Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Neurochem ; 166(1): 87-106, 2023 07.
Article in English | MEDLINE | ID: mdl-37328918

ABSTRACT

Ischemic stroke is a leading cause of disability worldwide. There is no simple treatment to alleviate ischemic brain injury, as thrombolytic therapy is applicable within a narrow time window. During the last years, the ketogenic diet (KD) and the exogenous administration of the ketone body ß-hydroxybutyrate (BHB) have been proposed as therapeutic tools for acute neurological disorders and both can reduce ischemic brain injury. However, the mechanisms involved are not completely clear. We have previously shown that the D enantiomer of BHB stimulates the autophagic flux in cultured neurons exposed to glucose deprivation (GD) and in the brain of hypoglycemic rats. Here, we have investigated the effect of the systemic administration of D-BHB, followed by its continuous infusion after middle cerebral artery occlusion (MCAO), on the autophagy-lysosomal pathway and the activation of the unfolded protein response (UPR). Results show for the first time that the protective effect of BHB against MCAO injury is enantiomer selective as only D-BHB, the physiologic enantiomer of BHB, significantly reduced brain injury. D-BHB treatment prevented the cleavage of the lysosomal membrane protein LAMP2 and stimulated the autophagic flux in the ischemic core and the penumbra. In addition, D-BHB notably reduced the activation of the PERK/eIF2α/ATF4 pathway of the UPR and inhibited IRE1α phosphorylation. L-BHB showed no significant effect relative to ischemic animals. In cortical cultures under GD, D-BHB prevented LAMP2 cleavage and decreased lysosomal number. It also abated the activation of the PERK/eIF2α/ATF4 pathway, partially sustained protein synthesis, and reduced pIRE1α. In contrast, L-BHB showed no significant effects. Results suggest that protection elicited by D-BHB treatment post-ischemia prevents lysosomal rupture allowing functional autophagy, preventing the loss of proteostasis and UPR activation.


Subject(s)
Brain Injuries , Stroke , Rats , Animals , Ketone Bodies/pharmacology , Ketone Bodies/metabolism , Endoribonucleases/pharmacology , Protein Serine-Threonine Kinases , Endoplasmic Reticulum Stress , 3-Hydroxybutyric Acid/metabolism , 3-Hydroxybutyric Acid/pharmacology , Glucose/metabolism , Autophagy , Infarction, Middle Cerebral Artery , Models, Theoretical , Stroke/drug therapy
2.
Cells ; 12(3)2023 02 02.
Article in English | MEDLINE | ID: mdl-36766827

ABSTRACT

Mitochondrial activity and quality control are essential for neuronal homeostasis as neurons rely on glucose oxidative metabolism. The ketone body, D-ß-hydroxybutyrate (D-BHB), is metabolized to acetyl-CoA in brain mitochondria and used as an energy fuel alternative to glucose. We have previously reported that D-BHB sustains ATP production and stimulates the autophagic flux under glucose deprivation in neurons; however, the effects of D-BHB on mitochondrial turnover under physiological conditions are still unknown. Sirtuins (SIRTs) are NAD+-activated protein deacetylases involved in the regulation of mitochondrial biogenesis and mitophagy through the activation of transcription factors FOXO1, FOXO3a, TFEB and PGC1α coactivator. Here, we aimed to investigate the effect of D-BHB on mitochondrial turnover in cultured neurons and the mechanisms involved. Results show that D-BHB increased mitochondrial membrane potential and regulated the NAD+/NADH ratio. D-BHB enhanced FOXO1, FOXO3a and PGC1α nuclear levels in an SIRT2-dependent manner and stimulated autophagy, mitophagy and mitochondrial biogenesis. These effects increased neuronal resistance to energy stress. D-BHB also stimulated the autophagic-lysosomal pathway through AMPK activation and TFEB-mediated lysosomal biogenesis. Upregulation of SIRT2, FOXOs, PGC1α and TFEB was confirmed in the brain of ketogenic diet (KD)-treated mice. Altogether, the results identify SIRT2, for the first time, as a target of D-BHB in neurons, which is involved in the regulation of autophagy/mitophagy and mitochondrial quality control.


Subject(s)
NAD , Sirtuin 2 , Animals , Mice , 3-Hydroxybutyric Acid/pharmacology , 3-Hydroxybutyric Acid/metabolism , Autophagy , Glucose/metabolism , Ketone Bodies/metabolism , Ketone Bodies/pharmacology , Lysosomes/metabolism , Mitochondria/metabolism , NAD/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 2/metabolism
3.
Front Aging Neurosci ; 13: 766306, 2021.
Article in English | MEDLINE | ID: mdl-34924995

ABSTRACT

The decline in brain function during aging is one of the most critical health problems nowadays. Although senescent astrocytes have been found in old-age brains and neurodegenerative diseases, their impact on the function of other cerebral cell types is unknown. The aim of this study was to evaluate the effect of senescent astrocytes on the mitochondrial function of a neuron. In order to evaluate neuronal susceptibility to a long and constant senescence-associated secretory phenotype (SASP) exposure, we developed a model by using cellular cocultures in transwell plates. Rat primary cortical astrocytes were seeded in transwell inserts and induced to premature senescence with hydrogen peroxide [stress-induced premature senescence (SIPS)]. Independently, primary rat cortical neurons were seeded at the bottom of transwells. After neuronal 6 days in vitro (DIV), the inserts with SIPS-astrocytes were placed in the chamber and cocultured with neurons for 6 more days. The neuronal viability, the redox state [reduced glutathione/oxidized glutathione (GSH/GSSG)], the mitochondrial morphology, and the proteins and membrane potential were determined. Our results showed that the neuronal mitochondria functionality was altered after being cocultured with senescent astrocytes. In vivo, we found that old animals had diminished mitochondrial oxidative phosphorylation (OXPHOS) proteins, redox state, and senescence markers as compared to young rats, suggesting effects of the senescent astrocytes similar to the ones we observed in vitro. Overall, these results indicate that the microenvironment generated by senescent astrocytes can affect neuronal mitochondria and physiology.

4.
J Am Med Dir Assoc ; 21(12): 1811-1814, 2020 12.
Article in English | MEDLINE | ID: mdl-33256960

ABSTRACT

Older adults living in nursing homes are the most vulnerable group of the COVID-19 pandemic. There are many difficulties in isolating residents and limiting the spread in this setting. We have developed a simple algorithm with a traffic light format for resident classification and sectorization within nursing homes, based on basic diagnostic tests, surveillance of symptoms onset, and close contact monitoring. We have implemented the algorithm in several centers with good data on adherence. Suggestions for implementation and evaluation are discussed.


Subject(s)
Algorithms , COVID-19/prevention & control , Nursing Homes , Humans , Patient Isolation/organization & administration , SARS-CoV-2
5.
Front Cell Neurosci ; 14: 547215, 2020.
Article in English | MEDLINE | ID: mdl-33173467

ABSTRACT

Glucose supply from blood is mandatory for brain functioning and its interruption during acute hypoglycemia or cerebral ischemia leads to brain injury. Alternative substrates to glucose such as the ketone bodies (KB), acetoacetate (AcAc), and ß-hydroxybutyrate (BHB), can be used as energy fuels in the brain during hypoglycemia and prevent neuronal death, but the mechanisms involved are still not well understood. During glucose deprivation adaptive cell responses can be activated such as autophagy, a lysosomal-dependent degradation process, to support cell survival. However, impaired or excessive autophagy can lead to cell dysfunction. We have previously shown that impaired autophagy contributes to neuronal death induced by glucose deprivation in cortical neurons and that D isomer of BHB (D-BHB) reestablishes the autophagic flux increasing viability. Here, we aimed to investigate autophagy dynamics in the brain of rats subjected to severe hypoglycemia (SH) without glucose infusion (GI), severe hypoglycemia followed by GI (SH + GI), and a brief period of hypoglycemic coma followed by GI (Coma). The effect of D-BHB administration after the coma was also tested (Coma + BHB). The transformation of LC3-I to LC3-II and the abundance of autophagy proteins, Beclin 1 (BECN1), ATG7, and ATG12-ATG5 conjugate, were analyzed as an index of autophagosome formation, and the levels of sequestrosome1/p62 (SQSTM1/p62) were determined as a hallmark of autophagic degradation. Data suggest that autophagosomes accumulate in the cortex and the hippocampus of rats after SH, likely due to impaired autophagic degradation. In the cortex, autophagosome accumulation persisted at 6 h after GI in animals exposed to SH but recovered basal levels at 24 h, while in the hippocampus no significant effect was observed. In animals subjected to coma, autophagosome accumulation was observed at 24 h after GI in both regions. D-BHB treatment reduced LC3-II and SQSTM1/p62 content and reduced ULK1 phosphorylation by AMPK, suggesting it stimulates the autophagic flux and decreases AMPK activity reducing autophagy initiation. D-BHB also reduced the number of degenerating cells. Together, data suggest different autophagy dynamics after GI in rats subjected to SH or the hypoglycemic coma and support that D-BHB treatment can modulate autophagy dynamics favoring the autophagic flux.

6.
Curr Pharm Des ; 26(12): 1377-1387, 2020.
Article in English | MEDLINE | ID: mdl-31957603

ABSTRACT

BACKGROUND: The ketone bodies (KB), ß-hydroxybutyrate (BHB) and acetoacetate, have been proposed for the treatment of acute and chronic neurological disorders, however, the molecular mechanisms involved in KB protection are not well understood. KB can substitute for glucose and support mitochondrial metabolism increasing cell survival. We have reported that the D-isomer of BHB (D-BHB) stimulates autophagic degradation during glucose deprivation in cultured neurons increasing cell viability. Autophagy is a lysosomal degradation process of damaged proteins and organelles activated during nutrient deprivation to obtain building blocks and energy. However, impaired or excessive autophagy can contribute to neuronal death. OBJECTIVE: The aim of the present study was to test whether D-BHB can preserve autophagic function in an in vivo model of excitotoxic damage induced by the administration of the glutamate receptor agonist, N-methyl-Daspartate (NMDA), in the rat striatum. METHODS: D-BHB was administered through an intravenous injection followed by either an intraperitoneal injection (i.v+i.p) or a continuous epidural infusion (i.v+pump), or through a continuous infusion of D-BHB alone. Changes in the autophagy proteins ATG7, ATG5, BECLIN 1 (BECN1), LC3, Sequestrosome1/p62 (SQSTM1/ p62) and the lysosomal membrane protein LAMP2, were evaluated by immunoblot. The lesion volume was measured in cresyl violet-stained brain sections. RESULTS: Autophagy is activated early after NMDA injection but autophagic degradation is impaired due to the cleavage of LAMP2. Twenty-four h after NMDA intrastriatal injection, the autophagic flux is re-established, but LAMP2 cleavage is still observed. The administration of D-BHB through the i.v+pump protocol reduced the content of autophagic proteins and the cleavage of LAMP2, suggesting decreased autophagosome formation and lysosomal membrane preservation, improving autophagic degradation. D-BHB also reduced brain injury. The i.v+i.p administration protocol and the infusion of D-BHB alone showed no effect on autophagy activation or degradation.


Subject(s)
Autophagy , N-Methylaspartate , 3-Hydroxybutyric Acid , Animals , Ketone Bodies/chemistry , Neurons/chemistry , Neurons/physiology , Rats
7.
J Neuroinflammation ; 16(1): 91, 2019 Apr 17.
Article in English | MEDLINE | ID: mdl-30995916

ABSTRACT

BACKGROUND: During excitotoxic damage, neuronal death results from the increase in intracellular calcium, the induction of oxidative stress, and a subsequent inflammatory response. NADPH oxidases (NOX) are relevant sources of reactive oxygen species (ROS) during excitotoxic damage. NADPH oxidase-2 (NOX-2) has been particularly related to neuronal damage and death, as well as to the resolution of the subsequent inflammatory response. As ROS are crucial components of the regulation of inflammatory response, in this work, we evaluated the role of NOX-2 in the progression of inflammation resulting from glutamate-induced excitotoxic damage of the striatum in an in vivo model. METHODS: The striata of wild-type C57BL/6 J and NOX-2 KO mice (gp91Cybbtm1Din/J) were stereotactically injected with monosodium glutamate either alone or in combination with IL-4 or IL-10. The damage was evaluated in histological sections stained with cresyl violet and Fluoro-Jade B. The enzymatic activity of caspase-3 and NOX were also measured. Additionally, the cytokine profile was identified by ELISA and motor activity was verified by the tests of the cylinder, the adhesive tape removal, and the inverted grid. RESULTS: Our results show a neuroprotective effect in mice with a genetic inhibition of NOX-2, which is partially due to a differential response to excitotoxic damage, characterized by the production of anti-inflammatory cytokines. In NOX-2 KO animals, the excitotoxic condition increased the production of interleukin-4, which could contribute to the production of interleukin-10 that decreased neuronal apoptotic death and the magnitude of striatal injury. Treatment with interleukin-4 and interleukin-10 protected from excitotoxic damage in wild-type animals. CONCLUSIONS: The release of proinflammatory cytokines during the excitotoxic event promotes an additional apoptotic death of neurons that survived the initial damage. During the subsequent inflammatory response to excitotoxic damage, ROS generated by NOX-2 play a decisive role in the extension of the lesion and consequently in the severity of the functional compromise, probably by regulating the anti-inflammatory cytokines production.


Subject(s)
Corpus Striatum/enzymology , Corpus Striatum/pathology , Inflammation/enzymology , Inflammation/pathology , NADPH Oxidase 2/metabolism , Animals , Corpus Striatum/immunology , Disease Progression , Glutamic Acid/toxicity , Inflammation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
J Cereb Blood Flow Metab ; 39(5): 808-821, 2019 05.
Article in English | MEDLINE | ID: mdl-29047291

ABSTRACT

Moderate recurrent hypoglycemia (RH) is frequent in Type 1 diabetes mellitus (TIDM) patients who are under intensive insulin therapy increasing the risk for severe hypoglycemia (SH). The consequences of RH are not well understood and its repercussions on neuronal damage and cognitive function after a subsequent episode of SH have been poorly investigated. In the current study, we have addressed this question and observed that previous RH during seven consecutive days exacerbated oxidative damage and neuronal death induced by a subsequent episode of SH accompanied by a short period of coma, in the parietal cortex, the striatum and mainly in the hippocampus. These changes correlated with a severe decrease in reduced glutathione content (GSH), and a significant spatial and contextual memory deficit. Administration of the antioxidant, N-acetyl-L-cysteine, (NAC) reduced neuronal death and prevented cognitive impairment. These results demonstrate that previous RH enhances brain vulnerability to acute hypoglycemia and suggests that this effect is mediated by the decline in the antioxidant defense and oxidative damage. The present results highlight the importance of an adequate control of moderate hypoglycemic episodes in TIDM.


Subject(s)
Cognitive Dysfunction/etiology , Coma/complications , Hypoglycemia/complications , Oxidative Stress , Animals , Blood Glucose/metabolism , Cell Death , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Coma/metabolism , Coma/pathology , Glutathione/metabolism , Humans , Hypoglycemia/metabolism , Hypoglycemia/pathology , Male , Neurons/metabolism , Neurons/pathology , Rats, Wistar
9.
Arch Toxicol ; 92(3): 1037-1048, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29204679

ABSTRACT

Early life exposure to environmental pollutants and toxic chemicals has been linked to learning and behavioral alterations in children. iAs exposure is associated with different types neurological disorders such as memory and learning impairment. iAs is methylated in the brain by the arsenic III-methyltransferase in a process that requires glutathione (GSH). The xCT-antiporter cell membrane transporter participates in the influx of cystine for GSH synthesis in exchange for glutamate in a 1:1 ratio. In CD-1 mice gestationally exposed to 20 ppm of sodium arsenite in drinking water, we have previously observed up-regulation of xCT in the male mouse hippocampus which caused glutamatergic synapse alterations affecting learning and memory processes. Here, we used the same gestational iAs exposure model to investigate whether the up-regulation of xCT and down-regulation of GLT-1 transporters were associated with higher levels of extracellular glutamate and changes in the expression of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor, responsible for excitatory fast synaptic transmission. The induction of LTP in the perforant-dentate gyrus pathway (PP-DG) of the hippocampus was also studied, as well as learning and memory formation using the water maze test. Changes in GSH levels were also tested in the hippocampus of animals exposed to iAs. Results showed increased GSH synthesis (p < 0.05), associated with significantly higher extracellular glutamate levels in iAs exposed mice. Exposure was also significantly associated with AMPA subunits down-regulation, deficient LTP induction, and lower excitability of the PP-DG pathway. In addition, animals showed deficient learning and memory in the Morris Water Maze test.


Subject(s)
Arsenic/toxicity , Glutamic Acid/metabolism , Hippocampus/drug effects , Memory Disorders/chemically induced , Prenatal Exposure Delayed Effects , Receptors, Glutamate/metabolism , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Female , Glutathione/metabolism , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Male , Memory Disorders/etiology , Mice, Inbred Strains , Perforant Pathway/drug effects , Pregnancy , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
10.
Cell Death Dis ; 8(6): e2911, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28661473

ABSTRACT

Autophagy is triggered during nutrient and energy deprivation in a variety of cells as a homeostatic response to metabolic stress. In the CNS, deficient autophagy has been implicated in neurodegenerative diseases and ischemic brain injury. However, its role in hypoglycemic damage is poorly understood and the dynamics of autophagy during the hypoglycemic and the glucose reperfusion periods, has not been fully described. In the present study, we analyzed the changes in the content of the autophagy proteins BECN1, LC3-II and p62/SQSTM1 by western blot, and autophagosome formation was followed through time-lapse experiments, during glucose deprivation (GD) and glucose reintroduction (GR) in cortical cultures. According to the results, autophagosome formation rapidly increased during GD, and was followed by an active autophagic flux early after glucose replenishment. However, cells progressively died during GR and autophagy inhibition reduced neuronal death. Neurons undergoing apoptosis during GR did not form autophagosomes, while those surviving up to late GR showed autophagosomes. Calpain activity strongly increased during GR and remained elevated during progressive neuronal death. Its activation led to the cleavage of LAMP2 resulting in lysosome membrane permeabilization (LMP) and release of cathepsin B to the cytosol. Calpain inhibition prevented LMP and increased the number of neurons containing lysosomes and autophagosomes increasing cell viability. Taken together, the present results suggest that calpain-mediated lysosome dysfunction during GR turns an adaptive autophagy response to energy stress into a defective autophagy pathway, which contributes to neuronal death. In these conditions, autophagy inhibition results in the improvement of cell survival.


Subject(s)
Autophagy/genetics , Calpain/genetics , Glucose/metabolism , Neurodegenerative Diseases/genetics , Neurons/metabolism , Animals , Apoptosis/genetics , Autophagosomes/metabolism , Autophagosomes/pathology , Calpain/metabolism , Cell Survival/genetics , Glucose/adverse effects , Humans , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/pathology , Rats
11.
Sci Rep ; 6: 24028, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27063549

ABSTRACT

Parkinson's disease (PD) is a multifactorial neurodegenerative disorder, characterised by the progressive loss of midbrain dopaminergic neurons and a variety of motor symptoms. The gene coding for the phospholipid phosphatase 3, PLPP3 (formerly PPAP2B or LPP3), maps within the PARK10 locus, a region that has been linked with increased risk to late-onset PD. PLPP3 modulates the levels of a range of bioactive lipids controlling fundamental cellular processes within the central nervous system. Here we show that PLPP3 is enriched in astroglial cells of the adult murine ventral midbrain. Conditional inactivation of Plpp3 using a Nestin::Cre driver results in reduced mesencephalic levels of sphingosine-1-phosphate receptor 1 (S1P1), a well-known mediator of pro-survival responses. Yet, adult PLPP3-deficient mice exhibited no alterations in the number of dopaminergic neurons or in the basal levels of striatal extracellular dopamine (DA). Potassium-evoked DA overflow in the striatum, however, was significantly decreased in mutant mice. Locomotor evaluation revealed that, although PLPP3-deficient mice exhibit motor impairment, this is not progressive or responsive to acute L-DOPA therapy. These findings suggest that disruption of Plpp3 during early neural development leads to dopaminergic transmission deficits in the absence of nigrostriatal degeneration, and without causing an age-related locomotor decline consistent with PD.


Subject(s)
Dopaminergic Neurons/metabolism , Parkinson Disease/genetics , Phospholipid Transfer Proteins/metabolism , Animals , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/pathology , Female , Genetic Loci , Locomotion/physiology , Mesencephalon/metabolism , Mesencephalon/pathology , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phospholipid Transfer Proteins/deficiency , Phospholipid Transfer Proteins/genetics , Proprotein Convertases/metabolism , Serine Endopeptidases/metabolism , Signal Transduction
12.
Neurochem Res ; 41(3): 600-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26303508

ABSTRACT

Glucose is the major energy substrate in brain, however, during ketogenesis induced by starvation or prolonged hypoglycemia, the ketone bodies (KB), acetoacetate and ß-hydroxybutyrate (BHB) can substitute for glucose. KB improve neuronal survival in diverse injury models, but the mechanisms by which KB prevent neuronal damage are still not well understood. In the present study we have investigated whether protection by the D isomer of BHB (D-BHB) against neuronal death induced by glucose deprivation (GD), is related to autophagy. Autophagy is a lysosomal-dependent degradation process activated during nutritional stress, which leads to the digestion of damaged proteins and organelles providing energy for cell survival. Results show that autophagy is activated in cortical cultured neurons during GD, as indicated by the increase in the levels of the lipidated form of the microtubule associated protein light chain 3 (LC3-II), and the number of autophagic vesicles. At early phases of glucose reintroduction (GR), the levels of p62 declined suggesting that the degradation of the autophagolysosomal content takes place at this time. In cultures exposed to GD and GR in the presence of D-BHB, the levels of LC3-II and p62 rapidly declined and remained low during GR, suggesting that the KB stimulates the autophagic flux preventing autophagosome accumulation and improving neuronal survival.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Autophagy , Cerebral Cortex/cytology , Glucose/metabolism , Neurons/cytology , 3-Hydroxybutyric Acid/chemistry , 3-Hydroxybutyric Acid/pharmacology , Animals , Cell Survival , Cells, Cultured , Cerebral Cortex/metabolism , Heat-Shock Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Rats, Wistar , Sequestosome-1 Protein , Stereoisomerism
13.
Brain Res ; 1627: 80-9, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26410780

ABSTRACT

Endoplasmic reticulum (ER) and mitochondria dysfunction contribute to insulin resistance generation during obesity and diabetes. ER and mitochondria interact through Mitofusin 2 (MTF2), which anchors in the outer mitochondrial and ER membranes regulating energy metabolism. Ablation of MTF2 leads to ER stress activation and insulin resistance. Here we determine whether lipotoxic insult induced by saturated lipids decreases MTF2 expression leading to ER stress response in hypothalamus and its effects on insulin sensitivity using in vitro and in vivo models. We found that lipotoxic stimulation induced by palmitic acid, but not the monounsaturated palmitoleic acid, decreases MTF2 protein levels in hypothalamic mHypoA-CLU192 cells. Also, palmitic acid incubation activates ER stress response evidenced by increase in the protein levels of GRP78/BIP marker at later stage than MTF2 downregulation. Additionally, we found that MTF2 alterations induced by palmitic, but not palmitoleic, stimulation exacerbate insulin resistance in hypothalamic cells. Insulin resistance induced by palmitic acid is prevented by pre-incubation of the anti-inflammatory and the ER stress release reagents, sodium salicylate and 4 phenylbutirate, respectively. Finally, we demonstrated that lipotoxic insult induced by high fat feeding to mice decreases MTF2 proteins levels in arcuate nucleus of hypothalamus. Our data indicate that saturated lipids modulate MTF2 expression in hypothalamus coordinating the ER stress response and the susceptibility to insulin resistance.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/pharmacology , Hypothalamus/cytology , Insulin Resistance/physiology , Neurons/drug effects , Palmitic Acid/pharmacology , Polycomb Repressive Complex 2/metabolism , Analysis of Variance , Animals , Body Weight/drug effects , Body Weight/physiology , Diet, High-Fat , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/ultrastructure , Oncogene Protein v-akt/metabolism , Polycomb Repressive Complex 2/genetics , Reactive Oxygen Species/metabolism , Time Factors
14.
J Cereb Blood Flow Metab ; 35(5): 851-60, 2015 May.
Article in English | MEDLINE | ID: mdl-25649993

ABSTRACT

Glucose is the main energy substrate in brain but in certain circumstances such as prolonged fasting and the suckling period alternative substrates can be used such as the ketone bodies (KB), beta-hydroxybutyrate (BHB), and acetoacetate. It has been shown that KB prevent neuronal death induced during energy limiting conditions and excitotoxicity. The protective effect of KB has been mainly attributed to the improvement of mitochondrial function. In the present study, we have investigated the protective effect of D-BHB against neuronal death induced by severe noncoma hypoglycemia in the rat in vivo and by glucose deprivation (GD) in cortical cultures. Results show that systemic administration of D-BHB reduces reactive oxygen species (ROS) production in distinct cortical areas and subregions of the hippocampus and efficiently prevents neuronal death in the cortex of hypoglycemic animals. In vitro results show that D-BHB stimulates ATP production and reduces ROS levels, while the nonphysiologic isomer of BHB, L-BHB, has no effect on energy production but reduces ROS levels. Data suggest that protection by BHB, not only results from its metabolic action but is also related to its capability to reduce ROS, rendering this KB as a suitable candidate for the treatment of ischemic and traumatic injury.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Cerebral Cortex , Energy Metabolism/drug effects , Hypoglycemia , Neurons , Reactive Oxygen Species/metabolism , Acetoacetates/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Glucose/metabolism , Hypoglycemia/drug therapy , Hypoglycemia/metabolism , Hypoglycemia/pathology , Ketone Bodies/metabolism , Male , Neurons/metabolism , Neurons/pathology , Rats , Rats, Wistar
15.
Metab Brain Dis ; 29(3): 711-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24590689

ABSTRACT

Hypoglycemia is a serious complication of insulin therapy in patients suffering from type 1 Diabetes Mellitus. Severe hypoglycemia leading to coma (isoelectricity) induces massive neuronal death in vulnerable brain regions such as the hippocampus, the striatum and the cerebral cortex. It has been suggested that the production of reactive oxygen species (ROS) and oxidative stress is involved in hypoglycemic brain damage, and that ROS generation is stimulated by glucose reintroduction (GR) after the hypoglycemic coma. However, the distribution of ROS in discrete brain regions has not been studied in detail. Using the oxidation sensitive marker dihydroethidium (DHE) we have investigated the distribution of ROS in different regions of the mouse brain during prolonged severe hypoglycemia without isoelectricity, as well as the effect of GR on ROS levels. Results show that ROS generation increases in the hippocampus, the cerebral cortex and the striatum after prolonged severe hypoglycemia before the coma. The hippocampus showed the largest increases in ROS levels. GR further stimulated ROS production in the hippocampus and the striatum while in the cerebral cortex, only the somatosensory and parietal areas were significantly affected by GR. Results suggest that ROS are differentially produced during the hypoglycemic insult and that a different response to GR is present among distinct brain regions.


Subject(s)
Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Hippocampus/metabolism , Hypoglycemia/metabolism , Reactive Oxygen Species/metabolism , Animals , Male , Mice , Organ Specificity , Oxidative Stress/physiology
16.
Int J Biochem Cell Biol ; 45(11): 2596-604, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23994487

ABSTRACT

The mechanisms leading to neuronal death during glucose deprivation have not been fully elucidated, but a role of oxidative stress has been suggested. In the present study we have investigated whether the production of reactive oxygen species during glucose deprivation, contributes to the activation of calpain, a calcium-dependent protease involved in neuronal injury associated with brain ischemia and cerebral trauma. We have observed a rapid activation of calpain, as monitored by the cleavage of the cytoskeletal protein α-spectrin, after glucose withdrawal, which is reduced by inhibitors of xanthine oxidase, phospholipase A2 and NADPH oxidase. Results suggest that phospholipase A2 and NADPH oxidase contribute to the early activation of calpain after glucose deprivation. In particular NOX2, a member of the NADPH oxidase family is involved, since reduced stimulation of calpain activity is observed after glucose deprivation in hippocampal slices from transgenic mice lacking a functional NOX2. We observed an additive effect of the inhibitors of xanthine oxidase and phospholipase A2 on both ROS production and calpain activity, suggesting a synergistic action of these two enzymes. The present results provide new evidence showing that reactive oxygen species stimulate calpain activation during glucose deprivation and that this mechanism is involved in neuronal death.


Subject(s)
Calpain/metabolism , Glucose/deficiency , Neurons/enzymology , Neurons/pathology , Oxidative Stress , Animals , Cell Death/drug effects , Enzyme Activation/drug effects , Glucose/pharmacology , Hippocampus/pathology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Mice , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Phospholipases A2/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/metabolism
17.
Neurochem Int ; 63(4): 331-43, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23876631

ABSTRACT

The aim of the present review is to offer a current perspective about the consequences of hypoglycemia and its impact on the diabetic disorder due to the increasing incidence of diabetes around the world. The main consequence of insulin treatment in type 1 diabetic patients is the occurrence of repetitive periods of hypoglycemia and even episodes of severe hypoglycemia leading to coma. In the latter, selective neuronal death is observed in brain vulnerable regions both in humans and animal models, such as the cortex and the hippocampus. Cognitive damage subsequent to hypoglycemic coma has been associated with neuronal death in the hippocampus. The mechanisms implicated in selective damage are not completely understood but many factors have been identified including excitotoxicity, oxidative stress, zinc release, PARP-1 activation and mitochondrial dysfunction. Importantly, the diabetic condition aggravates neuronal damage and cognitive failure induced by hypoglycemia. In the absence of coma prolonged and severe hypoglycemia leads to increased oxidative stress and discrete neuronal death mainly in the cerebral cortex. The mechanisms responsible for cell damage in this condition are still unknown. Recurrent moderate hypoglycemia is far more common in diabetic patients than severe hypoglycemia and currently important efforts are being done in order to elucidate the relationship between cognitive deficits and recurrent hypoglycemia in diabetics. Human studies suggest impaired performance mainly in memory and attention tasks in healthy and diabetic individuals under the hypoglycemic condition. Only scarce neuronal death has been observed under moderate repetitive hypoglycemia but studies suggest that impaired hippocampal synaptic function might be one of the causes of cognitive failure. Recent studies have also implicated altered mitochondrial function and mitochondrial oxidative stress.


Subject(s)
Cognition Disorders/complications , Hypoglycemia/complications , Neurons/pathology , Animals , Cognition Disorders/pathology , Humans , Hypoglycemia/pathology , Hypoglycemia/prevention & control
18.
Exp Neurol ; 233(1): 182-92, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21985864

ABSTRACT

Manganese is essential for life, yet chronic exposure to this metal can cause a neurodegenerative disease named manganism that affects motor function. In the present study we have evaluated Mn neurotoxicity after its administration in the rat striatum. The participation of the calcium-dependent protease calpain and the apoptosis-related protease caspase-3, in Mn-induced cell death was monitored in the striatum and globus pallidus. Mn induced the activation of both proteases, although calpain activation seems to be an earlier event. Moreover, while the broad-spectrum caspase inhibitor QVD did not significantly prevent Mn-induced cell death, the specific calpain inhibitor MDL-28170 did. The role of NMDA glutamate receptors on calpain activity was also investigated; blockage of these receptors by MK-801 and memantine did not prevent calpain activation, nor Mn-induced cell death. Finally, studies in striatal homogenates suggest a direct activation of calpain by Mn ions. Altogether the present study suggests that additional mechanisms to excitotoxicity are involved in Mn-induced cell death, placing calpain as an important mediator of acute Mn neurotoxicity in vivo.


Subject(s)
Calpain/metabolism , Corpus Striatum/metabolism , Manganese Poisoning/pathology , Manganese/toxicity , Analysis of Variance , Animals , Caspase 3 , Cell Death/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Excitatory Amino Acid Antagonists/pharmacology , Fluoresceins , Globus Pallidus/metabolism , Globus Pallidus/pathology , Male , Manganese Poisoning/drug therapy , Manganese Poisoning/etiology , Memantine/pharmacology , Memantine/therapeutic use , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Organic Chemicals , Rats , Rats, Wistar , Time Factors
19.
J Neuropathol Exp Neurol ; 70(11): 1020-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22002428

ABSTRACT

Prolonged activation of glutamate receptors leads to excitotoxicity. Several processes such as reactive oxygen species (ROS) production and activation of the calcium-dependent protease, calpain, contribute to glutamate-induced damage. It has been suggested that the ROS-producing enzyme, NADPH oxidase (NOX), plays a role in excitotoxicity. Studies have reported NOX activation after NMDA receptor stimulation during excitotoxic damage, but the role of non-NMDA and metabotropic receptors is unknown. We evaluated the roles of different glutamate receptor subtypes on NOX activation and neuronal death induced by the intrastriatal administration of glutamate in mice. In wild-type mice, NOX2 immunoreactivity in neurons and microglia was stimulated by glutamate administration, and it progressively increased as microglia became activated; calpain activity was also induced. By contrast, mice lacking NOX2 were less vulnerable to excitotoxicity, and there was reduced ROS production and protein nitrosylation, microglial reactivity, and calpain activation. These results suggest that NOX2 is stimulated by glutamate in neurons and reactive microglia through the activation of ionotropic and metabotropic receptors. Neuronal damage involves ROS production by NOX2, which, in turn, contributes to calpain activation.


Subject(s)
Calpain/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Neurotoxicity Syndromes/metabolism , Reactive Oxygen Species/metabolism , Receptors, Ionotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Calcium-Binding Proteins/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Drug Interactions , Excitatory Amino Acid Agents/pharmacology , Fluoresceins , Gene Expression Regulation/drug effects , Glutamic Acid/toxicity , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , NADPH Oxidase 2 , NADPH Oxidases/deficiency , Neurons/drug effects , Neurons/metabolism , Neurotoxicity Syndromes/etiology , Organic Chemicals , Phosphopyruvate Hydratase/metabolism , Receptors, Immunologic/metabolism , Time Factors
20.
Neurochem Res ; 35(8): 1156-63, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20401690

ABSTRACT

Excitotoxicity has been associated with the loss of medium spiny neurons (MSN) in Huntington's disease (HD). We have previously observed that the content of the glial glutamate transporters, glutamate transporter 1 (GLT-1) and glutamate-aspartate transporter (GLAST), diminishes in R6/2 mice at 14 weeks of age but not at 10 weeks, and that this change correlates with an increased vulnerability of striatal neurons to glutamate toxicity. We have also reported that inhibition of the glycolytic pathway decreases glutamate uptake and enhances glutamate neurotoxicity in the rat brain. We now show that at 10-weeks of age, glutamate excitotoxicity is precipitated in R6/2 mice, after the treatment with iodoacetate (IOA), an inhibitor of the glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase (GAPDH). IOA induces a larger inhibition of GAPDH in R6/2 mice, while it similarly reduces the levels of GLT-1 and GLAST in wild-type and transgenic animals. Results suggest that metabolic failure and altered glutamate uptake are involved in the vulnerability of striatal neurons to glutamate excitotoxicity in HD.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Glutamic Acid/metabolism , Huntington Disease/metabolism , Animals , Brain/drug effects , Brain/metabolism , Female , Glutamic Acid/toxicity , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/antagonists & inhibitors , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis , Huntington Disease/genetics , Iodoacetates/pharmacology , Mice , Mice, Inbred BALB C , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...