribute to the pathogenesis of adverse neurological outcome in survivors of preterm birth. In infant rodent brains, hyperoxia triggers widespread apoptotic neurodegeneration, induces proinflammatory cytokines and inhibits growth factor signaling cascades. Since a tissue-protective effect has been observed for recombinant erythropoietin (rEpo), we hypothesized that rEpo would influence the expression of proinflammatory cytokines and matrix metalloproteinase (MMP)-2 and MMP-9. Six-day-old Wistar rats were exposed to 80% oxygen for 2-48 h and received 20,000 IU rEpo i.p. Sex-matched littermates kept in room air and injected with normal saline or rEpo served as controls. Treatment with rEpo significantly reduced hyperoxia-induced upregulation of the proinflammatory cytokines IL-1beta and IL-18 in infant rodent brains on the mRNA and protein levels. In parallel, gelatin zymography in hyperoxia-treated immature rat brains revealed an upregulation of active MMP-2 which was reduced by concomitant rEpo treatment. Furthermore, hyperoxia induced upregulation of MMP-9 following 12 h of oxygen exposure and this was attenuated by rEpo treatment. Our results suggest that rEpo generates its protective effect against oxygen toxicity through a reduction of proinflammatory mediator levels.


Asunto(s)
Encéfalo/patología , Muerte Celular/efectos de los fármacos , Eritropoyetina/uso terapéutico , Hiperoxia/tratamiento farmacológico , Hiperoxia/patología , Análisis de Varianza , Animales , Animales Recién Nacidos , Western Blotting , Encéfalo/efectos de los fármacos , Interleucina-18/biosíntesis , Interleucina-1beta/biosíntesis , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Oxígeno/toxicidad , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Histochem Cell Biol ; 132(4): 435-45, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19526364

RESUMEN

Emerging evidence suggests a role for glutamate and its receptors in the biology of cancer. This study was designed to systematically analyze the expression of ionotropic and metabotropic glutamate receptor subunits in various human cancer cell lines, compare expression levels to those in human brain tissue and, using electrophysiological techniques, explore whether cancer cells respond to glutamate receptor agonists and antagonists. Expression analysis of glutamate receptor subunits NR1-NR3B, GluR1-GluR7, KA1, KA2 and mGluR1-mGluR8 was performed by means of RT-PCR in human rhabdomyosarcoma/medulloblastoma (TE671), neuroblastoma (SK-NA-S), thyroid carcinoma (FTC 238), lung carcinoma (SK-LU-1), astrocytoma (MOGGCCM), multiple myeloma (RPMI 8226), glioma (U87-MG and U343), lung carcinoma (A549), colon adenocarcinoma (HT 29), T cell leukemia cells (Jurkat E6.1), breast carcinoma (T47D) and colon adenocarcinoma (LS180). Analysis revealed that all glutamate receptor subunits were differentially expressed in the tumor cell lines. For the majority of tumors, expression levels of NR2B, GluR4, GluR6 and KA2 were lower compared to human brain tissue. Confocal imaging revealed that selected glutamate receptor subunit proteins were expressed in tumor cells. By means of patch-clamp analysis, it was shown that A549 and TE671 cells depolarized in response to application of glutamate agonists and that this effect was reversed by glutamate receptor antagonists. This study reveals that glutamate receptor subunits are differentially expressed in human tumor cell lines at the mRNA and the protein level, and that their expression is associated with the formation of functional channels. The potential role of glutamate receptor antagonists in cancer therapy is a feasible goal to be explored in clinical trials.


Asunto(s)
Neoplasias/metabolismo , Receptores AMPA/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Encéfalo/metabolismo , Línea Celular Tumoral , Antagonistas de Aminoácidos Excitadores/farmacología , Humanos , Técnicas de Placa-Clamp , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores AMPA/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Valina/análogos & derivados , Valina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA