Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(7)2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-37047246

RESUMEN

Neurons have long been central to the study of cellular networks in the nervous system [...].


Asunto(s)
Enfermedades del Sistema Nervioso , Neuroglía , Humanos , Neuroglía/fisiología , Neuronas/fisiología
2.
Int J Mol Sci ; 24(8)2023 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-37108406

RESUMEN

Maternal immune activation (MIA) is an important risk factor for neurodevelopmental disorders such as autism. The aim of the current study was to investigate the development-dependent changes in the mitochondrial function of MIA-exposed offspring, which may contribute to autism-like deficits. MIA was evoked by the single intraperitoneal administration of lipopolysaccharide to pregnant rats at gestation day 9.5, and several aspects of mitochondrial function in fetuses and in the brains of seven-day-old pups and adolescent offspring were analyzed along with oxidative stress parameters measurement. It was found that MIA significantly increased the activity of NADPH oxidase (NOX), an enzyme generating reactive oxygen species (ROS) in the fetuses and in the brain of seven-day-old pups, but not in the adolescent offspring. Although a lower mitochondrial membrane potential accompanied by a decreased ATP level was already observed in the fetuses and in the brain of seven-day-old pups, persistent alterations of ROS, mitochondrial membrane depolarization, and lower ATP generation with concomitant electron transport chain complexes downregulation were observed only in the adolescent offspring. We suggest that ROS observed in infancy are most likely of a NOX activity origin, whereas in adolescence, ROS are produced by damaged mitochondria. The accumulation of dysfunctional mitochondria leads to the intense release of free radicals that trigger oxidative stress and neuroinflammation, resulting in an interlinked vicious cascade.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Femenino , Ratas , Animales , Especies Reactivas de Oxígeno , Encéfalo , Vitaminas , Mitocondrias , Adenosina Trifosfato , Conducta Animal/fisiología , Modelos Animales de Enfermedad
3.
Int J Mol Sci ; 24(7)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37047292

RESUMEN

Neurotrophic factors (NTFs) play an important role in maintaining homeostasis of the central nervous system (CNS) by regulating the survival, differentiation, maturation, and development of neurons and by participating in the regeneration of damaged tissues. Disturbances in the level and functioning of NTFs can lead to many diseases of the nervous system, including degenerative diseases, mental diseases, and neurodevelopmental disorders. Each CNS disease is characterized by a unique pathomechanism, however, the involvement of certain processes in its etiology is common, such as neuroinflammation, dysregulation of NTFs levels, or mitochondrial dysfunction. It has been shown that NTFs can control the activation of glial cells by directing them toward a neuroprotective and anti-inflammatory phenotype and activating signaling pathways responsible for neuronal survival. In this review, our goal is to outline the current state of knowledge about the processes affected by NTFs, the crosstalk between NTFs, mitochondria, and the nervous and immune systems, leading to the inhibition of neuroinflammation and oxidative stress, and thus the inhibition of the development and progression of CNS disorders.


Asunto(s)
Encefalopatías , Enfermedades del Sistema Nervioso Central , Humanos , Enfermedades Neuroinflamatorias , Factores de Crecimiento Nervioso/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Encefalopatías/metabolismo , Enfermedades del Sistema Nervioso Central/etiología , Enfermedades del Sistema Nervioso Central/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
4.
Int J Mol Sci ; 24(8)2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-37108467

RESUMEN

Tuberous sclerosis complex (TSC) is a rare genetic multisystem disorder caused by loss-of-function mutations in the tumour suppressors TSC1/TSC2, both of which are negative regulators of the mammalian target of rapamycin (mTOR) kinase. Importantly, mTOR hyperactivity seems to be linked with the pathobiology of autism spectrum disorders (ASD). Recent studies suggest the potential involvement of microtubule (MT) network dysfunction in the neuropathology of "mTORopathies", including ASD. Cytoskeletal reorganization could be responsible for neuroplasticity disturbances in ASD individuals. Thus, the aim of this work was to study the effect of Tsc2 haploinsufficiency on the cytoskeletal pathology and disturbances in the proteostasis of the key cytoskeletal proteins in the brain of a TSC mouse model of ASD. Western-blot analysis indicated significant brain-structure-dependent abnormalities in the microtubule-associated protein Tau (MAP-Tau), and reduced MAP1B and neurofilament light (NF-L) protein level in 2-month-old male B6;129S4-Tsc2tm1Djk/J mice. Alongside, pathological irregularities in the ultrastructure of both MT and neurofilament (NFL) networks as well as swelling of the nerve endings were demonstrated. These changes in the level of key cytoskeletal proteins in the brain of the autistic-like TSC mice suggest the possible molecular mechanisms responsible for neuroplasticity alterations in the ASD brain.


Asunto(s)
Trastorno del Espectro Autista , Esclerosis Tuberosa , Ratones , Animales , Masculino , Trastorno del Espectro Autista/genética , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas del Citoesqueleto/genética , Microtúbulos/metabolismo , Mamíferos/metabolismo
5.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768946

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disease that is characterized by a deficit in social interactions and communication, as well as repetitive and restrictive behaviors. Increasing lines of evidence suggest an important role for immune dysregulation and/or inflammation in the development of ASD. Recently, a relationship between inflammation, oxidative stress, and mitochondrial dysfunction has been reported in the brain tissue of individuals with ASD. Some recent studies have also reported oxidative stress and mitochondrial abnormalities in animal models of maternal immune activation (MIA). This review is focused on the hypothesis that MIA induces microglial activation, oxidative stress, and mitochondrial dysfunction, a deleterious trio in the brain that can lead to neuroinflammation and neurodevelopmental pathologies in offspring. Infection during pregnancy activates the mother's immune system to release proinflammatory cytokines, such as IL-6, TNF-α, and others. Furthermore, these cytokines can directly cross the placenta and enter the fetal circulation, or activate resident immune cells, resulting in an increased production of proinflammatory cytokines, including IL-6. Proinflammatory cytokines that cross the blood-brain barrier (BBB) may initiate a neuroinflammation cascade, starting with the activation of the microglia. Inflammatory processes induce oxidative stress and mitochondrial dysfunction that, in turn, may exacerbate oxidative stress in a self-perpetuating vicious cycle that can lead to downstream abnormalities in brain development and behavior.


Asunto(s)
Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Animales , Trastorno del Espectro Autista/terapia , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Mediadores de Inflamación/inmunología , Intercambio Materno-Fetal/inmunología , Microglía/inmunología , Mitocondrias/inmunología , Modelos Inmunológicos , Neuroinmunomodulación , Estrés Oxidativo/inmunología , Embarazo
6.
Int J Mol Sci ; 22(6)2021 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-33809910

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental condition characterized by deficient social interaction and communication besides repetitive, stereotyped behaviours. A characteristic feature of ASD is altered dendritic spine density and morphology associated with synaptic plasticity disturbances. Since microtubules (MTs) regulate dendritic spine morphology and play an important role in spine development and plasticity the aim of the present study was to investigate the alterations in the content of neuronal α/ß-tubulin and Tau protein level as well as phosphorylation state in the valproic acid (VPA)-induced rat model of autism. Our results indicated that maternal exposure to VPA induces: (1) decrease the level of α/ß-tubulin along with Tau accumulation in the hippocampus and cerebral cortex; (2) excessive Tau phosphorylation and activation of Tau-kinases: CDK5, ERK1/2, and p70S6K in the cerebral cortex; (3) up-regulation of mTOR kinase-dependent signalling in the hippocampus and cerebral cortex of adolescent rat offspring. Moreover, immunohistochemical staining showed histopathological changes in neurons (chromatolysis) in both analysed brain structures of rats prenatally exposed to VPA. The observed changes in Tau protein together with an excessive decrease in α/ß-tubulin level may suggest destabilization and thus dysfunction of the MT cytoskeleton network, which in consequence may lead to the disturbance in synaptic plasticity and the development of autistic-like behaviours.


Asunto(s)
Trastorno Autístico/etiología , Trastorno Autístico/metabolismo , Encéfalo/metabolismo , Exposición Materna/efectos adversos , Efectos Tardíos de la Exposición Prenatal , Ácido Valproico/efectos adversos , Proteínas tau/metabolismo , Animales , Trastorno Autístico/patología , Biomarcadores , Encéfalo/patología , Susceptibilidad a Enfermedades , Activación Enzimática , Femenino , Inmunohistoquímica , Fosforilación , Embarazo , Ratas , Transducción de Señal , Tubulina (Proteína)/metabolismo
7.
Int J Mol Sci ; 22(18)2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34576223

RESUMEN

Tuberous sclerosis complex (TSC) is a rare, multi-system genetic disease with serious neurological and mental symptoms, including autism. Mutations in the TSC1/TSC2 genes lead to the overactivation of mTOR signalling, which is also linked to nonsyndromic autism. Our aim was to analyse synaptic pathology in a transgenic model of TSC: two-month-old male B6;129S4-Tsc2tm1Djk/J mice with Tsc2 haploinsufficiency. Significant brain-region-dependent alterations in the expression of several synaptic proteins were identified. The most prominent changes were observed in the immunoreactivity of presynaptic VAMP1/2 (ca. 50% increase) and phospho-synapsin-1 (Ser62/67) (ca. 80% increase). Transmission electron microscopy demonstrated serious ultrastructural abnormalities in synapses such as a blurred structure of synaptic density and a significantly increased number of synaptic vesicles. The impairment of synaptic mitochondrial ultrastructure was represented by excessive elongation, swelling, and blurred crista contours. Polyribosomes in the cytoplasm and swollen Golgi apparatus suggest possible impairment of protein metabolism. Moreover, the delamination of myelin and the presence of vacuolar structures in the cell nucleus were observed. We also report that Tsc2+/- mice displayed increased brain weights and sizes. The behavioural analysis demonstrated the impairment of memory function, as established in the novel object recognition test. To summarise, our data indicate serious synaptic impairment in the brains of male Tsc2+/- mice.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Sinapsis , Animales , Animales Modificados Genéticamente , Trastorno del Espectro Autista/genética , Conducta Animal , Encéfalo/fisiología , Núcleo Celular/metabolismo , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Densitometría , Haploinsuficiencia , Hipocampo/metabolismo , Masculino , Ratones , Microscopía Electrónica de Transmisión , Tamaño de los Órganos , Fosforilación , ARN Mensajero/metabolismo , Reconocimiento en Psicología , Transducción de Señal , Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética
8.
Int J Mol Sci ; 21(23)2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33291628

RESUMEN

Copper is one of the most abundant basic transition metals in the human body. It takes part in oxygen metabolism, collagen synthesis, and skin pigmentation, maintaining the integrity of blood vessels, as well as in iron homeostasis, antioxidant defense, and neurotransmitter synthesis. It may also be involved in cell signaling and may participate in modulation of membrane receptor-ligand interactions, control of kinase and related phosphatase functions, as well as many cellular pathways. Its role is also important in controlling gene expression in the nucleus. In the nervous system in particular, copper is involved in myelination, and by modulating synaptic activity as well as excitotoxic cell death and signaling cascades induced by neurotrophic factors, copper is important for various neuronal functions. Current data suggest that both excess copper levels and copper deficiency can be harmful, and careful homeostatic control is important. This knowledge opens up an important new area for potential therapeutic interventions based on copper supplementation or removal in neurodegenerative diseases including Wilson's disease (WD), Menkes disease (MD), Alzheimer's disease (AD), Parkinson's disease (PD), and others. However, much remains to be discovered, in particular, how to regulate copper homeostasis to prevent neurodegeneration, when to chelate copper, and when to supplement it.


Asunto(s)
Cobre/metabolismo , Susceptibilidad a Enfermedades , Degeneración Hepatolenticular/complicaciones , Degeneración Hepatolenticular/metabolismo , Enfermedades Neurodegenerativas/etiología , Animales , Astrocitos/metabolismo , Transporte Biológico , Biomarcadores , Encéfalo/metabolismo , Encéfalo/patología , Cobre/deficiencia , Manejo de la Enfermedad , Degeneración Hepatolenticular/genética , Homeostasis , Humanos , Redes y Vías Metabólicas , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/terapia , Neuronas/metabolismo , Especificidad de Órganos
9.
Int J Mol Sci ; 21(11)2020 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-32486485

RESUMEN

The purinergic P2X7 receptor (P2X7R) belongs to a family of trimeric ion channels that are gated by extracellular adenosine 5'-triphosphate (ATP). Several studies have pointed to a role of P2X7R-dependent signalling in Parkinson's disease (PD)-related neurodegeneration. The pathology of (PD) is characterized by the formation of insoluble alpha-synuclein (α-Syn) aggregates-Lewy bodies, but the mechanisms underlying α-Syn-induced dopaminergic cell death are still partially unclear. Our previous studies indicate that extracellular α-Syn directly interact with neuronal P2X7R and induces intracellular free calcium mobilization in neuronal cells. The main objective of this study was to examine the involvement of P2X7R receptor in α-Syn-induced mitochondrial dysfunction and cell death. We found that P2X7R stimulation is responsible for α-Syn-induced oxidative stress and activation of the molecular pathways of programmed cell death. Exogenous α-Syn treatment led to P2X7R-dependent decrease in mitochondrial membrane potential as well as elevation of mitochondrial ROS production resulting in breakdown of cellular energy production. Moreover, P2X7R-dependent deregulation of AMP-activated protein kinase as well as decrease in parkin protein level could be responsible for α-Syn-induced mitophagy impairment and accumulation of dysfunctional mitochondria. P2X7R might be putative pharmacological targets in molecular mechanism of extracellular α-Syn toxicity.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Mitocondrias/patología , Neuroblastoma/metabolismo , Receptores Purinérgicos P2X7/metabolismo , alfa-Sinucleína/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adenosina Trifosfato/química , Línea Celular Tumoral , Supervivencia Celular , Radicales Libres , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Mitofagia , Neuronas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Transducción de Señal
10.
Int J Mol Sci ; 21(10)2020 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-32443651

RESUMEN

Autism spectrum disorders (ASD) are a heterogeneous group of neurodevelopmental conditions categorized as synaptopathies. Environmental risk factors contribute to ASD aetiology. In particular, prenatal exposure to the anti-epileptic drug valproic acid (VPA) may increase the risk of autism. In the present study, we investigated the effect of prenatal exposure to VPA on the synaptic morphology and expression of key synaptic proteins in the hippocampus and cerebral cortex of young-adult male offspring. To characterize the VPA-induced autism model, behavioural outcomes, microglia-related neuroinflammation, and oxidative stress were analysed. Our data showed that prenatal exposure to VPA impaired communication in neonatal rats, reduced their exploratory activity, and led to anxiety-like and repetitive behaviours in the young-adult animals. VPA-induced pathological alterations in the ultrastructures of synapses accompanied by deregulation of key pre- and postsynaptic structural and functional proteins. Moreover, VPA exposure altered the redox status and expression of proinflammatory genes in a brain region-specific manner. The disruption of synaptic structure and plasticity may be the primary insult responsible for autism-related behaviour in the offspring. The vulnerability of specific synaptic proteins to the epigenetic effects of VPA may highlight the potential mechanisms by which prenatal VPA exposure generates behavioural changes.


Asunto(s)
Trastorno del Espectro Autista/inducido químicamente , Microglía/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Sinapsis/efectos de los fármacos , Ácido Valproico/efectos adversos , Animales , Anticonvulsivantes/efectos adversos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Inflamación , Masculino , Microglía/metabolismo , Microglía/patología , Estrés Oxidativo , Embarazo , Ratas , Sinapsis/patología , Ácido Valproico/toxicidad
11.
Int J Mol Sci ; 21(11)2020 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-32521803

RESUMEN

Maternal immune activation (MIA), induced by infection during pregnancy, is an important risk factor for neuro-developmental disorders, such as autism. Abnormal maternal cytokine signaling may affect fetal brain development and contribute to neurobiological and behavioral changes in the offspring. Here, we examined the effect of lipopolysaccharide-induced MIA on neuro-inflammatory changes, as well as synaptic morphology and key synaptic protein level in cerebral cortex of adolescent male rat offspring. Adolescent MIA offspring showed elevated blood cytokine levels, microglial activation, increased pro-inflammatory cytokines expression and increased oxidative stress in the cerebral cortex. Moreover, pathological changes in synaptic ultrastructure of MIA offspring was detected, along with presynaptic protein deficits and down-regulation of postsynaptic scaffolding proteins. Consequently, ability to unveil MIA-induced long-term alterations in synapses structure and protein level may have consequences on postnatal behavioral changes, associated with, and predisposed to, the development of neuropsychiatric disorders.


Asunto(s)
Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Encefalitis/etiología , Encefalitis/metabolismo , Inmunidad , Exposición Materna , Efectos Tardíos de la Exposición Prenatal , Sinapsis/metabolismo , Factores de Edad , Animales , Trastorno Autístico/etiología , Trastorno Autístico/metabolismo , Trastorno Autístico/psicología , Conducta Animal , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Encefalitis/patología , Femenino , Lipopolisacáridos/efectos adversos , Exposición Materna/efectos adversos , Estrés Oxidativo , Fenotipo , Embarazo , Ratas
12.
J Neuroinflammation ; 15(1): 1, 2018 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-29301548

RESUMEN

BACKGROUND: Cyclin-dependent kinase 5 (Cdk5) belongs to the family of proline-directed serine/threonine kinases and plays a critical role in neuronal differentiation, migration, synaptogenesis, plasticity, neurotransmission and apoptosis. The deregulation of Cdk5 activity was observed in post mortem analysis of brain tissue of Alzheimer's disease (AD) patients, suggesting the involvement of Cdk5 in the pathomechanism of this neurodegenerative disease. However, our recent study demonstrated the important function of Cdk5 in regulating inflammatory reaction. METHODS: Since the role of Cdk5 in regulation of inflammatory signalling in AD is unknown, we investigated the involvement of Cdk5 in neuroinflammation induced by single intracerebroventricular (icv) injection of amyloid beta protein (Aß) oligomers in mouse. The brain tissue was analysed up to 35 days post injection. Roscovitine (intraperitoneal administration) was used as a potent Cdk5 inhibitor. The experiments were also performed on human neuroblastoma SH-SY5Y as well as mouse BV2 cell lines treated with exogenous oligomeric Aß. RESULTS: Our results demonstrated that single injection of Aß oligomers induces long-lasting activation of microglia and astrocytes in the hippocampus. We observed also profound, early inflammatory response in the mice hippocampus, leading to the significant elevation of pro-inflammatory cytokines expression (e.g. TNF-α, IL-1ß, IL-6). Moreover, Aß oligomers elevated the formation of truncated protein p25 in mouse hippocampus and induced overactivation of Cdk5 in neuronal cells. Importantly, administration of roscovitine reduced the inflammatory processes evoked by Aß in the hippocampus, leading to the significant decrease of cytokines level. CONCLUSIONS: These studies clearly show the involvement of Cdk5 in modulation of brain inflammatory response induced by Aß and may indicate this kinase as a novel target for pharmacological intervention in AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 5 Dependiente de la Ciclina/metabolismo , Hipocampo/metabolismo , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Fragmentos de Péptidos/toxicidad , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología , Roscovitina/farmacología
13.
Purinergic Signal ; 13(3): 347-361, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28516276

RESUMEN

Abnormalities of alpha-synuclein (ASN), the main component of protein deposits (Lewy bodies), were observed in Parkinson's disease (PD), dementia with Lewy bodies, Alzheimer's disease, and other neurodegenerative disorders. These alterations include increase in the levels of soluble ASN oligomers in the extracellular space. Numerous works have identified several mechanisms of their toxicity, including stimulation of the microglial P2X7 receptor leading to oxidative stress. While the significant role of purinergic signaling-particularly, P2 family receptors-in neurodegenerative disorders is well known, the interaction of extracellular soluble ASN with neuronal purinergic receptors is yet to be studied. Therefore, in this study, we have investigated the effect of ASN on P2 purinergic receptors and ATP-dependent signaling. We used neuroblastoma SH-SY5Y cell line and rat synaptoneurosomes treated with exogenous soluble ASN. The experiments were performed using spectrofluorometric, radiochemical, and immunochemical methods. We found the following: (i) ASN-induced intracellular free calcium mobilization in neuronal cells and nerve endings depends on the activation of purinergic P2X7 receptors; (ii) activation of P2X7 receptors leads to pannexin 1 recruitment to form an active complex responsible for ATP release; and (iii) ASN greatly decreases the activity of extracellular ecto-ATPase responsible for ATP degradation. Thus, it is concluded that purinergic receptors might be putative pharmacological targets in the molecular mechanism of extracellular ASN toxicity. Interference with P2X7 signaling seems to be a promising strategy for the prevention or therapy of PD and other neurodegenerative disorders.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Microglía/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Receptores Purinérgicos P2X7/metabolismo , alfa-Sinucleína/farmacología , Animales , Calcio/metabolismo , Línea Celular Tumoral , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Humanos , Masculino , Ratas Wistar
14.
J Neurochem ; 136(2): 222-33, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26376455

RESUMEN

Cyclin-dependent kinase 5 (Cdk5) is involved in proper neurodevelopment and brain function and serves as a switch between neuronal survival and death. Overactivation of Cdk5 is associated with many neurodegenerative disorders such as Alzheimer's or Parkinson's diseases. It is believed that in those diseases Cdk5 may be an important link between disease-initiating factors and cell death effectors. A common hallmark of neurodegenerative disorders is incorrect folding of specific proteins, thus leading to their intra- and extracellular accumulation in the nervous system. Abnormal Cdk5 signaling contributes to dysfunction of individual proteins and has a substantial role in either direct or indirect interactions of proteins common to, and critical in, different neurodegenerative diseases. While the roles of Cdk5 in α-synuclein (ASN) - tau or ß-amyloid peptide (Aß) - tau interactions are well documented, its contribution to many other pertinent interactions, such as that of ASN with Aß, or interactions of the Aß - ASN - tau triad with prion proteins, did not get beyond plausible hypotheses and remains to be proven. Understanding of the exact position of Cdk5 in the deleterious feed-forward loop critical for development and progression of neurodegenerative diseases may help designing successful therapeutic strategies of several fatal neurodegenerative diseases. Cyclin-dependent kinase 5 (Cdk5) is associated with many neurodegenerative disorders such as Alzheimer's or Parkinson's diseases. It is believed that in those diseases Cdk5 may be an important factor involved in protein misfolding, toxicity and interaction. We suggest that Cdk5 may contribute to the vicious circle of neurotoxic events involved in the pathogenesis of different neurodegenerative diseases.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Pliegue de Proteína , Péptidos beta-Amiloides/metabolismo , Animales , Humanos , Transducción de Señal , alfa-Sinucleína , Proteínas tau
15.
Neurochem Res ; 41(12): 3215-3226, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27590497

RESUMEN

Selol is an organic selenitetriglyceride formulation containing selenium at +4 oxidation level that can be effectively incorporated into catalytic sites of of Se-dependent antioxidants. In the present study, the potential antioxidative and cytoprotective effects of Selol against sodium nitroprusside (SNP)-evoked oxidative/nitrosative stress were investigated in PC12 cells and the underlying mechanisms analyzed. Spectrophoto- and spectrofluorimetic methods as well as fluorescence microscopy were used in this study; mRNA expression was quantified by real-time PCR. Selol dose-dependently improved the survival and decreased the percentage of apoptosis in PC12 cells exposed to SNP. To determine the mechanism of this protective action, the effect of Selol on free radical generation and on antioxidative potential was evaluated. Selol offered significant protection against the elevation of reactive oxidative species (ROS) evoked by SNP. Moreover, this compound restored glutathione homeostasis by ameliorating the SNP-evoked disturbance of GSH/GSSG ratio. The protective effect exerted by Selol was associated with the prevention of SNP-mediated down-regulation of antioxidative enzymes: glutathione peroxidase (Se-GPx), glutathione reductase (GR), and thioredoxin reductase (TrxR). Finally, GPx inhibition significantly abolished the cytoprotective effect of Selol. In conclusion, these results suggest that Selol effectively protected PC12 cells against SNP-induced oxidative damage and death by adjusting free radical levels and antioxidant system, and suppressing apoptosis. Selol could be successfully used in the treatments of diseases that involve oxidative stress and resulting apoptosis.


Asunto(s)
Antioxidantes/farmacología , Nitroprusiato/farmacología , Estrés Oxidativo/efectos de los fármacos , Compuestos de Selenio/farmacología , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citoprotección , Radicales Libres/metabolismo , Glutatión/metabolismo , Nitrosación , Células PC12 , Ratas
16.
Pol Merkur Lekarski ; 39(234): 382-8, 2015 Dec.
Artículo en Polaco | MEDLINE | ID: mdl-26802693

RESUMEN

Ventilator-associated pneumonia (VAP) is one of the most common hospital-acquired infections. VAP is associated with prolonged hospitalization and visibly increased mortality, which in the group of patients with VAP ranges from 25% to 47%. In January 2013 Centers of Disease Control and Prevention introduced a new definition for VAP. Subjective criteria in the previous VAP definition were the reason for difficulties in VAP surveillance and assessment of efficacy of ventilator bundles and other quality improvement initiatives. The purpose of this article is to summarise the new definition of VAP and the first researches after two years of use of the new definition. The new definition of ventilator-associated events (VAE) identifies a broader group of patients than the previous VAP definition. Surveillance of all complications of mechanical ventilation aimed to create more efficient prophylaxis bundles and to decrease the mortality in critically ill patients. The latest published studies suggest that most of the complications defined as VAE are patient-related, not modifiable risk factors and these patients had no evidence of hospital-acquired complications. The new definition failed to detect many patients with VAP and it has not resolved the ambiguities related to the diagnosis of this complication. It seems that the new surveillance program will not lead to introducing new prevention strategies that could decrease the mortality in intensive care unit patients.


Asunto(s)
Neumonía Asociada al Ventilador/etiología , Respiración Artificial/efectos adversos , Cuidados Críticos , Humanos , Neumonía Asociada al Ventilador/clasificación , Neumonía Asociada al Ventilador/epidemiología , Factores de Riesgo
18.
Postepy Hig Med Dosw (Online) ; 67: 1047-57, 2013 Nov 04.
Artículo en Polaco | MEDLINE | ID: mdl-24184956

RESUMEN

Recently published data demonstrated that increased release, oligomerization and toxicity of α-synuclein (ASN) is a key molecular process in pathophysiology of neurodegenerative diseases classified as synucleinopathies (e.g. Parkinson disease or Alzheimer's disease). It was proved that the excessive release of ASN into the extracellular space, driven by environmental factors as well as neurodegeneration, may have a significant role in the spread of the neurodegeneration process within the brain. Extracellular ASN was shown to be toxic both to neurons and glial cells and the mechanism of its action depends on the concentration of this protein in the extracellular space. Exogenous ASN leads to the activation of plasma membrane receptors, causes increased calcium influx, and stimulates the synthesis of proinflammatory cytokines and nitric oxide, which in turn leads to activation of programmed cell death. These data provide new insights into the involvement of ASN in the neurodegenerative diseases, and thus can serve effectively for the development of their new therapy.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Muerte Celular/fisiología , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/metabolismo , Apoptosis , Encéfalo/metabolismo , Espacio Extracelular/metabolismo , Humanos , Neuroglía/metabolismo , Neuronas/metabolismo , Óxido Nítrico/metabolismo
19.
Brain Sci ; 13(7)2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37509018

RESUMEN

Recent data suggest that defects in purinergic signalling are a common denominator of autism spectrum disorders (ASDs), though nothing is known about whether the disorder-related imbalance occurs at the receptor level. In this study, we investigated whether prenatal exposure to valproic acid (VPA) induces changes in purinergic receptor expression in adolescence and whether it corresponds to glial cell activation. Pregnant dams were subjected to an intraperitoneal injection of VPA at embryonic day 12.5. In the hippocampi of adolescent male VPA offspring, we observed an increase in the level of P2X1, with concomitant decreases in P2X7 and P2Y1 receptors. In contrast, in the cortex, the level of P2X1 was significantly reduced. Also, significant increases in cortical P2Y1 and P2Y12 receptors were detected. Additionally, we observed profound alterations in microglial cell numbers and morphology in the cortex of VPA animals, leading to the elevation of pro-inflammatory cytokine expression. The changes in glial cells were partially reduced via a single administration of a non-selective P2 receptor antagonist. These studies show the involvement of purinergic signalling imbalance in the modulation of brain inflammatory response induced via prenatal VPA exposure and may indicate that purinergic receptors are a novel target for pharmacological intervention in ASDs.

20.
Folia Neuropathol ; 60(4): 390-402, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36734381

RESUMEN

Purinergic signalling is involved in the control of several processes related to brain development, such as neurogenesis and gliogenesis, migration and differentiation of neuronal precursors, synaptogenesis and synaptic elimination to achieve a fully wired and efficient mature brain. Therefore, any deregulation of purine-dependent signalling mediated by stimulation of specific adenosine and purinergic receptor subtypes: P1, P2X, or P2Y, can lead to functional deficits and the development of neuropsychiatric disorders, including autism spectrum disorders (ASD). In this study, we investigated the changes in expression and activity of selected purinergic receptors during rat brain development in an animal model of ASD. Pregnant dams received an intraperitoneal injection of valproic acid (VPA; 450 mg/kg body weight) at embryonic day (ED) 12.5, around the time of neural tube closure. Subsequently, changes in the expression and activity of specific purinergic receptor subtypes were analysed at ED19, an important prenatal stage of brain development. Our results suggest that prenatal VPA exposure leads to a significant increase in the level and activity of adenosinergic receptors A1, A2b and A3, which are involved in the regulation of progenitor cell proliferation and nerve growth, and upregulation of purinergic P2X2/P2X3 receptors, which in turn may contribute to the postnatal neuroanatomical abnormalities and synaptic dysfunction. Conversely, the significant downregulation of P2Y1 and P2X7 receptors, together with their reduced activity in the embryonic VPA brain, may indicate disturbances in the processes of neuronal precursor migration and differentiation, dendritic and axonal formation, and glutamate/GABA imbalance, thereby altering neuronal excitability. In conclusion, defects in purinergic signalling induced by prenatal VPA exposure could have a profound impact on brain development during embryogenesis and on intellectual and behavioural functions after birth. These observations could provide clues for future implementation of potential therapeutic strategies for ASD.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Receptores Purinérgicos P2 , Animales , Femenino , Ratas , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Encéfalo/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptores Purinérgicos/metabolismo , Receptores Purinérgicos P2/metabolismo , Ácido Valproico/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA