Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Alzheimers Dement ; 18(2): 318-338, 2022 02.
Article in English | MEDLINE | ID: mdl-34057756

ABSTRACT

INTRODUCTION: The second most common form of early-onset dementia-frontotemporal dementia (FTD)-is often characterized by the aggregation of the microtubule-associated protein tau. Here we studied the mechanism of tau-induced neuronal dysfunction in neurons with the FTD-related 10+16 MAPT mutation. METHODS: Live imaging, electrophysiology, and redox proteomics were used in 10+16 induced pluripotent stem cell-derived neurons and a model of tau spreading in primary cultures. RESULTS: Overproduction of mitochondrial reactive oxygen species (ROS) in 10+16 neurons alters the trafficking of specific glutamate receptor subunits via redox regulation. Increased surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptors containing GluA1 and NR2B subunits leads to impaired glutamatergic signaling, calcium overload, and excitotoxicity. Mitochondrial antioxidants restore the altered response and prevent neuronal death. Importantly, extracellular 4R tau induces the same pathological response in healthy neurons, thus proposing a mechanism for disease propagation. DISCUSSION: These results demonstrate mitochondrial ROS modulate glutamatergic signaling in FTD, and suggest a new therapeutic strategy.


Subject(s)
Frontotemporal Dementia , Induced Pluripotent Stem Cells , Frontotemporal Dementia/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mitochondria , Neurons/metabolism , Reactive Oxygen Species/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , tau Proteins/metabolism
2.
Cell Calcium ; 91: 102268, 2020 11.
Article in English | MEDLINE | ID: mdl-32827867

ABSTRACT

Increasing evidence suggests that metabolic dysfunctions are at the roots of neurodegenerative disorders such as Alzheimer's disease (AD). In particular, defects in cerebral glucose metabolism, which have been often noted even before the occurrence of clinical symptoms and histopathological lesions, are now regarded as critical contributors to the pathogenesis of AD. Hence, the stimulation of energy metabolism, by enhancing the availability of specific metabolites, might be an alternative way to improve ATP synthesis and to positively affect AD progression. For instance, glutamate may serve as an intermediary metabolite for ATP synthesis through the tricarboxylic acid (TCA) cycle and the oxidative phosphorylation. We have recently shown that two transporters are critical for the anaplerotic use of glutamate: the Na+-dependent Excitatory Amino Acids Carrier 1 (EAAC1) and the Na+-Ca2+ exchanger 1 (NCX1). Therefore, in the present study, we established an AD-like phenotype by perturbing glucose metabolism in both primary rat cortical neurons and retinoic acid (RA)-differentiated SH-SY5Y cells, and we explored the potential of glutamate to halt cell damage by monitoring neurotoxicity, AD markers, ATP synthesis, cytosolic Ca2+ levels and EAAC1/NCX1 functional activities. We found that glutamate significantly increased ATP production and cell survival, reduced the increase of AD biomarkers (amyloid ß protein and the hyperphosphorylated form of tau protein), and recovered the increase of NCX reverse-mode activity. The RNA silencing of either EAAC1 or NCX1 caused the loss of the beneficial effects of glutamate, suggesting the requirement of a functional interplay between these transporters for glutamate-induced protection. Remarkably, our results indicate, as proof-of-principle, that facilitating the use of alternative fuels, like glutamate, may be an effective approach to overcome deficits in glucose utilization and significantly slow down neuronal degenerative process in AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Protective Agents/metabolism , Sodium-Calcium Exchanger/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cerebral Cortex/pathology , Glyceraldehyde , Humans , Models, Biological , Neurons/metabolism , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Tretinoin/pharmacology
3.
J Neurosci ; 39(31): 6038-6048, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31147524

ABSTRACT

Glutamate is one of the most important neurotransmitters in the process of signal transduction in the CNS. Excessive amounts of this neurotransmitter lead to glutamate excitotoxicity, which is accountable for neuronal death in acute neurological disorders, including stroke and trauma, and in neurodegenerative diseases. Inorganic polyphosphate (PolyP) plays multiple roles in the mammalian brain, including function as a calcium-dependent gliotransmitter mediating communication between astrocytes, while its role in the regulation of neuronal activity is unknown. Here we studied the effect of PolyP on glutamate-induced calcium signal in primary rat neurons in both physiological and pathological conditions. We found that preincubation of primary neurons with PolyP reduced glutamate-induced and AMPA-induced but not the NMDA-induced calcium signal. However, in rat hippocampal acute slices, PolyP reduced ion flux through NMDA and AMPA receptors in native neurons. The effect of PolyP on glutamate and specifically on the AMPA receptors was dependent on the presence of P2Y1 but not of P2X receptor inhibitors and also could be mimicked by P2Y1 agonist 2MeSADP. Preincubation of cortical neurons with PolyP significantly reduced the initial calcium peak as well as the number of neurons with delayed calcium deregulation in response to high concentrations of glutamate and resulted in protection of neurons against glutamate-induced cell death. As a result, activation of P2Y1 receptors by PolyP reduced calcium signal acting through AMPA receptors, thus protecting neurons against glutamate excitotoxicity by reduction of the calcium overload and restoration of mitochondrial function.SIGNIFICANCE STATEMENT One of the oldest polymers in the evolution of living matter is the inorganic polyphosphate (PolyP). It is shown to play a role of gliotransmitter in the brain; however, the role of polyphosphate in neuronal signaling is not clear. Here we demonstrate that inorganic polyphosphate is able to reduce calcium signaling induced by physiological or high concentrations of glutamate. The effect of polyphosphate on glutamate-induced calcium signal in neurons is due to the effect of this polymer on the AMPA receptors. The effect of PolyP on glutamate-induced and AMPA-induced calcium signal is dependent on P2Y receptor antagonist. The ability of PolyP to restrict the glutamate-induced calcium signal lies in the basis of its protection of neurons against glutamate excitotoxicity.


Subject(s)
Glutamic Acid/metabolism , Neurons/metabolism , Polyphosphates/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Purinergic P2Y1/metabolism , Animals , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Female , Glutamic Acid/toxicity , Male , Neurons/drug effects , Polyphosphates/pharmacology , Rats , Rats, Sprague-Dawley
4.
Eur J Pharmacol ; 850: 158-166, 2019 May 05.
Article in English | MEDLINE | ID: mdl-30721704

ABSTRACT

Ticagrelor is a direct acting and reversibly binding P2Y12 antagonist approved for the prevention of thromboembolic events. Clinical effects of ticagrelor cannot be simply accounted for by pure platelet inhibition, and off-target mechanisms can potentially play a role. In particular, recent evidence suggests that ticagrelor may also influence heart function and improve the evolution of myocardial ischemic injury by more direct effects on myocytes. The cardiac sodium/calcium exchanger 1 (NCX1) is a critical player in the generation and control of calcium (Ca2+) signals, which orchestrate multiple myocyte activities in health and disease. Altered expression and/or activity of NCX1 can have profound consequences for the function and fate of myocytes. Whether ticagrelor affects cardiac NCX1 has not been investigated yet. To explore this hypothesis, we analyzed the expression, localization and activity of NCX1 in the heart derived H9c2-NCX1 cells following ticagrelor exposure. We found that ticagrelor concentration- and time-dependently reduced the activity of the cardiac NCX1 in H9c2 cells. In particular, the inhibitory effect of ticagrelor on the Ca2+-influx mode of NCX1 was evident within 1 h and further developed after 24 h, when NCX1 activity was suppressed by about 55% in cells treated with 1 µM ticagrelor. Ticagrelor-induced inhibition of exchanger activity was reached at clinically relevant concentrations, without affecting the expression levels and subcellular distribution of NCX1. Collectively, these findings suggest that cardiac NCX1 is a new downstream target of ticagrelor, which may contribute to the therapeutic profile of ticagrelor in clinical practice.


Subject(s)
Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors , Ticagrelor/pharmacology , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Protein Transport/drug effects , Sodium-Calcium Exchanger/metabolism
5.
Front Cell Neurosci ; 12: 264, 2018.
Article in English | MEDLINE | ID: mdl-30333728

ABSTRACT

Friedreich's Ataxia (FRDA) is a neurodegenerative disorder, characterized by degeneration of dorsal root ganglia, cerebellum and cardiomyopathy. Heart failure is one of the most common causes of death for FRDA patients. Deficiency of frataxin, a small mitochondrial protein, is responsible for all clinical and morphological manifestations of FRDA. The focus of our study was to investigate the unexplored Ca2+ homeostasis in cerebellar granule neurons (CGNs) and in cardiomyocytes of FRDA cellular models to understand the pathogenesis of degeneration. Ca2+ homeostasis in neurons and cardiomyocytes is not only crucial for the cellular wellbeing but more importantly to generate action potential in both neurons and cardiomyocytes. By challenging Ca2+ homeostasis in CGNs, and in adult and neonatal cardiomyocytes of FRDA models, we have assessed the impact of frataxin decrease on both neuronal and cardiac physiopathology. Interestingly, we have found that Ca2+ homeostasis is altered both cell types. CGNs showed a Ca2+ mishandling under depolarizing conditions and this was also reflected in the endoplasmic reticulum (ER) content. In cardiomyocytes we found that the sarcoplasmic reticulum (SR) Ca2+ content was pathologically reduced, and that mitochondrial Ca2+ uptake was impaired. This phenomenon is due to the excess of oxidative stress under FRDA like conditions and the consequent aberrant modulation of key players at the SR/ER and mitochondrial level that usually restore the Ca2+ homeostasis. Our findings demonstrate that in both neurons and cardiomyocytes the decreased Ca2+ level within the stores has a comparable detrimental impact in their physiology. In cardiomyocytes, we found that ryanodine receptors (RyRs) may be leaking and expel more Ca2+ out from the SR. At the same time mitochondrial uptake was altered and we found that Vitamin E can restore this defect. Moreover, Vitamin E protects from cell death induced by hypoxia-reperfusion injury, revealing novel properties of Vitamin E as potential therapeutic tool for FRDA cardiomyopathy.

6.
Sci Rep ; 7(1): 13073, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026150

ABSTRACT

Myocardial ischemia culminates in ATP production impairment, ionic derangement and cell death. The provision of metabolic substrates during reperfusion significantly increases heart tolerance to ischemia by improving mitochondrial performance. Under normoxia, glutamate contributes to myocardial energy balance as substrate for anaplerotic reactions, and we demonstrated that the Na+/Ca2+ exchanger1 (NCX1) provides functional support for both glutamate uptake and use for ATP synthesis. Here we investigated the role of NCX1 in the potential of glutamate to improve energy metabolism and survival of cardiac cells subjected to hypoxia/reoxygenation (H/R). Specifically, in H9c2-NCX1 myoblasts, ATP levels, mitochondrial activities and cell survival were significantly compromised after H/R challenge. Glutamate supplementation at the onset of the reoxygenation phase significantly promoted viability, improved mitochondrial functions and normalized the H/R-induced increase of NCX1 reverse-mode activity. The benefits of glutamate were strikingly lost in H9c2-WT (lacking NCX1 expression), or in H9c2-NCX1 and rat cardiomyocytes treated with either NCX or Excitatory Amino Acid Transporters (EAATs) blockers, suggesting that a functional interplay between these transporters is critically required for glutamate-induced protection. Collectively, these results revealed for the first time the key role of NCX1 for the beneficial effects of glutamate against H/R-induced cell injury.


Subject(s)
Cell Survival/drug effects , Glutamic Acid/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Blotting, Western , Cell Hypoxia/drug effects , Male , Rats , Rats, Wistar
7.
J Biomed Sci ; 24(1): 56, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28810874

ABSTRACT

Sudden cardiac death (SCD) describes a natural and unexpected death from cardiac causes occurring within a short period of time (generally within 1 h of symptom onset) in the absence of any other potentially lethal condition. Most SCD-related diseases have a genetic basis; in particular congenital cardiac channelopathies and cardiomyopathies have been described as leading causes of SCD. Congenital cardiac channelopathies are primary electric disorders caused by mutations affecting genes encoding cardiac ion channels or associated proteins, whereas cardiomyopathies are related to mutations in genes encoding several categories of proteins, including those of sarcomeres, desmosomes, the cytoskeleton, and the nuclear envelope. The purpose of this review is to provide a general overview of the main genetic variants that have been linked to the major congenital cardiac channelopathies and cardiomyopathies. Functional alterations of the related proteins are also described.


Subject(s)
Caveolin 3/genetics , Death, Sudden, Cardiac/etiology , Genetic Variation , Cardiomyopathies/complications , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Caveolin 3/metabolism , Channelopathies/complications , Channelopathies/congenital , Channelopathies/genetics , Channelopathies/physiopathology , Humans , Mutation
8.
Eur J Pharmacol ; 794: 246-256, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27894810

ABSTRACT

Ca2+-handling disturbances play an important role in the genesis of myocardial ischemia/reperfusion (I/R) injury. Ischemic preconditioning (IPC) is a powerful strategy to induce tolerance against subsequent ischemic episodes. IPC signaling pathways may be triggered by Ca2+ ion. Since Na+/Ca2+ exchanger 1 (NCX1) participates in modulating intracellular Ca2+ homeostasis, here we further defined its role in I/R and investigated its potential involvement in IPC-induced cardioprotection. In isolated ventricular cardiomyocytes, perfused rat heart and H9c2 cardiomyoblasts, I/R produced a significant cell injury, assessed by measuring extracellular lactate dehydrogenase (LDH) and, for the whole heart, also by estimating myocardial infarct size area. Characterization of cell death revealed the involvement of apoptotic processes. Interestingly, I/R challenge induced NCX1 protein upregulation. In NCX1-transfected H9c2 cells, exchanger protein upregulation was accompanied by an increase in its reverse mode activity. The effects of I/R on extracellular LDH and infarct size area were drastically reduced by 1µM SN-6, a selective NCX1 inhibitor. Moreover, SN-6 also prevented I/R-induced increase of NCX1 reverse-mode activity and protein upregulation. These results suggested a deleterious role of NCX1 in I/R-induced cell damage. In both isolated cardiomyocytes and perfused heart, IPC followed by I/R afforded cardioprotection, reducing extracellular LDH release and limiting ischemic area extent. Interestingly, NCX1 blockade (1µM SN-6) completely abolished IPC protection against I/R, leading to exacerbation of cell injury, massive infarct size area and restoration of NCX1 protein expression. These findings suggest that NCX1 is deleterious in I/R, whereas it may be beneficial in promoting IPC-induced cardioprotection.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/therapy , Myocytes, Cardiac/drug effects , Sodium-Calcium Exchanger/antagonists & inhibitors , Animals , Benzyl Compounds/pharmacology , Cell Death/drug effects , Cell Line , Extracellular Space/metabolism , L-Lactate Dehydrogenase/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , Thiazolidines/pharmacology
9.
Biomed Res Int ; 2016: 6701324, 2016.
Article in English | MEDLINE | ID: mdl-27340665

ABSTRACT

Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by progressive neuronal loss. AD is associated with aberrant processing of the amyloid precursor protein, which leads to the deposition of amyloid-ß plaques within the brain. Together with plaques deposition, the hyperphosphorylation of the microtubules associated protein tau and the formation of intraneuronal neurofibrillary tangles are a typical neuropathological feature in AD brains. Cellular dysfunctions involving specific subcellular compartments, such as mitochondria and endoplasmic reticulum (ER), are emerging as crucial players in the pathogenesis of AD, as well as increased oxidative stress and dysregulation of calcium homeostasis. Specifically, dysregulation of intracellular calcium homeostasis has been suggested as a common proximal cause of neural dysfunction in AD. Aberrant calcium signaling has been considered a phenomenon mainly related to the dysfunction of intracellular calcium stores, which can occur in both neuronal and nonneuronal cells. This review reports the most recent findings on cellular mechanisms involved in the pathogenesis of AD, with main focus on the control of calcium homeostasis at both cytosolic and mitochondrial level.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Calcium Signaling/genetics , Calcium/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Neurons/metabolism , Neurons/pathology , Oxidative Stress/genetics , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...