Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Int J Mol Sci ; 25(7)2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38612861

ABSTRACT

Alzheimer's disease (AD), the most common neurodegenerative disease (NDD), is characterized by chronic neuronal cell death through progressive loss of cognitive function. Amyloid beta (Aß) deposition, neuroinflammation, oxidative stress, and hyperphosphorylated tau proteins are considered the hallmarks of AD pathology. Different therapeutic approaches approved by the Food and Drug Administration can only target a single altered pathway instead of various mechanisms that are involved in AD pathology, resulting in limited symptomatic relief and almost no effect in slowing down the disease progression. Growing evidence on modulating the components of the endocannabinoid system (ECS) proclaimed their neuroprotective effects by reducing neurochemical alterations and preventing cellular dysfunction. Recent studies on AD mouse models have reported that the inhibitors of the fatty acid amide hydrolase (FAAH) and monoacylglycerol (MAGL), hydrolytic enzymes for N-arachidonoyl ethanolamine (AEA) and 2-arachidonoylglycerol (2-AG), respectively, might be promising candidates as therapeutical intervention. The FAAH and MAGL inhibitors alone or in combination seem to produce neuroprotection by reversing cognitive deficits along with Aß-induced neuroinflammation, oxidative responses, and neuronal death, delaying AD progression. Their exact signaling mechanisms need to be elucidated for understanding the brain intrinsic repair mechanism. The aim of this review was to shed light on physiology and pathophysiology of AD and to summarize the experimental data on neuroprotective roles of FAAH and MAGL inhibitors. In this review, we have also included CB1R and CB2R modulators with their diverse roles to modulate ECS mediated responses such as anti-nociceptive, anxiolytic, and anti-inflammatory actions in AD. Future research would provide the directions in understanding the molecular mechanisms and development of new therapeutic interventions for the treatment of AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , United States , Animals , Mice , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Endocannabinoids , Neuroinflammatory Diseases
2.
Diagnostics (Basel) ; 14(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38201409

ABSTRACT

The colloid cyst is a non-malignant tumor growth made of a gelatinous material covered by a membrane of epithelial tissue. It is usually located posterior to the foramen of Monro, in the anterior aspect of the third ventricle of the brain. Due to its location, it can cause obstructive hydrocephalus, increased intracranial pressure, and sudden cardiac death, catecholamine-mediated, through hypothalamus compression. All the mechanisms are still controversial, but the role of catecholamine has been confirmed with histological findings that highlighted myocardial injury (coagulative myocytolysis and contraction band necrosis, CBN). This study presents a case of sudden death in a previously healthy 22-year-old male due to a colloid cyst of the third ventricle. A complete autopsy was performed, highlighting in the brain an abundant quantity of cerebrospinal fluid (CSF) and a 2 cm pale grayish-green rounded cyst formation partially filling and distending the third ventricle. The diagnosis was confirmed through immunohistochemical investigation: positivity for Periodic acid-Schiff (PAS) staining and CK7 expression. In cases such as the one reported here, a combined approach of autopsy, histology, and immunohistochemistry is mandatory in order to identify the neoformation's location and morpho-structural characteristics for a correct differential diagnosis, as well as to identify the cause of death.

3.
Cells ; 9(9)2020 09 06.
Article in English | MEDLINE | ID: mdl-32899900

ABSTRACT

Increasing evidence suggests that metabolic alterations may be etiologically linked to neurodegenerative disorders such as Parkinson's disease (PD) and in particular empathizes the possibility of targeting mitochondrial dysfunctions to improve PD progression. Under different pathological conditions (i.e., cardiac and neuronal ischemia/reperfusion injury), we showed that supplementation of energetic substrates like glutamate exerts a protective role by preserving mitochondrial functions and enhancing ATP synthesis through a mechanism involving the Na+-dependent excitatory amino acid transporters (EAATs) and the Na+/Ca2+ exchanger (NCX). In this study, we investigated whether a similar approach aimed at promoting glutamate metabolism would be also beneficial against cell damage in an in vitro PD-like model. In retinoic acid (RA)-differentiated SH-SY5Y cells challenged with α-synuclein (α-syn) plus rotenone (Rot), glutamate significantly improved cell viability by increasing ATP levels, reducing oxidative damage and cytosolic and mitochondrial Ca2+ overload. Glutamate benefits were strikingly lost when either EAAT3 or NCX1 expression was knocked down by RNA silencing. Overall, our results open the possibility of targeting EAAT3/NCX1 functions to limit PD pathology by simultaneously favoring glutamate uptake and metabolic use in dopaminergic neurons.


Subject(s)
Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Parkinson Disease/genetics , Sodium-Calcium Exchanger/metabolism , Cell Line, Tumor , Humans , Neuroprotection , Parkinson Disease/metabolism , Transfection
4.
Cell Calcium ; 86: 102160, 2020 03.
Article in English | MEDLINE | ID: mdl-31962228

ABSTRACT

Energy metabolism impairment is a central event in the pathophysiology of ischemia. The limited availability of glucose and oxygen strongly affects mitochondrial activity, thus leading to ATP depletion. In this setting, the switch to alternative energy sources could ameliorate cells survival by enhancing ATP production, thus representing an attractive strategy for ischemic treatment. In this regard, some studies have recently re-evaluated the metabolic role of glutamate and its potential to promote cell survival under pathological conditions. In the present review, we discuss the ability of glutamate to exert an "energizing role" in cardiac and neuronal models of hypoxia/reoxygenation (H/R) injury, focusing on the Na+/Ca2+ exchanger (NCX) and the Na+-dependent excitatory amino acid transporters (EAATs) as key players in this metabolic pathway.


Subject(s)
Glutamate Plasma Membrane Transport Proteins/metabolism , Glutamic Acid/metabolism , Sodium-Calcium Exchanger/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cell Survival , Energy Metabolism , Humans
5.
Eur Radiol ; 29(7): 3927-3934, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30989349

ABSTRACT

OBJECTIVES: To estimate the incidence of contrast-induced acute kidney injury (CI-AKI) after intravenous (iv) iodinated contrast material (ICM) exposure. METHODS: This prospective cohort study included all consecutive patients who underwent radiological investigations using low-osmolar iopamidol 370 mg/ml in a regional hospital over a period of 36 months, without any exclusion criteria. The estimated glomerular filtration rate (eGFR) was evaluated using the MRDR equation before (2-10 days) and after (24-36 h) radiological investigations. CI-AKI was defined as a ≥ 25% decrease in eGFR from baseline. CI-AKI incidence was estimated using a binomial distribution. The association between CI-AKI and demographic and clinical characteristics was modeled using logistic regression. RESULTS: The study included 1541 patients with a median age of 68 (1st-3rd quartiles 58-76) years with various comorbidities, 30% of whom had pre-existing CKD. Patients affected by stage III or IV chronic kidney disease (CKD) received an infusion of 0.9% normal saline (1.0-1.5 ml/kg/h) before and after iso-osmolar iodixanol administration. CI-AKI was observed in 33 patients (2.1%, 95% CI 1.5-3.0). The logistic regression analysis showed that antibiotic and statin therapies were significantly associated with CI-AKI. The probability of developing CI-AKI decreased by 80% in patients taking statins (OR = 0.20, 95% CI 0.03; 0.68) and increased approximately three times in patients with antibiotic therapy compared with those who did not take statins and antibiotics (OR = 2.92, 95% CI 1.21; 6.36). CONCLUSIONS: Our data suggest that low-osmolar iopamidol carries a low incidence of nephrotoxicity, even in subjects with various comorbid conditions or reduced renal function. KEY POINTS: • IV administration of ICM carries a low incidence of nephrotoxicity, which was transient in observed patients. • Statin therapy is negatively associated with AKI in patients exposed to ICM. • Pre-existing impairment of renal function is not associated with AKI in patients exposed to ICM.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/epidemiology , Contrast Media/administration & dosage , Iopamidol/adverse effects , Triiodobenzoic Acids/adverse effects , Administration, Intravenous , Aged , Female , Glomerular Filtration Rate , Humans , Incidence , Iopamidol/administration & dosage , Logistic Models , Male , Middle Aged , Odds Ratio , Prospective Studies , Triiodobenzoic Acids/administration & dosage
6.
Cell Death Dis ; 10(2): 80, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692508

ABSTRACT

Progressive accumulation of α-synuclein (α-syn) and exposure to environmental toxins are risk factors that may both concur to Parkinson's disease (PD) pathogenesis. Electrophysiological recordings of field postsynaptic potentials (fEPSPs) and Ca2+ measures in striatal brain slices and differentiated SH-SY5Y cells showed that co-application of α-syn and the neurotoxic pesticide rotenone (Rot) induced Ca2+ dysregulation and alteration of both synaptic transmission and cell function. Interestingly, the presence of the mitochondrial NCX inhibitor CGP-37157 prevented these alterations. The specific involvement of the mitochondrial NCX was confirmed by the inability of the plasma membrane inhibitor SN-6 to counteract such phenomenon. Of note, using a siRNA approach, we found that NCX1 was the isoform specifically involved. These findings suggested that NCX1, operating on the mitochondrial membrane, may have a critical role in the maintenance of ionic Ca2+ homeostasis in PD and that its inhibition most likely exerts a protective effect in the toxicity induced by α-syn and Rot.


Subject(s)
Corpus Striatum/metabolism , Mitochondria/metabolism , Neurons/metabolism , Rotenone/adverse effects , Sodium-Calcium Exchanger/metabolism , alpha-Synuclein/adverse effects , Animals , Disease Models, Animal , Humans , Parkinson Disease , Rats , Rats, Wistar
7.
Cell Death Dis ; 9(7): 731, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29955038

ABSTRACT

In brain ischemia, reduction in oxygen and substrates affects mitochondrial respiratory chain and aerobic metabolism, culminating in ATP production impairment, ionic imbalance, and cell death. The restoration of blood flow and reoxygenation are frequently associated with exacerbation of tissue injury, giving rise to ischemia/reperfusion (I/R) injury. In this setting, the imbalance of brain bioenergetics induces important metabolic adaptations, including utilization of alternative energy sources, such as glutamate. Although glutamate has long been considered as a neurotoxin, it can also be used as intermediary metabolite for ATP synthesis, and both the Na+/Ca2+ exchanger (NCX) and the Na+-dependent excitatory amino-acid transporters (EAATs) are essential in this pathway. Here we analyzed the role of NCX in the potential of glutamate to improve metabolism and survival of neuronal cells subjected to hypoxia/reoxygenation (H/R). In SH-SY5Y neuroblastoma cells differentiated into a neuron-like state, H/R produced a significant cell damage, a decrease in ATP cellular content, and intracellular Ca2+ alterations. Exposure to glutamate at the onset of the reoxygenation phase attenuated H/R-induced cell damage and evoked a significant raise in intracellular ATP levels. Furthermore, we found that in H/R cells NCX reverse-mode activity was reduced, and that glutamate limited such reduction. All the effects induced by glutamate supplementation were lost when cells were transfected with small interfering RNA against NCX1 and EAAT3, suggesting the need of a specific functional interplay between these proteins for glutamate-induced protection. Collectively, our results revealed the potential beneficial effect of glutamate in an in vitro model of H/R injury and focused on the essential role exerted by NCX1. Although preliminary, these findings could be a starting point to further investigate in in vivo systems such protective effect in ischemic settings, shedding a new light on the classical view of glutamate as detrimental factor.


Subject(s)
Glutamic Acid/metabolism , Models, Biological , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Sodium-Calcium Exchanger/metabolism , Adenosine Triphosphate/biosynthesis , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Deoxyglucose/pharmacology , Energy Metabolism , Excitatory Amino Acid Transporter 3/metabolism , Humans , Neuroprotective Agents/pharmacology , Oligomycins/pharmacology
8.
J Forensic Sci ; 63(3): 965-968, 2018 May.
Article in English | MEDLINE | ID: mdl-28886207

ABSTRACT

Kambo is a substance obtained from the skin secretions of a frog, Phyllomedusa bicolor, popular in the Amazon region, which is administered via the transdermal route. We report a case of 42-year-old man found dead in his house. Near the corpse, a plastic box labeled as "Kambo sticks" was found. The man was a chronic consumer of Kambo and no previous pathology or genetic disease emerged in clinical history from the declaration of his general practitioner. Autopsy investigations and toxicological analysis were performed. The histopathological examination showed left ventricular hypertrophy. Toxicological screening was negative for ethanol and other drugs. Phyllocaerulein, phyllokinin, and deltorphin A were isolated from the Kambo sticks but, only deltorphin A was detected in blood sample. We describe the first forensic case of death associated with Kambo administration. We attempt to explain how its use could be related to the cause of sudden death in this case.


Subject(s)
Anura , Ceremonial Behavior , Death, Sudden/etiology , Oligopeptides/blood , Adult , Animals , Coronary Artery Disease/pathology , Coronary Vessels/pathology , Humans , Hypertrophy, Left Ventricular/pathology , Male , Skin/metabolism
9.
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
10.
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
11.
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
12.
Eur J Pharmacol ; 746: 31-40, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25445045

ABSTRACT

Several molecular pathways involved in the development of cardiac hypertrophy are triggered by perturbation of intracellular Ca(2+) homeostasis. Within the heart, Na(+)/Ca(2+) exchanger 1 (NCX1) is one of the main determinant in controlling Ca(2+) homeostasis. In cardiac hypertrophy and heart failure NCX1 expression and activity have been reported to be altered. It has been shown that chronic bacterial infections (sepsis, endocarditis, and myocarditis) can promote cardiac hypertrophy. Bacterial stressors, such as the Gram-negative endotoxin lipopolysaccharide (LPS), can directly or indirectly affect intracellular Ca(2+) homeostasis in the heart and induce the development of cardiac hypertrophy. The present study aimed at evaluating the potential link between the signal pathways activated in LPS-exposed myocytes and NCX1. In the whole rat heart, LPS perfusion induced an early hypertrophy response during which NCX1 expression significantly increased. Notably, all these changes were completely prevented by the NCX inhibitor SN-6. We further dissect the role of NCX1 in the LPS-induced hypertrophic response in an in vitro cardiac model based on two H9c2 cardiomyoblast clones, namely H9c2-WT (lacking endogenous NCX1 expression) and H9c2-NCX1 (stably transfected with a functional NCX1). H9c2-NCX1 were more susceptible than H9c2-WT to develop a hypertrophic phenotype, and they displayed a significant increase in NCX1 expression and function after LPS treatment. SN-6 completely counteracted both hypertrophic response and exchanger alterations induced by LPS in H9c2-NCX1 cells, but it had no effects on H9c2-WT. Collectively, our results suggest that NCX1 plays a critical role in promoting myocardial hypertrophy triggered by LPS.


Subject(s)
Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Lipopolysaccharides/toxicity , Sodium-Calcium Exchanger/metabolism , Animals , Benzyl Compounds/pharmacology , Cardiomegaly/genetics , Cell Line , Male , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myoblasts, Cardiac/pathology , Rats , Rats, Wistar , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Thiazolidines/pharmacology , Up-Regulation/drug effects
13.
Mol Pharmacol ; 84(4): 603-14, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23913256

ABSTRACT

It is known that glutamate (Glu), the major excitatory amino acid in the central nervous system, can be an essential source for cell energy metabolism. Here we investigated the role of the plasma membrane Na(+)/Ca(2+) exchanger (NCX) and the excitatory amino acid transporters (EAATs) in Glu uptake and recycling mechanisms leading to ATP synthesis. We used different cell lines, such as SH-SY5Y neuroblastoma, C6 glioma and H9c2 as neuronal, glial, and cardiac models, respectively. We first observed that Glu increased ATP production in SH-SY5Y and C6 cells. Pharmacological inhibition of either EAAT or NCX counteracted the Glu-induced ATP synthesis. Furthermore, Glu induced a plasma membrane depolarization and an intracellular Ca(2+) increase, and both responses were again abolished by EAAT and NCX blockers. In line with the hypothesis of a mutual interplay between the activities of EAAT and NCX, coimmunoprecipitation studies showed a physical interaction between them. We expanded our studies on EAAT/NCX interplay in the H9c2 cells. H9c2 expresses EAATs but lacks endogenous NCX1 expression. Glu failed to elicit any significant response in terms of ATP synthesis, cell depolarization, and Ca(2+) increase unless a functional NCX1 was introduced in H9c2 cells by stable transfection. Moreover, these responses were counteracted by EAAT and NCX blockers, as observed in SH-SY5Y and C6 cells. Collectively, these data suggest that plasma membrane EAAT and NCX are both involved in Glu-induced ATP synthesis, with NCX playing a pivotal role.


Subject(s)
Adenosine Triphosphate/biosynthesis , Cell Membrane/metabolism , Excitatory Amino Acid Transporter 1/metabolism , Glutamic Acid/pharmacology , Sodium-Calcium Exchanger/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/drug effects , Heart/drug effects , Heart/physiology , Humans , Rats
14.
PLoS One ; 7(3): e34015, 2012.
Article in English | MEDLINE | ID: mdl-22479505

ABSTRACT

Glutamate is emerging as a major factor stimulating energy production in CNS. Brain mitochondria can utilize this neurotransmitter as respiratory substrate and specific transporters are required to mediate the glutamate entry into the mitochondrial matrix. Glutamate transporters of the Excitatory Amino Acid Transporters (EAATs) family have been previously well characterized on the cell surface of neuronal and glial cells, representing the primary players for glutamate uptake in mammalian brain. Here, by using western blot, confocal microscopy and immunoelectron microscopy, we report for the first time that the Excitatory Amino Acid Carrier 1 (EAAC1), an EAATs member, is expressed in neuronal and glial mitochondria where it participates in glutamate-stimulated ATP production, evaluated by a luciferase-luciferin system. Mitochondrial metabolic response is counteracted when different EAATs pharmacological blockers or selective EAAC1 antisense oligonucleotides were used. Since EAATs are Na(+)-dependent proteins, this raised the possibility that other transporters regulating ion gradients across mitochondrial membrane were required for glutamate response. We describe colocalization, mutual activity dependency, physical interaction between EAAC1 and the sodium/calcium exchanger 1 (NCX1) both in neuronal and glial mitochondria, and that NCX1 is an essential modulator of this glutamate transporter. Only NCX1 activity is crucial for such glutamate-stimulated ATP synthesis, as demonstrated by pharmacological blockade and selective knock-down with antisense oligonucleotides. The EAAC1/NCX1-dependent mitochondrial response to glutamate may be a general and alternative mechanism whereby this neurotransmitter sustains ATP production, since we have documented such metabolic response also in mitochondria isolated from heart. The data reported here disclose a new physiological role for mitochondrial NCX1 as the key player in glutamate-induced energy production.


Subject(s)
Adenosine Triphosphate/metabolism , Brain/metabolism , Excitatory Amino Acid Transporter 3/metabolism , Glutamic Acid/metabolism , Mitochondria/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Humans , Ions , Malates/chemistry , Microscopy, Confocal/methods , Microscopy, Immunoelectron/methods , Neurons/metabolism , Oxidative Stress , PC12 Cells , Pyruvic Acid/chemistry , Rats , Rats, Wistar , Sodium/metabolism , Swine
15.
Curr Drug Metab ; 12(3): 278-86, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21470106

ABSTRACT

It is well known that interindividual variability can affect the response to many drugs in relation to age, gender, diet, and organ function. Pharmacogenomic studies have also documented that genetic polymorphisms can exert clinically significant effects in terms of drug resistance, efficacy and toxicity by modifying the expression of critical gene products (drug-metabolizing enzymes, transporters, and target molecules) as well as pharmacokinetic and pharmacodynamic parameters. A growing body of in vitro and clinical evidence suggests that common polymorphisms in the folate gene pathway are associated with an altered response to methotrexate (MTX) in patients with malignancy and autoimmune disease. Such polymorphisms may also induce significant MTX toxicity requiring expensive monitoring and treatment. Although the available data are not conclusive, they suggest that in the future MTX pharmacogenetics could play a key role in clinical practice by improving and tailoring treatment. This review describes the genetic polymorphisms that significantly influence MTX resistance, efficacy, and toxicity.


Subject(s)
Methotrexate/metabolism , Methotrexate/pharmacology , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacology , Drug Resistance , Folic Acid Antagonists/adverse effects , Folic Acid Antagonists/metabolism , Folic Acid Antagonists/pharmacology , Humans , Methotrexate/adverse effects , Pharmacogenetics , Polymorphism, Genetic
16.
Pharmacol Res ; 61(4): 334-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19941959

ABSTRACT

The long-term effects of perinatal Delta(9)-tetrahydrocannabinol (Delta(9)-THC) exposure - from gestational day (GD) 15 to postnatal day (PND) 9 - on hippocampal glutamatergic neurotransmission were studied in slices from the 40-day-old offspring of Delta(9)-THC exposed (Delta(9)-THC-rats) and vehicle-exposed (control) dams. Basal and in K+-evoked endogenous hippocampal glutamate outflow were both significantly decreased in Delta(9)-THC-rats. The effect of short Delta(9)-THC exposure (0.1microM) on K(+)-evoked glutamate release disclosed a loss of the stimulatory effect of Delta(9)-THC on hippocampal glutamate release in Delta(9)-THC-rats, but not in controls. In addition, l-[(3)H]-glutamate uptake was significantly lower in hippocampal slices from Delta(9)-THC-rats, where a significant decrease in glutamate transporter 1 (GLT1) and glutamate/aspartate transporter (GLAST) protein was also detected. Collectively, these data demonstrate that perinatal exposure to cannabinoids induces long-term impairment in hippocampal glutamatergic neurotransmission that persist into adolescence.


Subject(s)
Dronabinol/toxicity , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Hippocampus/drug effects , Synaptic Transmission/drug effects , Animals , Animals, Newborn , Female , Hippocampus/metabolism , In Vitro Techniques , Potassium/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar
17.
Pharmacol Res ; 56(6): 556-65, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18024055

ABSTRACT

In neural cells, Na+/Ca2+ exchanger (NCX) participates in Ca2+ recycling across mitochondrial membranes, thus contributing to shape Ca2+ responses. NCX exchanger isoform proteins, NCX1-3, are widely distributed in mammalian brain, where they localize to neuronal, glial and endothelial cells, but anatomical data on their mitochondrial expression are scanty. In the present work, mitochondrial localization of NCX1-3 was investigated in rat neocortex and hippocampus by means of western blotting analysis and in situ electron microscopy immunocytochemistry. Results showed that a conspicuous population of neuronal and astrocytic mitochondria express NCX1-3, with distinct isoforms exhibiting differential patterns of mitochondrial expression. In neurons, percentages of NCXs-labelled mitochondria varied significantly between diverse subcellular regions: the majority of NCXs-expressing mitochondria were found in dendrites, often located beneath the plasmalemma and near postsynaptic sites. In astrocytes, most NCXs-labelled mitochondria were situated close to the cellular surface. Present quantitative and qualitative immunocytochemical data suggest that all NCX isoforms contribute to mitochondrial Ca2+ homeostasis in neurons and glial cells in vivo, and that NCXs may be particularly involved in handling Ca2+ in dendritic, subplasmalemmal mitochondria, thus emphasizing the role of mitochondrial NCX1-3 in shaping postsynaptic calcium transients.


Subject(s)
Astrocytes/metabolism , Mitochondria/metabolism , Neurons/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Male , Protein Isoforms , Rats , Rats, Sprague-Dawley
18.
Neuropharmacology ; 53(3): 369-78, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17631920

ABSTRACT

Prenatal exposure to the CB1 receptor agonist (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)-pyrrolo[1,2,3-de]-1,4-benzoxazinyl]-(1-naphthalenyl)methanone) mesylate (WIN) at a daily dose of 0.5 mg/kg, and Delta9-tetrahydrocannabinol (Delta9-THC) at a daily dose of 5 mg/kg, reduced dialysate glutamate levels in frontal cerebral cortex of adolescent offspring (40-day-old) with respect to those born from vehicle-treated mothers. WIN treatment induced a statistically significant enhancement of Vmaxl-[3H]glutamate uptake, whereas it did not modify glutamate Km, in frontal cerebral cortex synaptosomes of adolescent rats. Western blotting analysis, performed either in membrane proteins derived from homogenates and in proteins extracted from synaptosomes of frontal cerebral cortex, revealed that prenatal WIN exposure enhanced the expression of glutamate transporter 1 (GLT1) and excitatory amino acid carrier 1 (EAAC1). Moreover, immunocytochemical analyses of frontal cortex area revealed a more intense GLT1 and EAAC1 immunoreactivity (ir) distribution in the WIN-treated group. Collectively these results show that prenatal exposure to the cannabinoid CB1 receptor agonist WIN increases expression and functional activity of GLT1 and EAAC1 glutamate transporters (GluTs) associated to a decrease of cortical glutamate outflow, in adolescent rats. These findings may contribute to explain the mechanism underlying the cognitive impairment observed in the offspring of mothers who used marijuana during pregnancy.


Subject(s)
Benzoxazines/pharmacology , Excitatory Amino Acid Transporter 2/agonists , Excitatory Amino Acid Transporter 3/agonists , Frontal Lobe/drug effects , Glutamic Acid/metabolism , Morpholines/pharmacology , Naphthalenes/pharmacology , Prenatal Exposure Delayed Effects , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Dronabinol/administration & dosage , Excitatory Amino Acid Transporter 2/physiology , Excitatory Amino Acid Transporter 3/physiology , Female , Frontal Lobe/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Wistar
19.
J Neurosci ; 27(18): 4919-28, 2007 May 02.
Article in English | MEDLINE | ID: mdl-17475800

ABSTRACT

Heteromeric assembly of KCNQ2 and KCNQ3 subunits underlie the M-current (I(KM)), a slowly activating and noninactivating neuronal K(+) current. Mutations in KCNQ2 and KCNQ3 genes cause benign familial neonatal convulsions (BFNCs), a rare autosomal-dominant epilepsy of the newborn. In the present study, we describe the identification of a novel KCNQ2 heterozygous mutation (c587t) in a BFNC-affected family, leading to an alanine to valine substitution at amino acid position 196 located at the N-terminal end of the voltage-sensing S(4) domain. The consequences on KCNQ2 subunit function prompted by the A196V substitution, as well as by the A196V/L197P mutation previously described in another BFNC-affected family, were investigated by macroscopic and single-channel current measurements in CHO cells transiently transfected with wild-type and mutant subunits. When compared with KCNQ2 channels, homomeric KCNQ2 A196V or A196V/L197P channels showed a 20 mV rightward shift in their activation voltage dependence, with no concomitant change in maximal open probability or single-channel conductance. Furthermore, current activation kinetics of KCNQ2 A196V channels displayed an unusual dependence on the conditioning prepulse voltage, being markedly slower when preceded by prepulses to more depolarized potentials. Heteromeric channels formed by KCNQ2 A196V and KCNQ3 subunits displayed gating changes similar to those of KCNQ2 A196V homomeric channels. Collectively, these results reveal a novel role for noncharged residues in the N-terminal end of S(4) in controlling gating of I(KM) and suggest that gating changes caused by mutations at these residues may decrease I(KM) function, thus causing neuronal hyperexcitability, ultimately leading to neonatal convulsions.


Subject(s)
Epilepsy, Benign Neonatal/genetics , Epilepsy, Benign Neonatal/metabolism , Ion Channel Gating/genetics , KCNQ2 Potassium Channel/genetics , Mutation , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , CHO Cells , Child, Preschool , Cricetinae , Cricetulus , Female , Humans , Infant , Ion Channel Gating/physiology , KCNQ2 Potassium Channel/physiology , Male , Membrane Potentials/genetics , Membrane Potentials/physiology , Molecular Sequence Data , Pedigree
20.
Cell Calcium ; 41(3): 221-34, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16914199

ABSTRACT

Na(+)-Ca(2+) exchanger (NCX) controls cytosolic Ca(2+) and Na(+) concentrations ([Ca(2+)](i) and [Na(+)](i)) in eukaryotic cells. Here we investigated by immunocytochemistry the cellular and subcellular localization of the three known NCX isoforms, NCX1, NCX2 and NCX3, in adult rat neocortex and hippocampus. NCX1-3 were widely expressed in both brain areas: NCX1 immunoreactivity (ir) was exclusively associated to neuropilar puncta, while NCX2-3 were also detected in neuronal somata and dendrites. NCX1-3 ir was often identified around blood vessels. In both neocortex and hippocampus, all NCX isoforms were prominently expressed in dendrites and dendritic spines contacted by asymmetric axon terminals, whereas they were poorly expressed in presynaptic boutons. In addition, NCX1-3 ir was detected in astrocytes, notably in distal processes ensheathing excitatory synapses. All NCXs were expressed in perivascular astrocytic endfeet and endothelial cells. The robust expression of NCX1-3 in heterogeneous cell types in the brain in situ emphasizes their role in handling Ca(2+) and Na(+) in both excitable and non-excitable cells. Perisynaptic localization of NCX1-3 in dendrites and spines indicates that all isoforms are favourably located for buffering [Ca(2+)](i) in excitatory postsynaptic sites. NCX1-3 expressed in perisynaptic glial processes may participate in shaping astrocytic [Ca(2+)](i) transients evoked by ongoing synaptic activity.


Subject(s)
Cerebellar Cortex/metabolism , Membrane Transport Proteins/biosynthesis , Sodium-Calcium Exchanger/biosynthesis , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Axons/metabolism , Axons/ultrastructure , Cell Line , Cerebellar Cortex/ultrastructure , Cricetinae , Dendrites/metabolism , Dendrites/ultrastructure , Hippocampus/metabolism , Ion Transport/physiology , Male , Neuronal Plasticity , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Synapses/metabolism , Synapses/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...