Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Pharmacol Rep ; 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39298028

RESUMEN

The group III metabotropic glutamate receptors (mGluRs), comprising mGluR4, mGluR6, mGluR7, and mGluR8, offer neuroprotective potential in mitigating excitotoxicity during ischemic brain injury, particularly in neonatal contexts. They are G-protein coupled receptors that inhibit adenylyl cyclase and reduce neurotransmitter release, mainly located presynaptically and acting as autoreceptors. This review aims to examine the differential expression and function of group III mGluRs across various brain regions such as the cortex, hippocampus, and cerebellum, with a special focus on the neonatal stage of development. Glutamate excitotoxicity plays a crucial role in the pathophysiology of brain ischemia in neonates. While ionotropic glutamate receptors are traditional targets for neuroprotection, their direct inhibition often leads to severe side effects due to their critical roles in normal neurotransmission and synaptic plasticity. Group III mGluRs provide a more nuanced and potentially safer approach by modulating rather than blocking glutamatergic transmission. Their downstream signaling cascade results in the regulation of intracellular calcium levels, neuronal hyperpolarization, and reduced neurotransmitter release, effectively decreasing excitotoxic signaling without completely suppressing essential glutamatergic functions. Importantly, the neuroprotective effects of group III mGluRs extend beyond direct modulation of glutamate release influencing glial cell function, neuroinflammation, and oxidative stress, all of which contribute to secondary injury cascades in brain ischemia. This comprehensive analysis of group III mGluRs multifaceted neuroprotective potential provides valuable insights for developing novel therapeutic strategies to combat excitotoxicity in neonatal ischemic brain injury.

2.
Pharmacol Rep ; 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39289333

RESUMEN

Injury to the developing central nervous system resulting from perinatal hypoxia-ischemia (HI) is still a clinical challenge. The only approach currently available in clinical practice for severe cases of HI is therapeutic hypothermia, initiated shortly after birth and supported by medications to regulate blood pressure, control epileptic seizures, and dialysis to support kidney function. However, these treatments are not effective enough to significantly improve infant survival or prevent brain damage. The need to create a new effective therapy has focused attention on metabotropic glutamate receptors (mGluR), which control signaling pathways involved in HI-induced neurodegeneration. The complexity of mGluR actions, considering their localization and developmental changes, and the functions of each subtype in HI-evoked brain damage, combined with difficulties in the availability of safe and effective modulators, raises the question whether modulation of mGluRs with subtype-selective ligands can become a new treatment in neonatal HI. Addressing this question, this review presents the available information concerning the role of each of the eight receptor subtypes of the three mGluR groups (group I, II, and III). Data obtained from experiments performed on in vitro and in vivo neonatal HI models show the neuroprotective potential of group I mGluR antagonists, as well as group II and III agonists. The information collected in this work indicates that the neuroprotective effects of manipulating mGluR in experimental HI models, despite the need to create more safe and selective ligands for particular receptors, provide a chance to create new therapies for the sensitive brains of infants at risk.

4.
Front Neurol ; 15: 1386695, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38685945

RESUMEN

Birth asphyxia and its main sequel, hypoxic-ischemic encephalopathy, are one of the leading causes of children's deaths worldwide and can potentially worsen the quality of life in subsequent years. Despite extensive research efforts, efficient therapy against the consequences of hypoxia-ischemia occurring in the perinatal period of life is still lacking. The use of hyperbaric oxygen, improving such vital consequences of birth asphyxia as lowered partial oxygen pressure in tissue, apoptosis of neuronal cells, and impaired angiogenesis, is a promising approach. This review focused on the selected aspects of mainly experimental hyperbaric oxygen therapy. The therapeutic window for the treatment of perinatal asphyxia is very narrow, but administering hyperbaric oxygen within those days improves outcomes. Several miRNAs (e.g., mir-107) mediate the therapeutic effect of hyperbaric oxygen by modulating the Wnt pathway, inhibiting apoptosis, increasing angiogenesis, or inducing neural stem cells. Combining hyperbaric oxygen therapy with drugs, such as memantine or ephedrine, produced promising results. A separate aspect is the use of preconditioning with hyperbaric oxygen. Overall, preliminary clinical trials with hyperbaric oxygen therapy used in perinatal asphyxia give auspicious results.

5.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35806000

RESUMEN

Birth asphyxia causes brain injury in neonates, but a fully successful treatment has yet to be developed. This study aimed to investigate the effect of group II mGlu receptors activation after experimental birth asphyxia (hypoxia-ischemia) on the expression of factors involved in apoptosis and neuroprotective neurotrophins. Hypoxia-ischemia (HI) on 7-day-old rats was used as an experimental model. The effects of intraperitoneal application of mGluR2 agonist LY379268 (5 mg/kg) and the specific mGluR3 agonist NAAG (5 mg/kg) (1 h or 6 h after HI) on apoptotic processes and initiation of the neuroprotective mechanism were investigated. LY379268 and NAAG applied shortly after HI prevented brain damage and significantly decreased pro-apoptotic Bax and HtrA2/Omi expression, increasing expression of anti-apoptotic Bcl-2. NAAG or LY379268 applied at both times also decreased HIF-1α formation. HI caused a significant decrease in BDNF concentration, which was restored after LY379268 or NAAG administration. HI-induced increase in GDNF concentration was decreased after administration of LY379268 or NAAG. Our results show that activation of mGluR2/3 receptors shortly after HI prevents brain damage by the inhibition of excessive glutamate release and apoptotic damage decrease. mGluR2 and mGluR3 agonists produced comparable results, indicating that both receptors may be a potential target for early treatment in neonatal HI.


Asunto(s)
Asfixia , Lesiones Encefálicas , Factor Neurotrófico Derivado del Encéfalo , Factor Neurotrófico Derivado de la Línea Celular Glial , Receptores de Glutamato Metabotrópico , Aminoácidos/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Asfixia/metabolismo , Asfixia/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Dipéptidos/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hipoxia/metabolismo , Hipoxia/patología , Fármacos Neuroprotectores/farmacología , Ratas , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/metabolismo
6.
Antioxidants (Basel) ; 10(11)2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34829646

RESUMEN

The over-activation of NMDA receptors and oxidative stress are important components of neonatal hypoxia-ischemia (HI). Kynurenic acid (KYNA) acts as an NMDA receptor antagonist and is known as a reactive oxygen species (ROS) scavenger, which makes it a potential therapeutic compound. This study aimed to establish the neuroprotective and antioxidant potential of KYNA in an experimental model of HI. HI on seven-day-old rats was used as an experimental model. The animals were injected i.p. with different doses of KYNA 1 h or 6 h after HI. The neuroprotective effect of KYNA was determined by the measurement of brain damage and elements of oxidative stress (ROS and glutathione (GSH) level, SOD, GPx, and catalase activity). KYNA applied 1 h after HI significantly reduced weight loss of the ischemic hemisphere, and prevented neuronal loss in the hippocampus and cortex. KYNA significantly reduced HI-increased ROS, GSH level, and antioxidant enzyme activity. Only the highest used concentration of KYNA showed neuroprotection when applied 6 h after HI. The presented results indicate induction of neuroprotection at the ROS formation stage. However, based on the presented data, it is not possible to pinpoint whether NMDA receptor inhibition or the scavenging abilities are the dominant KYNA-mediated neuroprotective mechanisms.

7.
Oxid Med Cell Longev ; 2021: 8848015, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763176

RESUMEN

Hypoxia-ischemia (HI) in an immature brain results in energy depletion and excessive glutamate release resulting in excitotoxicity and oxidative stress. An increase in reactive oxygen species (ROS) production induces apoptotic processes resulting in neuronal death. Activation of group II mGluR was shown to prevent neuronal damage after HI. The application of agonists of mGluR3 (N-acetylaspartylglutamate; NAAG) or mGluR2 (LY379268) inhibits the release of glutamate and reduces neurodegeneration in a neonatal rat model of HI, although the exact mechanism is not fully recognized. In the present study, the effects of NAAG (5 mg/kg) and LY379268 (5 mg/kg) application (24 h or 1 h before experimental birth asphyxia) on apoptotic processes as the potential mechanism of neuroprotection in 7-day-old rats were investigated. Intraperitoneal application of NAAG or LY379268 at either time point before HI significantly reduced the number of TUNEL-positive cells in the CA1 region of the ischemic brain hemisphere. Both agonists reduced expression of the proapoptotic Bax protein and increased expression of Bcl-2. Decreases in HI-induced caspase-9 and caspase-3 activity were also observed. Application of NAAG or LY379268 24 h or 1 h before HI reduced HIF-1α formation likely by reducing ROS levels. It was shown that LY379268 concentration remains at a level that is required for activation of mGluR2 for up to 24 h; however, NAAG is quickly metabolized by glutamate carboxypeptidase II (GCPII) into glutamate and N-acetyl-aspartate. The observed effect of LY379268 application 24 h or 1 h before HI is connected with direct activation of mGluR2 and inhibition of glutamate release. Based on the data presented in this study and on our previous findings, we conclude that the neuroprotective effect of NAAG applied 1 h before HI is most likely the result of a combination of mGluR3 and NMDA receptor activation, whereas the beneficial effects of NAAG pretreatment 24 h before HI can be explained by the activation of NMDA receptors and induction of the antioxidative/antiapoptotic defense system triggered by mild excitotoxicity in neurons. This response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning.


Asunto(s)
Apoptosis , Hipoxia-Isquemia Encefálica/patología , Receptores de Glutamato Metabotrópico/agonistas , Aminoácidos/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Dipéptidos/farmacología , Femenino , Hipocampo/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas Wistar , Receptores de Glutamato Metabotrópico/metabolismo , Proteína X Asociada a bcl-2/metabolismo
8.
Antioxidants (Basel) ; 9(9)2020 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-32957477

RESUMEN

N-acetylaspartylglutamate (NAAG), the most abundant peptide transmitter in the mammalian nervous system, activates mGluR3 at presynaptic sites, inhibiting the release of glutamate, and acts on mGluR3 on astrocytes, stimulating the release of neuroprotective growth factors (TGF-ß). NAAG can also affect N-methyl-d-aspartate (NMDA) receptors in both synaptic and extrasynaptic regions. NAAG reduces neurodegeneration in a neonatal rat model of hypoxia-ischemia (HI), although the exact mechanism is not fully recognized. In the present study, the effect of NAAG application 24 or 1 h before experimental birth asphyxia on oxidative stress markers and the potential mechanisms of neuroprotection on 7-day old rats was investigated. The intraperitoneal application of NAAG at either time point before HI significantly reduced the weight deficit of the ischemic brain hemisphere, radical oxygen species (ROS) content and activity of antioxidant enzymes, and increased the concentration of reduced glutathione (GSH). No additional increase in the TGF-ß concentration was observed after NAAG application. The fast metabolism of NAAG and the decrease in TGF-ß concentration that resulted from NAAG pretreatment, performed up to 24 h before HI, excluded the involvement mGluR3 in neuroprotection. The observed effect may be explained by the activation of NMDA receptors induced by NAAG pretreatment 24 h before HI. Inhibition of the NAAG effect by memantine supports this conclusion. NAAG preconditioning 1 h before HI results in a mixture of mGluR3 and NMDA receptor activation. Preconditioning with NAAG induces the antioxidative defense system triggered by mild excitotoxicity in neurons. Moreover, this response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning. However, this theory requires further investigation.

9.
Cancers (Basel) ; 12(5)2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32413951

RESUMEN

Malignant brain tumor-glioblastoma is not only difficult to treat but also hard to study and model. One of the reasons for these is their heterogeneity, i.e., individual tumors consisting of cancer cells that are unlike each other. Such diverse cells can thrive due to the simultaneous co-evolution of anatomic niches and adaption into zones with distorted homeostasis of oxygen. It dampens cytotoxic and immune therapies as the response depends on the cellular composition and its adaptation to hypoxia. We explored what transcriptome reposition strategies are used by cells in the different areas of the tumor. We created the hypoxic map by differential expression analysis between hypoxic and cellular features using RNA sequencing data cross-referenced with the tumor's anatomic features (Ivy Glioblastoma Atlas Project). The molecular functions of genes differentially expressed in the hypoxic regions were analyzed by a systematic review of the gene ontology analysis. To put a hypoxic niche signature into a clinical context, we associated the model with patients' survival datasets (The Cancer Genome Atlas). The most unique class of genes in the hypoxic area of the tumor was associated with the process of autophagy. Both hypoxic and cellular anatomic features were enriched in immune response genes whose, along with autophagy cluster genes, had the power to predict glioblastoma patient survival. Our analysis revealed that transcriptome responsive to hypoxia predicted worse patients' outcomes by driving tumor cell adaptation to metabolic stress and immune escape.

10.
Neurobiol Learn Mem ; 171: 107209, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32147584

RESUMEN

An increase in the intracellular Ca2+ level in neurons is one of the main steps in the memory formation cascade. The increase results from extracellular Ca2+ influx by activation of ionotropic glutamate receptors and release from intracellular stores by the stimulation of IP3 receptors (IP3Rs) via group I metabotropic glutamate receptors (mGluR1/5). Recent data indicate an additional mechanism resulting in Ca2+ influx into neurons, triggered by intracellular signals that are directly connected to the activation of group I mGluRs. This influx occurs through transient receptor potential (TRP) channels, which are permeable to Na+, K+ and, mainly, Ca2+. These channels are activated by increases in intracellular Ca2+, diacylglycerol (DAC) and inositol 1,4,5-triphosphate (IP3) level resulting from a group I mGluR activation. The aim of the present study was to investigate the participation of TRP channels, especially from TRPC and TRPV groups, in memory consolidation and reconsolidation and memory retrieval processes in a passive avoidance task in one-day old chicks. TRP channels were blocked by the injection of the unspecific channel modulators SKF 96365 (2.5 µl 30 µM/hemisphere) and 2-APB (2.5 µl 10 µM/hemisphere) directly into the intermediate medial mesopallium (IMM) region of the chick brain immediately after initial training or after a reminder. The inhibition of specific TRP channels (TRPV1, TRPV3 or TRPC3) was achieved by the application of selective antibodies. Our results demonstrate that the inhibition of TRP channels by the application of both modulators disrupted memory consolidation, resulting in permanent task amnesia. The inhibition of the TRPV1, TRPC3 and TRPV3 channels by specific antibodies resulted in similar amnesia. Moreover, the inhibition of TRP channels by SKF 96365 and 2-APB at different time points after initial training or after the reminder also resulted in amnesia, indicating the role of TRP channels in memory retrieval. The inhibition of calcium influx through these channels resulted in permanent memory disruption, which suggests that the calcium signal generated by TRP channels is crucial for memory formation and retrieval processes. For the first time, the important role of TRPV3 channels in memory formation was demonstrated.


Asunto(s)
Reacción de Prevención/fisiología , Memoria/fisiología , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Reacción de Prevención/efectos de los fármacos , Compuestos de Boro/farmacología , Calcio/metabolismo , Pollos , Cognición/efectos de los fármacos , Imidazoles/farmacología , Masculino , Memoria/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores
11.
Noncoding RNA ; 5(1)2019 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-30875963

RESUMEN

Malignant glioblastoma (GBM, glioma) is the most common and aggressive primary adult brain tumor. The prognosis of GBM patients remains poor, despite surgery, radiation and chemotherapy. The major obstacles for successful remedy are invasiveness and therapy resistance of GBM cells. Invasive glioma cells leave primary tumor core and infiltrate surrounding normal brain leading to inevitable recurrence, even after surgical resection, radiation and chemotherapy. Therapy resistance allowing for selection of more aggressive and resistant sub-populations including GBM stem-like cells (GSCs) upon treatment is another serious impediment to successful treatment. Through their regulation of multiple genes, microRNAs can orchestrate complex programs of gene expression and act as master regulators of cellular processes. MicroRNA-based therapeutics could thus impact broad cellular programs, leading to inhibition of invasion and sensitization to radio/chemotherapy. Our data show that miR-451 attenuates glioma cell migration in vitro and invasion in vivo. In addition, we have found that miR-451 sensitizes glioma cells to conventional chemo- and radio-therapy. Our data also show that miR-451 is regulated in vivo by AMPK pathway and that AMPK/miR-451 loop has the ability to switch between proliferative and migratory pattern of glioma cells behavior. We therefore postulate that AMPK/miR-451 negative reciprocal feedback loop allows GBM cells/GSCs to adapt to tumor "ecosystem" by metabolic and behavioral flexibility, and that disruption of such a loop reduces invasiveness and diminishes therapy resistance.

12.
Noncoding RNA ; 5(1)2019 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-30813461

RESUMEN

The mammalian brain is made up of billions of neurons and supporting cells (glial cells), intricately connected. Molecular perturbations often lead to neurodegeneration by progressive loss of structure and malfunction of neurons, including their death. On the other side, a combination of genetic and cellular factors in glial cells, and less frequently in neurons, drive oncogenic transformation. In both situations, microenvironmental niches influence the progression of diseases and therapeutic responses. Dynamic changes that occur in cellular transcriptomes during the progression of developmental lineages and pathogenesis are controlled through a variety of regulatory networks. These include epigenetic modifications, signaling pathways, and transcriptional and post-transcriptional mechanisms. One prominent component of the latter is small non-coding RNAs, including microRNAs, that control the vast majority of these networks including genes regulating neural stemness, differentiation, apoptosis, projection fates, migration and many others. These cellular processes are also profoundly dependent on the microenvironment, stemness niche, hypoxic microenvironment, and interactions with associated cells including endothelial and immune cells. Significantly, the brain of all other mammalian organs expresses the highest number of microRNAs, with an additional gain in expression in the early stage of neurodegeneration and loss in expression in oncogenesis. However, a mechanistic explanation of the concept of an apparent inverse correlation between the odds of cancer and neurodegenerative diseases is only weakly developed. In this review, we thus will discuss widespread de-regulation of microRNAome observed in these two major groups of brain pathologies. The deciphering of these intricacies is of importance, as therapeutic restoration of pre-pathological microRNA landscape in neurodegeneration must not lead to oncogenesis and vice versa. We thus focus on microRNAs engaged in cellular processes that are inversely regulated in these diseases. We also aim to define the difference in microRNA networks between pro-survival and pro-apoptotic signaling in the brain.

13.
Chemosphere ; 223: 64-73, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30769291

RESUMEN

The brominated flame retardant tetrabromobisphenol A (TBBPA) is toxic to cultured brain neurons, and glutamate receptors partially mediate this effect; consequently, the depolarizing effect of TBBPA on neurons is to be expected, but it is yet to be actually demonstrated. The aim of this study was to detect TBBPA-evoked depolarization and identify the underlying mechanisms. The plasma membrane potential of rat cerebellar granule cells (CGC) in cerebellar slices or in primary cultures was measured using whole-cell current clamp recordings, or the fluorescent probe oxonol VI, respectively. The contribution of NMDA and AMPA receptors, voltage-gated sodium channels and intracellular calcium mobilization was tested using their selective antagonists or inhibitors. Direct interactions of TBBPA with NMDARs were tested by measuring the specific binding of radiolabeled NMDAR ligands to isolated rat cortical membrane fraction. TBBPA (25 µM) strongly depolarized CGC in cerebellar slices, and at ≥ 7.5 µM concentration-dependently depolarized primary CGC cultures. Depolarization of the primary CGC by 25 µM TBBPA was partly reduced when MK-801 was applied alone or in combination with either TTX or CNQX, or where bastadin 12 was applied in combination with ryanodine, whereas depolarization was completely prevented when MK-801, CNQX and TTX where combined. TBBPA had no effect on the specific binding of NMDAR radio-ligands to isolated cortical membranes. These results demonstrate the depolarizing effect of TBBPA on CGC, which is mainly mediated by ionotropic glutamate receptors, while voltage-gated sodium channels are also involved. We found no evidence for the direct activation of NMDARs by TBBPA.


Asunto(s)
Cerebelo/patología , Potenciales de la Membrana/efectos de los fármacos , Bifenilos Polibrominados/toxicidad , Animales , Células Cultivadas , Retardadores de Llama/toxicidad , Fármacos Neuromusculares Despolarizantes , Neuronas/patología , Técnicas de Placa-Clamp , Ratas , Receptores Ionotrópicos de Glutamato/metabolismo , Receptores Ionotrópicos de Glutamato/fisiología
14.
PLoS One ; 13(7): e0200933, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30044838

RESUMEN

Birth asphyxia resulting in brain hypoxia-ischemia (H-I) can cause neonatal death or lead to persistent brain damage. Recent investigations have shown that group II metabotropic glutamate receptor (mGluR2/3) activation can provide neuroprotection against H-I but the mechanism of this effect is not clear. The aim of this study was to investigate whether mGluR2/3 agonists applied a short time after H-I reduce brain damage in an experimental model of birth asphyxia, and whether a decrease in oxidative stress plays a role in neuroprotection. Neonatal H-I in 7-day-old rats was used as an experimental model of birth asphyxia. Rats were injected intra peritoneally with mGluR2 (LY 379268) or mGluR3 (NAAG) agonists 1 h or 6 h after H-I (5 mg/kg). The weight deficit of the ischemic brain hemisphere, radical oxygen species (ROS) content levels, antioxidant enzymes activity and the concentrations of reduced glutathione (GSH) were measured. Both agonists reduced weight loss in the ischemic hemisphere and mitigated neuronal degeneration in the CA1 hippocampal region and cerebral cortex. Both agonists reduced the elevated levels of ROS in the ipsilateral hemisphere observed after H-I and prevented an increase in antioxidant enzymes activity in the injured hemisphere restoring them to control levels. A decrease in GSH level was also restored after agonists application. The results show that the activation of mGluR2 and mGluR3 a short time after H-I triggers neuroprotective mechanisms that act through the inhibition of oxidative stress and ROS production. The prevention of ROS production by the inhibition of glutamate release and decrease in its extracellular concentration is likely the main mechanism involved in the observed neuroprotection.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Receptores de Glutamato Metabotrópico/agonistas , Animales , Animales Recién Nacidos , Antioxidantes/metabolismo , Encéfalo/enzimología , Femenino , Glutatión/metabolismo , Hipoxia-Isquemia Encefálica/enzimología , Masculino , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
15.
Brain Sci ; 8(3)2018 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-29562588

RESUMEN

Hypoxia-ischemia (H-I) at the time of birth may cause neonatal death or lead to persistent brain damage. The search for an effective treatment of asphyxiated infants has not resulted in an effective therapy, and hypothermia remains the only available therapeutic strategy. Among possible experimental therapies, the induction of ischemic tolerance is promising. Recent investigations have shown that activation of group II metabotropic glutamate receptors (mGluR2/3) can provide neuroprotection against H-I, but the mechanism of this effect is not clear. The aim of this study was to investigate whether an mGluR2/3 agonist applied before H-I reduces brain damage in an experimental model of birth asphyxia and whether a decrease in oxidative stress plays a role in neuroprotection. Neonatal H-I on seven-day-old rats was used as an experimental model of birth asphyxia. Rats were injected intraperitoneally with the mGluR2/3 agonist LY379268 24 or 1 h before H-I (5 mg/kg). LY379268 reduced the infarct area in the ischemic hemisphere. Application of the agonist at both times also reduced the elevated levels of reactive oxygen species (ROS) in the ipsilateral hemisphere observed after H-I and prevented the increase in antioxidant enzyme activity in the injured hemisphere. The decrease in glutathione (GSH) level was also restored after agonist application. The results suggest that the neuroprotective mechanisms triggered by the activation of mGluR2/3 before H-I act through the decrease of glutamate release and its extracellular concentration resulting in the inhibition of ROS production and reduction of oxidative stress. This, rather than induction of ischemic tolerance, is probably the main mechanism involved in the observed neuroprotection.

16.
Gene Expr Patterns ; 25-26: 85-91, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28625896

RESUMEN

Real-time quantitative PCR is an exceptionally sensitive method that can detect even very small differences in gene expression and, as such, it is essential to use suitable reference genes. Domestic chickens are used in a wide range of studies including neurobiology, behavior, ecology and disease transmission. In recent avian gene expression experiments, 18S (18S ribosomal RNA), beta actin (ACTB) and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) have frequently been used; however, there is not enough evidence that these reference genes are suitable for all types of experiments. There is considerable evidence for lateralization in numerous learning tasks and for differences in the functional contribution of the two brain hemispheres. Therefore, the purpose of this study was to identify a set of reference genes for chick brain region called an intermediate medial mesopallium (IMM), which is connected with memory formation in the chick brain, whilst also taking into consideration the differences between the left and right hemispheres. This study evaluated the expression stability of eleven candidate housekeeping genes in the IMM region of the 1-day old chick brain. In our experimental system, the most reliable results were given by the NormFinder algorithm. The results show for the first time that ACTB, commonly used as an avian reference gene, is not suitable for investigation of gene expression in the chick brain and that brain lateralization exact selection of different reference gens for each hemisphere. For memory process investigations using tasks in one-day old chicks the most effective reference genes for the left hemisphere were HMBS and SDHA, and for the right hemisphere the most effective was RPL19.


Asunto(s)
Proteínas Aviares/genética , Encéfalo/fisiología , Pollos/genética , Perfilación de la Expresión Génica/normas , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Actinas/genética , Algoritmos , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Lateralidad Funcional , Genes Esenciales , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , ARN Ribosómico 18S/genética
17.
Stem Cell Reports ; 8(6): 1497-1505, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28528698

RESUMEN

Despite the importance of molecular subtype classification of glioblastoma (GBM), the extent of extracellular vesicle (EV)-driven molecular and phenotypic reprogramming remains poorly understood. To reveal complex subpopulation dynamics within the heterogeneous intratumoral ecosystem, we characterized microRNA expression and secretion in phenotypically diverse subpopulations of patient-derived GBM stem-like cells (GSCs). As EVs and microRNAs convey information that rearranges the molecular landscape in a cell type-specific manner, we argue that intratumoral exchange of microRNA augments the heterogeneity of GSC that is reflected in highly heterogeneous profile of microRNA expression in GBM subtypes.


Asunto(s)
Neoplasias Encefálicas/patología , Vesículas Extracelulares/metabolismo , Glioblastoma/patología , MicroARNs/metabolismo , Antígeno AC133/metabolismo , Animales , Neoplasias Encefálicas/genética , Exosomas/metabolismo , Femenino , Glioblastoma/genética , Humanos , Ratones , Ratones Desnudos , MicroARNs/genética , Invasividad Neoplásica , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Fenotipo , Tetraspanina 30/metabolismo , Transcriptoma , Trasplante Heterólogo , Células Tumorales Cultivadas
18.
Pharmacol Rep ; 68(5): 1076-83, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27552063

RESUMEN

BACKGROUND: Perinatal hypoxia-ischemia causes brain injury in neonates, but a fully successful treatment to prevent changes in the brain has yet to be developed. The aim of this study was to evaluate the effect of combining memantine treatment with HBO (2.5 ATA) or HH (0.47 ATA) on neonatal hypoxia-ischemia brain injury. METHODS: 7-day old rats were subjected to hypoxia-ischemia (H-I) and treated with combination of memantine and HBO or HH. The brain damage was evaluated by examination of infarct area and the number of apoptotic cells in CA1 region of hippocampus. Additionally, the level of reactive oxygen species (ROS) was measured. RESULTS: Memantine, HBO or HH postconditioning applied at short time (1-6h) after H-I, and repeated for two subsequent days, resulted in significant neuroprotection. The reduction in ipsilateral hemisphere weight deficit and in the size of infarct area was observed 14days after H-I. A reduction in apoptosis and ROS level was also observed. Combining memantine with HBO or HH resulted in a loss of neuroprotection. CONCLUSIONS: Our results show that, combining HBO or HH postconditioning with memantine produce no additive increase in the neuroprotective effect. On the contrary, combining the treatments resulted in lower neuroprotection in comparison to the effects of memantine, HBO or HH alone.


Asunto(s)
Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Memantina/farmacología , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oxígeno/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Modelos Animales de Enfermedad , Oxigenoterapia Hiperbárica/métodos , Hipoxia/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
19.
Brain Res ; 1648(Pt A): 257-265, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27431936

RESUMEN

Ischemic preconditioning with sublethal stress triggers defensive mechanisms against ischemic brain damage; however, such manipulations are potentially dangerous and, therefore, safe stimuli have been sought. Hyperoxia preconditioning by administration of hyperbaric (HBO) or normobaric oxygen (NBO) may have neuroprotective potential. The aim of this study was to determine whether preconditioning with HBO and air (HBA) applied at 2.5 absolute pressure (ATA) or NBO preconditioning induces ischemic tolerance in the brain of gerbils subjected to 3min transient cerebral ischemia. Neuronal cell survival, changes in brain temperature, the generation of factors involved in neurodegeneration and basic behavior in nest building were all tested. Hyperoxic preconditioning prevented ischemia-induced neuronal cell loss, reduced the number of TUNEL positive cells in the CA1 region of the hippocampus and improved the nest building process compared to untreated ischemic animals. Preconditioning also suppressed the production of reactive oxygen species and increased Bax expression normally observed after an ischemic episode. Only HBO preconditioning inhibited ischemia-evoked increases in brain temperature. Our results show that hyperoxic preconditioning results in induction of ischemic tolerance and prevents ischemia-induced neuronal damage in the gerbil brain. Pressurized air preconditioning was as effective as HBO or NBO preconditioning in providing neuroprotection. The observed neuroprotection probably results from mild oxidative stress evoked by increased brain tissue oxidation and activation of antioxidant and antiapoptotic defenses.


Asunto(s)
Oxigenoterapia Hiperbárica/métodos , Ataque Isquémico Transitorio/prevención & control , Precondicionamiento Isquémico/métodos , Animales , Apoptosis , Temperatura Corporal , Encéfalo/metabolismo , Supervivencia Celular , Gerbillinae , Hipocampo/patología , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Masculino , Comportamiento de Nidificación , Prosencéfalo/fisiopatología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno , Proteína X Asociada a bcl-2/metabolismo
20.
Neurochem Res ; 40(11): 2200-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26318863

RESUMEN

The study assessed involvement of Ca(2+) signaling mediated by the metabotropic glutamate receptors mGluR1/5 in brain tolerance induced by hypoxic preconditioning. Acute slices of rat piriform cortex were tested 1 day after exposure of adult rats to mild hypobaric hypoxia for 2 h at a pressure of 480 hPa once a day for three consecutive days. We detected 44.1 ± 11.6 % suppression of in vitro anoxia-induced increases of intracellular Ca(2+) levels and a fivefold increase in Ca(2+) transients evoked by selective mGluR1/5 agonist, DHPG. Western blot analysis of cortical homogenates demonstrated a 11 ± 4 % decrease in mGluR1 immunoreactivity (IR), and in the nuclei-enriched fraction a 12 ± 3 % increase in IR of phospholipase Cß1 (PLCß1), which is a major mediator of mGluR1/5 signaling. Immunocytochemical analysis of the cortex revealed increase in the mGluR1/5 and PLCß1 IR in perikarya, and a decrease in IR of the neuronal inositol trisphosphate receptors (IP3Rs). We suggest that enhanced expression of mGluR5 and PLCß1 and potentiation of Ca(2+) signaling may represent pro-survival upregulation of Ca(2+)-dependent genomic processes, while decrease in mGluR1 and IP3R IR may be attributed to a feedback mechanism preventing excessive intracellular Ca(2+) release.


Asunto(s)
Presión del Aire , Corteza Cerebral/metabolismo , Hipoxia/metabolismo , Receptor del Glutamato Metabotropico 5/biosíntesis , Receptores de Glutamato Metabotrópico/biosíntesis , Transducción de Señal/genética , Animales , Señalización del Calcio/genética , Receptores de Inositol 1,4,5-Trifosfato/biosíntesis , Receptores de Inositol 1,4,5-Trifosfato/genética , Masculino , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Fosfolipasa C beta/biosíntesis , Fosfolipasa C beta/genética , Corteza Piriforme/metabolismo , Ratas , Ratas Wistar , Receptor del Glutamato Metabotropico 5/agonistas , Receptor del Glutamato Metabotropico 5/genética , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/genética , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA