Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Antioxidants (Basel) ; 12(4)2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37107312

ABSTRACT

Attention-Deficit/Hyperactivity Disorder (ADHD) is one of the most prevalent neurodevelopmental disorders. Interestingly, children with ADHD seem to experience more ophthalmologic abnormalities, and the impact of methylphenidate (MPH) use on retinal physiology remains unclear. Thus, we aimed to unravel the retina's structural, functional, and cellular alterations and the impact of MPH in ADHD versus the control conditions. For that, spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY) were used as animal models of ADHD and the controls, respectively. Animals were divided into four experimental groups as follows: WKY vehicle (Veh; tap water), WKY MPH (1.5 mg/kg/day), SHR Veh, SHR MPH. Individual administration was performed by gavage between P28-P55. Retinal physiology and structure were evaluated at P56 followed by tissue collection and analysis. The ADHD animal model presents the retinal structural, functional, and neuronal deficits, as well as the microglial reactivity, astrogliosis, blood-retinal barrier (BRB) hyperpermeability and a pro-inflammatory status. In this model, MPH had a beneficial effect on reducing microgliosis, BRB dysfunction, and inflammatory response, but did not correct the neuronal and functional alterations in the retina. Curiously, in the control animals, MPH showed an opposite effect since it impaired the retinal function, neuronal cells, and BRB integrity, and also promoted both microglia reactivity and upregulation of pro-inflammatory mediators. This study unveils the retinal alterations in ADHD and the opposite effects induced by MPH in the retina of ADHD and the control animal models.

2.
Eur J Clin Invest ; 52(4): e13694, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34694635

ABSTRACT

BACKGROUND: Methamphetamine abuse is a worldwide concern with long-term health complications. Its impact on neurons has been extensively investigated, and it is currently known that glial cells, including astrocytes, are involved in drug-induced outcomes. Importantly, METH also causes blood-brain barrier (BBB) disruption and astrocytes are critical for BBB (dys)function. Therefore, we aimed to clarify the involvement of neuroinflammation mediated by astrocytes in BBB permeability and brain oedema induced by METH. Further, we aimed to identify a new approach to counteract METH effects. METHODS: Mice were administered with a METH binge regimen (4 × 10 mg/kg) alone or in combination with parthenolide (PTL; 4 × 1 mg/kg), and hippocampi were analysed. For in vitro studies, mouse primary cultures of astrocytes were exposed to 250 µM METH, alone or co-treated with 10 µM PTL. RESULTS: We observed a neuroinflammatory response characterized by astrocytic morphological changes and increased TNF-α, iNOS and ICAM-1 protein levels (213.62%, 205.76% and 191.47% of control, respectively). Additionally, brain oedema and BBB disruption were identified by increased water content (81.30% of tissue weight) and albumin (224.40% of control) in the hippocampal tissue, as well as a significant decrease in vessel coverage by astrocytes after METH exposure. Regarding astrocyte cultures, we further identified TNF-α as a key player in METH-induced cell swelling. Importantly, PTL (present in feverfew plant) prevented both animal and in vitro effects induced by METH. CONCLUSIONS: We provided important insights on brain dysfunction induced by METH, and we also suggest a new approach to counteract such negative effects.


Subject(s)
Astrocytes/drug effects , Blood-Brain Barrier/drug effects , Methamphetamine/pharmacology , Sesquiterpenes/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL
3.
Pharmaceuticals (Basel) ; 14(3)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33809599

ABSTRACT

3,4-Methylenedioxypyrovalerone (MDPV), a widely available synthetic cathinone, is a popular substitute for classical controlled drugs of abuse, such as methamphetamine (METH). Although MDPV poses public health risks, its neuropharmacological profile remains poorly explored. This study aimed to provide evidence on that direction. Accordingly, C57BL/6J mice were exposed to a binge MDPV or METH regimen (four intraperitoneal injections every 2 h, 10 mg/kg). Locomotor, exploratory, and emotional behavior, in addition to striatal neurotoxicity and glial signature, were assessed within 18-24 h, a known time-window encompassing classical amphetamine dopaminergic neurotoxicity. MDPV resulted in unchanged locomotor activity (open field test) and emotional behavior (elevated plus maze, splash test, tail suspension test). Additionally, striatal TH (METH neurotoxicity hallmark), Iba-1 (microglia), GFAP (astrocyte), RAGE, and TLR2/4/7 (immune modulators) protein densities remained unchanged after MDPV-exposure. Expectedly, and in sheer contrast with MDPV, METH resulted in decrease general locomotor activity paralleled by a significant striatal TH depletion, astrogliosis, and microglia arborization alterations (Sholl analysis). This comparative study newly highlights that binge MDPV-exposure comes without evident behavioral, neurochemical, and glial changes at a time-point where METH-induced striatal neurotoxicity is clearly evident. Nevertheless, neuropharmacological MDPV signature needs further profiling at different time-points, regimens, and brain regions.

4.
Toxicol Lett ; 334: 53-59, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32956829

ABSTRACT

Methamphetamine (METH) consumption is a health problem that leads to neurological and psychiatric disturbances. The cellular alterations behind these conditions have been extensively investigated and it is now well-established that METH causes cerebrovascular alterations being a key feature in drug-induced neuropathology. Although promising advances in understanding the blood-brain barrier (BBB) alterations induced by METH, there is still no available approach to counteract or diminish such effects. Interestingly, several studies show that neuropeptide Y (NPY) has an important protective role against METH-induced neuronal and glial toxicity, as well as behavioral deficits. Despite these beneficial effects of the NPY system, nothing is known about its role in brain endothelial cells under conditions of METH exposure. Thus, our aim was to unravel the effect of NPY and its receptors against METH-induced endothelial cell dysfunction. For that, we used a human brain microvascular endothelial cell line (hCMEC/D3) and our results demonstrate that endothelial cells express both NPY Y1 (Y1R) and Y2 (Y2R) receptors, but only Y2R is upregulated after METH exposure. Moreover, this drug of abuse induced endothelial cell death and elicited the production of reactive oxygen species (ROS) by these cells, which were prevented by the activation of Y2R. Additional, cell death and oxidative stress triggered by METH were dependent on the concentration of the drug. In sum, with the present study we identified for the first time the NPY system, and particularly the Y2R subtype, as a promising target to protect against METH-induced neurovascular dysfunction.


Subject(s)
Brain/blood supply , Central Nervous System Stimulants/toxicity , Endothelial Cells/drug effects , Methamphetamine/toxicity , Oxidative Stress/drug effects , Receptors, Neuropeptide Y/agonists , Blood-Brain Barrier/metabolism , Brain/cytology , Cell Death/drug effects , Cell Line , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Humans , Microvessels/cytology , Microvessels/drug effects , Microvessels/metabolism , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/pharmacology , Peptide Fragments/pharmacology , Reactive Oxygen Species/metabolism , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/genetics , Up-Regulation
5.
Mol Pharm ; 17(6): 2145-2154, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32339462

ABSTRACT

Photodynamic therapy (PDT) has demonstrated encouraging anticancer therapeutic results, but the current clinically approved photosensitizers (PSs) are not ideal in the treatment of bladder cancer. Conventional PSs have low selectivity to the bladder tumor tissue and induce toxicity or bystander effects on nontumor urothelium. Previous studies demonstrated that the use of galactose-photosensitizer (PS) conjugates is a more selective method of delivering PDT-mediated toxicity due to their ability to recognize carbohydrate-binding domains overexpressed in bladder tumors. Using patient-derived bladder tumor specimens cultured ex vivo and bladder cancer cell lines with different PDT sensitivity, we find that a galactose-phthalocyanine (PcGal16) accumulates in bladder tumors expressing galactose-binding proteins and internalizes through an endocytic process. The endocytosis mechanism is cell line-dependent. In HT-1376 bladder cancer lines resistant to PDT, depletion of caveolin-1-the main structural protein of caveolae structures-increased the amount of sugar-binding proteins, i.e. GLUT1, at the cell membrane resulting in an improved PcGal16 uptake and PDT efficacy. These data show the potential of ex vivo cultures of bladder cancer, that ideally could mimic the original microenvironment, in screening galacto-PDT agents. Additionally, our studies demonstrate that PDT efficacy in bladder cancer depends on the endocytic mechanisms that regulate PS accumulation and internalization in cancer cells.


Subject(s)
Caveolin 1/metabolism , Indoles/chemistry , Indoles/therapeutic use , Photochemotherapy/methods , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/therapy , Aged , Blotting, Western , Caveolin 1/genetics , Cell Line, Tumor , Endocytosis/drug effects , Female , Galectin 1/genetics , Galectin 1/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , In Vitro Techniques , Isoindoles , Male , Microscopy, Fluorescence
6.
Eur Neuropsychopharmacol ; 29(2): 195-210, 2019 12.
Article in English | MEDLINE | ID: mdl-30554860

ABSTRACT

Methylphenidate (MPH) is the classic treatment for attention deficit hyperactivity disorder (ADHD) among children and adults. Despite its beneficial effects, non-medical use of MPH is nowadays a problem with high impact on society. Thus, our goal was to uncover the neurovascular and cognitive effects of MPH chronic use during a critical period of development in control conditions. For that, male Wistar Kyoto rats were treated with MPH (1.5 or 5 mg/kg/day at weekdays, per os) from P28 to P55. We concluded that the higher dose of MPH caused hippocampal blood-brain barrier (BBB) hyperpermeability by vesicular transport (transcytosis) concomitantly with the presence of peripheral immune cells in the brain parenchyma. These observations were confirmed by in vitro studies, in which the knockdown of caveolin-1 in human brain endothelial cells prevented the increased permeability and leukocytes transmigration triggered by MPH (100 µM, 24 h). Furthermore, MPH led to astrocytic atrophy and to a decrease in the levels of several synaptic proteins and impairment of AKT/CREB signaling, together with working memory deficit assessed in the Y-maze test. On the contrary, we verified that the lower dose of MPH (1.5 mg/kg/day) increased astrocytic processes and upregulated several neuronal proteins as well as signaling pathways involved in synaptic plasticity culminating in working memory improvement. In conclusion, the present study reveals that a lower dose of MPH in normal rats improves memory performance being associated with the modulation of astrocytic morphology and synaptic machinery. However, a higher dose of MPH leads to BBB dysfunction and memory impairment.


Subject(s)
Central Nervous System Stimulants/pharmacology , Hippocampus/drug effects , Memory/drug effects , Methylphenidate/pharmacology , Transcytosis/drug effects , Animals , Animals, Newborn , Antioxidants/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Hippocampus/anatomy & histology , Hippocampus/ultrastructure , Lipid Peroxidation/drug effects , Male , Maze Learning/drug effects , Nerve Tissue Proteins/metabolism , Neuroglia/drug effects , Neuroglia/ultrastructure , Neurons/drug effects , Neurons/ultrastructure , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Transcytosis/physiology , Up-Regulation/drug effects
7.
Oncotarget ; 9(68): 32929-32942, 2018 Aug 31.
Article in English | MEDLINE | ID: mdl-30250640

ABSTRACT

Brain cognitive reserve refers to the ability of the brain to manage different challenges that arise throughout life, making it resilient to neuropathology. Hippocampal adult neurogenesis has been considered to be a relevant contributor for brain cognitive reserve and brain plasticity. Coriolus versicolor (CV), a common healthful mushroom, has been receiving increasing attention by its antitumoral, anti-inflammatory, antioxidant, antibacterial, and immunomodulatory properties, including in the hippocampus. Herein, we evaluated whether CV biomass oral administration for 2.5 months enhances hippocampal neurogenic reserve under normal/physiological conditions, by quantifying hippocampal dentate gyrus (DG) granular cell layer (GCL) and subgranular zone (SGZ) volumes, proliferation, number and dendritic complexity features of hippocampal newly-generated neurons. We also analyzed ß-catenin levels in DG newly-generated immature neurons, because it plays a major role in neurogenesis. Although no differences were observed in the volume of GCL and SGZ layers, in proliferation and in the number of newly-generated neurons of controls and CV-administered mice, we found that CV administration promotes a significant increase in dendritic length and branching and total dendritic volume of immature neurons, suggesting a positive effect of oral CV administration in the hippocampal neurogenic reserve. We also observed that ß-catenin levels are increased both in the nucleus and cytoplasm of DG immature neurons, suggesting that Wnt/ß-catenin signalling may play an important role in the CV positive effect on the differentiation of these cells. These data unveil a so far unexplored neurogenic potential of CV supplementation, which emerges as a possible preventive strategy for different neurological conditions.

8.
BMC Pediatr ; 18(1): 212, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29970045

ABSTRACT

BACKGROUND: Exploring the osteogenic effect of different bone-loading sports is particular relevant to understand the interaction between skeletal muscle and bone health during growth. This study aimed to compare total and regional bone and soft-tissue composition between female adolescent swimmers (n=20, 15.71±0.93 years) and volleyball players (n=26, 16.20±0.77 years). METHODS: Dietary intake was obtained using food frequency questionnaires. Body size was given by stature, sitting height, and body mass. Six skinfolds were measured. Bone mineral content (BMC) and density (BMD), lean soft tissue, and fat tissue were assessed using dual-energy X-ray absorptiometry. Pearson's product moment correlation coefficients were calculated to examine the relationships among variables, by type of sport. Comparisons between swimmers and volleyball players were performed using student t-tests for independent samples and multivariate analysis of covariance (controlling for age, training history and body size). RESULTS: Swimmers (BMC: 2328±338 g) and volleyball players (BMC: 2656±470 g) exceeded respectively by 2.1 and 2.8 standard deviation scores the average of international standards for whole body BMC of healthy adolescents. Years of training in swimmers were positively related to the upper limbs BMC (r=+0.49, p<0.05). In volleyball players, years of training correlated significantly with lower limbs BMD (r=+0.43, p<0.05). After adjustments for potential confounders, moderate differences (ES-r=0.32) between swimmers and volleyball players were noted in BMD at the lower limbs (volleyball players: +0.098 g∙cm-2, +7.8%). CONCLUSIONS: Youth female athletes who participate in high-intensity weight-loading activities such as volleyball exhibit moderately higher levels of BMD at the lower limbs compared to non-loading sports such as swimming.


Subject(s)
Body Composition , Bone Density , Resistance Training , Swimming/physiology , Volleyball/physiology , Absorptiometry, Photon , Adolescent , Body Size , Diet , Female , Humans , Lower Extremity/physiology , Skinfold Thickness , Upper Extremity/physiology
9.
Front Pharmacol ; 9: 219, 2018.
Article in English | MEDLINE | ID: mdl-29615903

ABSTRACT

The exposure to supra-physiological levels of glucocorticoids in prenatal life can lead to a long-term impact in brain cytoarchitecture, increasing the susceptibility to neuropsychiatric disorders. Dexamethasone, an exogenous glucocorticoid widely used in pregnant women in risk of preterm delivery, is associated with higher rates of neuropsychiatric conditions throughout life of the descendants. In animal models, prenatal dexamethasone exposure leads to anxious-like behavior and increased susceptibility to depressive-like behavior in adulthood, concomitant with alterations in neuronal morphology in brain regions implicated in the control of emotions and mood. The pharmacologic blockade of the purinergic adenosine A2A receptor, which was previously described as anxiolytic, is also able to modulate neuronal morphology, namely in the hippocampus. Additionally, recent observations point to an interaction between glucocorticoid receptors (GRs) and adenosine A2A receptors. In this work, we explored the impact of dexamethasone on neuronal morphology, and the putative implication of adenosine A2A receptor in the mediation of dexamethasone effects. We report that in vitro hippocampal neurons exposed to dexamethasone (250 nM), in the early phases of development, exhibit a polarized morphology alteration: dendritic atrophy and axonal hypertrophy. While the effect of dexamethasone in the axon is dependent on the activation of adenosine A2A receptor, the effect in the dendrites relies on the activation of GRs, regardless of the activation of adenosine A2A receptor. These results support the hypothesis of the interaction between GRs and adenosine A2A receptors and the potential therapeutic value of modulating adenosine A2A receptors activation in order to prevent glucocorticoid-induced alterations in developing neurons.

10.
Brain Behav Immun ; 68: 169-182, 2018 02.
Article in English | MEDLINE | ID: mdl-29061363

ABSTRACT

Attention deficit hyperactivity disorder (ADHD) is the most prevalent childhood mental disorders that often persists into adulthood. Moreover, methylphenidate (MPH) is the mainstay of medical treatment for this disorder. Yet, not much is known about the neurobiological impact of MPH on control versus ADHD conditions, which is crucial to simultaneously clarify the misuse/abuse versus therapeutic use of this psychostimulant. In the present study, we applied biochemical and behavioral approaches to broadly explore the early-life chronic exposure of two different doses of MPH (1.5 and 5 mg/kg/day) on control and ADHD rats (Wistar Kyoto and Spontaneously Hypertensive rats, respectively). We concluded that the higher dose of MPH promoted blood-brain barrier (BBB) permeability and elicited anxiety-like behavior in both control and ADHD animals. BBB dysfunction triggered by MPH was particularly prominent in control rats, which was characterized by a marked disruption of intercellular junctions, an increase of endothelial vesicles, and an upregulation of adhesion molecules concomitantly with the infiltration of peripheral immune cells into the prefrontal cortex. Moreover, both doses of MPH induced a robust neuroinflammatory and oxidative response in control rats. Curiously, in the ADHD model, the lower dose of MPH (1.5 mg/kg/day) had a beneficial effect since it balanced both immunity and behavior relative to vehicle animals. Overall, the contrasting effects of MPH observed between control and ADHD models support the importance of an appropriate MPH dose regimen for ADHD, and also suggest that MPH misuse negatively affects brain and behavior.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Immune Privilege/physiology , Methylphenidate/pharmacology , Animals , Anxiety/metabolism , Attention/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Central Nervous System Stimulants , Disease Models, Animal , Exploratory Behavior/drug effects , Immune Privilege/immunology , Male , Prefrontal Cortex/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY
11.
PLoS One ; 12(6): e0180357, 2017.
Article in English | MEDLINE | ID: mdl-28662190

ABSTRACT

Exploring the effect of non-impact and impact sports is particular relevant to understand the interaction between skeletal muscle and bone health during growth. The current study aimed to compare total and regional bone and soft-tissue composition, in parallel to measurements of blood lipid and inflammatory profiles between adolescent athletes and non-athletes. Anthropometry, biological maturity, dual energy X-ray absorptiometry (DXA) scans, training load and lipid and inflammatory profiles were assessed in a cross-sectional sample of 53 male adolescents (20 non-athletes, 15 swimmers and 18 basketball players) aged 12-19 years. Multiple comparisons between groups were performed using analysis of variance, covariance and magnitude effects (ES-r and Cohen's d). The comparisons of controls with other groups were very large for high-sensitivity C-reactive protein (d range: 2.17-2.92). The differences between sports disciplines, regarding tissue outputs obtained from DXA scan were moderate for all variables except fat tissue (d = 0.4). It was possible to determine small differences (ES-r = 0.17) between controls and swimmers for bone area at the lower limbs (13.0%). In parallel, between swimmers and basketball players, the gradient of the differences was small (ES-r range: 0.15-0.23) for bone mineral content (24.6%), bone area (11.3%) and bone mineral density (11.1%) at the lower limbs, favoring the basketball players. These observations highlight that youth male athletes presented better blood and soft tissues profiles with respect to controls. Furthermore, sport-specific differences emerged for the lower limbs, with basketball players presenting higher bone mineral content, area and density than swimmers.


Subject(s)
Bone and Bones/metabolism , Inflammation/blood , Lipids/blood , Sports , Stress, Physiological , Absorptiometry, Photon , Adolescent , Adult , Child , Cross-Sectional Studies , Humans , Male , Young Adult
12.
J Appl Physiol (1985) ; 123(1): 161-171, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28385921

ABSTRACT

Parkinson's disease (PD) prodromal stages comprise neuropsychiatric perturbations that critically compromise a patient's quality of life. These nonmotor symptoms (NMS) are associated with exacerbated innate immunity, a hallmark of overt PD. Physical exercise (PE) has the potential to improve neuropsychiatric deficits and to modulate immune network including receptor for advanced glycation end products (RAGE) and Toll-like receptors (TLRs) in distinct pathological settings. Accordingly, the present study aimed to test the hypothesis that PE 1) alleviates PD NMS and 2) modulates neuroimmune RAGE network in experimental PD. Adult Wistar rats subjected to long-term mild treadmill were administered intranasally with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and probed for PD NMS before the onset of motor abnormalities. Twelve days after MPTP, neuroimmune RAGE network transcriptomics (real-time quantitative PCR) was analyzed in frontal cortex, hippocampus, and striatum. Untrained MPTP animals displayed habit-learning and motivational deficits without gross motor impairments (cued version of water-maze, splash, and open-field tests, respectively). A suppression of RAGE and neuroimmune-related genes was observed in frontal cortex on chemical and physical stressors (untrained MPTP: RAGE, TLR5 and -7, and p22 NADPH oxidase; saline-trained animals: RAGE, TLR1 and -5 to -11, TNF-α, IL-1ß, and p22 NADPH oxidase), suggesting the recruitment of compensatory mechanisms to restrain innate inflammation. Notably, trained MPTP animals displayed normal cognitive/motivational performances. Additionally, these animals showed normal RAGE expression and neuroprotective PD-related DJ-1 gene upregulation in frontal cortex when compared with untrained MPTP animals. These findings corroborate PE efficacy in improving PD NMS and newly identify RAGE network as a neural substrate for exercise intervention. Additional research is warranted to unveil functional consequences of PE-induced modulation of RAGE/DJ-1 transcriptomics in PD premotor stages.NEW & NOTEWORTHY This study newly shows that physical exercise (PE) corrects nonmotor symptoms of the intranasal 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of experimental parkinsonism. Additionally, we show that suppression of neuroimmune receptor for advanced glycation end products (RAGE) network occurs in frontal cortex on chemical (MPTP) and physical (PE) interventions. Finally, PE normalizes frontal cortical RAGE transcriptomics and upregulates the neuroprotective DJ-1 gene in the intranasal MPTP model of experimental parkinsonism.


Subject(s)
Neuroimmunomodulation/physiology , Parkinsonian Disorders/immunology , Parkinsonian Disorders/rehabilitation , Physical Conditioning, Animal/physiology , Receptor for Advanced Glycation End Products/biosynthesis , Receptor for Advanced Glycation End Products/immunology , Animals , Brain/immunology , Brain/metabolism , Exercise Test/methods , Male , Physical Conditioning, Animal/methods , Rats , Rats, Wistar , Treatment Outcome
13.
CNS Neurosci Ther ; 23(2): 119-126, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27762079

ABSTRACT

INTRODUCTION: We recently showed that a single high dose of methamphetamine (METH) induces a persistent frontal cortical monoamine depletion that is accompanied by helpless-like behavior in mice. However, brain metabolic alterations underlying both neurochemical and mood alterations remain unknown. AIMS: Herein, we aimed at characterizing frontal cortical metabolic alterations associated with early negative mood behavior triggered by METH. Adult C57BL/6 mice were injected with METH (30 mg/kg, i.p.), and their frontal cortical metabolic status was characterized after probing their mood and anxiety-related phenotypes 3 days postinjection. RESULTS: Methamphetamine induced depressive-like behavior, as indicated by the decreased grooming time in the splash test and by a transient decrease in sucrose preference. At this time, METH did not alter anxiety-like behavior or motor functions. Depolarization-induced glucose uptake was reduced in frontocortical slices from METH-treated mice compared to controls. Consistently, astrocytic glucose transporter (GluT1) density was lower in the METH group. A proton high rotation magic angle spinning (HRMAS) spectroscopic approach revealed that METH induced a significant decrease in N-acetyl aspartate (NAA) and glutamate levels, suggesting that METH decreased neuronal glutamatergic function in frontal cortex. CONCLUSIONS: We report, for the first time, that a single METH injection triggers early self-care and hedonic deficits and impairs frontal cortical energetics in mice.


Subject(s)
Anhedonia/drug effects , Brain Injuries/chemically induced , Brain Injuries/pathology , Central Nervous System Stimulants/toxicity , Cerebral Cortex/drug effects , Methamphetamine/toxicity , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Exploratory Behavior/drug effects , Food Preferences/drug effects , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/metabolism , Glutamic Acid/metabolism , Grooming/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects
14.
J Neurochem ; 138(4): 598-609, 2016 08.
Article in English | MEDLINE | ID: mdl-27221633

ABSTRACT

Convincing evidence indicates that advanced glycation end-products and danger-associated protein S100B play a role in Parkinson's disease (PD). These agents operate through the receptor for advanced glycation end-products (RAGE), which displays distinct isoforms playing protective/deleterious effects. However, the nature of RAGE variants has been overlooked in PD studies. Hence, we attempted to characterize RAGE regulation in early stages of PD striatal pathology. A neurotoxin-based rodent model of PD was used in this study, through administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to C57BL/6 mice. Animals were killed 6 h post-MPTP to assess S100B/RAGE contents (RT-qPCR, ELISA) and RAGE isoform density (WB) and cellular distribution (immunohistochemistry). Dopaminergic and gliotic status were also mapped (HPLC-ED, WB, immunohistochemistry). At this preliminary stage of MPTP-induced PD in mice, RAGE inhibitory isoforms were increased whereas full-length RAGE was not affected. This putative cytoprotective RAGE phenotype paired an inflammatory and pro-oxidant setting fueling DAergic denervation. Increased RAGE inhibitory variants occur in astrocytes showing higher S100B density but no overt signs of hypertrophy or NF-κB activation, a canonical effector of RAGE. These findings expand our understanding of the toxic effect of MPTP on striatum and offer first in vivo evidence of RAGE being a responder in early stages of astrogliosis dynamics, supporting a protective rather tissue-destructive phenotype of RAGE in the initial phase of PD degeneration. These data lay the groundwork for future studies on the relevance of astrocytic RAGE in DAergic neuroprotection strategies. We report increased antagonistic RAGE variants paralleling S100B up-regulation in early stages of MPTP-induced astrogliosis dynamics . We propose that selective RAGE regulation reflects a self-protective mechanism to maintain low levels of RAGE ligands , preventing long-term inflammation and oxidative stress arising from sustained ligands/flRAGE activation . Understanding loss of RAGE protective response to stress may provide new therapeutic options to halt or slow down dopaminergic axonopathy and, ultimately, neuronal death .


Subject(s)
Astrocytes/metabolism , Corpus Striatum/metabolism , Neostriatum/metabolism , Parkinson Disease/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Disease Models, Animal , Male , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products/genetics
15.
CNS Neurosci Ther ; 22(5): 396-403, 2016 May.
Article in English | MEDLINE | ID: mdl-26843141

ABSTRACT

AIMS: Astrocytic S100B and receptor for advanced glycation endproducts (RAGE) have been implicated in Parkinson׳s disease (PD) pathogenesis through yet unclear mechanisms. This study attempted to characterize S100B/mRAGE (signaling isoform) axis in a dying-back dopaminergic (DAergic) axonopathy setting, which mimics an early event of PD pathology. METHODS: C57BL/6 mice were submitted to a chronic MPTP paradigm (20 mg/kg i.p., 2 i.d-12 h apart, 5 days/week for 2 weeks) and euthanized 7 days posttreatment to assess mRAGE cellular distribution and S100B/mRAGE density in striatum, after probing their locomotor activity (pole test and rotarod). Dopaminergic status, oxidative stress, and gliosis were also measured (HPLC-ED, WB, IHC). RESULTS: This MPTP regimen triggered increased oxidative stress (augmented HNE levels), gliosis (GS/Iba1-reactive morphology), loss of DAergic fibers (decreased tyrosine hydroxylase levels), and severe hypodopaminergia. Biochemical deficits were not translated into motor abnormalities, mimicking a presymptomatic PD period. Remarkably, striatal neurotrophic S100B/mRAGE levels and major neuronal mRAGE localization coexist with compensatory responses (3-fold increase in DA turnover), which are important to maintain normal motor function. CONCLUSION: Our findings rule out the involvement of S100B/mRAGE axis in striatal reactive gliosis, DAergic axonopathy and warrant further exploration of its neurotrophic effects in a presymptomatic compensatory PD stage, which is a fundamental period for successful implementation of therapeutic strategies.


Subject(s)
Corpus Striatum/metabolism , MPTP Poisoning/pathology , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Aldehydes/metabolism , Animals , Corpus Striatum/drug effects , Disease Models, Animal , Dopamine/metabolism , Gene Expression Regulation/drug effects , Gliosis/chemically induced , Gliosis/pathology , Hypokinesia/pathology , Hypokinesia/physiopathology , MPTP Poisoning/physiopathology , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Oxidative Stress/drug effects , Receptor for Advanced Glycation End Products/genetics , Rotarod Performance Test , S100 Calcium Binding Protein beta Subunit/genetics , Tyrosine 3-Monooxygenase/metabolism
16.
J Cereb Blood Flow Metab ; 35(8): 1260-71, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25899299

ABSTRACT

Methamphetamine (METH) is a psychostimulant that causes neurologic and psychiatric abnormalities. Recent studies have suggested that its neurotoxicity may also result from its ability to compromise the blood-brain barrier (BBB). Herein, we show that METH rapidly increased the vesicular transport across endothelial cells (ECs), followed by an increase of paracellular transport. Moreover, METH triggered the release of tumor necrosis factor-alpha (TNF-α), and the blockade of this cytokine or the inhibition of nuclear factor-kappa B (NF-κB) pathway prevented endothelial dysfunction. Since astrocytes have a crucial role in modulating BBB function, we further showed that conditioned medium obtained from astrocytes previously exposed to METH had a negative impact on barrier properties also via TNF-α/NF-κB pathway. Animal studies corroborated the in vitro results. Overall, we show that METH directly interferes with EC properties or indirectly via astrocytes through the release of TNF-α and subsequent activation of NF-κB pathway culminating in barrier dysfunction.


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System Stimulants/adverse effects , Endothelial Cells/metabolism , Methamphetamine/adverse effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Biological Transport/drug effects , Blood-Brain Barrier/pathology , Central Nervous System Stimulants/pharmacology , Endothelial Cells/pathology , Methamphetamine/pharmacology , Rats , Rats, Wistar
17.
Neurotox Res ; 25(3): 295-304, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24072398

ABSTRACT

Methamphetamine (METH) triggers a disruption of the monoaminergic system and METH abuse leads to negative emotional states including depressive symptoms during drug withdrawal. However, it is currently unknown if the acute toxic dosage of METH also causes a long-lasting depressive phenotype and persistent monoaminergic deficits. Thus, we now assessed the depressive-like behaviour in mice at early and long-term periods following a single high METH dose (30 mg/kg, i.p.). METH did not alter the motor function and procedural memory of mice as assessed by swimming speed and escape latency to find the platform in a cued version of the water maze task. However, METH significantly increased the immobility time in the tail suspension test at 3 and 49 days post-administration. This depressive-like profile induced by METH was accompanied by a marked depletion of frontostriatal dopaminergic and serotonergic neurotransmission, indicated by a reduction in the levels of dopamine, DOPAC and HVA, tyrosine hydroxylase and serotonin, observed at both 3 and 49 days post-administration. In parallel, another neurochemical feature of depression--astroglial dysfunction--was unaffected in the cortex and the striatal levels of the astrocytic protein marker, glial fibrillary acidic protein, were only transiently increased at 3 days. These findings demonstrate for the first time that a single high dose of METH induces long-lasting depressive-like behaviour in mice associated with a persistent disruption of frontostriatal dopaminergic and serotonergic homoeostasis.


Subject(s)
Depressive Disorder/chemically induced , Depressive Disorder/physiopathology , Methamphetamine/toxicity , Neurotoxicity Syndromes/physiopathology , Animals , Astrocytes/drug effects , Astrocytes/physiology , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Dopamine/metabolism , Escape Reaction/drug effects , Escape Reaction/physiology , Frontal Lobe/drug effects , Frontal Lobe/physiopathology , Locomotion/drug effects , Locomotion/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Memory/physiology , Mice , Mice, Inbred C57BL , Serotonin/metabolism , Swimming/physiology , Time Factors
18.
Int J Pharm ; 415(1-2): 73-82, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21641984

ABSTRACT

Implantable disks for glaucoma treatment were prepared by blending poly(ɛ-caprolactone), PCL, poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) and dorzolamide. Their in vivo performance was assessed by their capacity to decrease intraocular pressure (IOP) in normotensive and hypertensive eyes. Drug mapping showed that release was complete from blend disks and the low molecular weight (MW) PCL after 1 month in vivo. The high MW PCL showed non-cumulative release rates above the therapeutic level during 3 months in vitro. In vivo, the fibrous capsule formation around the implant controls the drug release, working as a barrier membrane. Histologic analysis showed normal foreign body reaction response to the implants. In normotensive eyes, a 20% decrease in IOP obtained with the disks during 1 month was similar to Trusopt eyedrops treatment. In hypertensive eyes, the most sustained decrease was shown by the high MW PCL (40% after 1 month, 30% after 2 months). It was shown that the implants can lower IOP in sustained manner in a rabbit glaucoma model.


Subject(s)
Antihypertensive Agents/administration & dosage , Drug Carriers/chemistry , Eye/drug effects , Glaucoma/drug therapy , Sulfonamides/administration & dosage , Thiophenes/administration & dosage , Animals , Antihypertensive Agents/adverse effects , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/therapeutic use , Calorimetry, Differential Scanning , Drug Carriers/adverse effects , Drug Implants/adverse effects , Eye/pathology , Glaucoma/pathology , Glaucoma/physiopathology , Intraocular Pressure/drug effects , Kinetics , Microscopy, Electron, Scanning , Polyesters/adverse effects , Polyesters/chemistry , Polyethylene Glycols/adverse effects , Polyethylene Glycols/chemistry , Propylene Glycols/adverse effects , Propylene Glycols/chemistry , Rabbits , Solubility , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Sulfonamides/therapeutic use , Surface Properties , Thiophenes/adverse effects , Thiophenes/pharmacokinetics , Thiophenes/therapeutic use
19.
Fundam Clin Pharmacol ; 21(3): 265-72, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17521295

ABSTRACT

To study the contractile responses of the human basilar artery to 5-hydroxytryptamine (5-HT), sumatriptan, zolmitriptan and naratriptan, and to characterize the 5-HT receptor subtypes involved on those responses, human basilar artery rings were prepared for isometric contraction, protein isolation and Western blotting analysis. Concentration-response (CR) curves were made for all agonists in the absence or in the presence of selective antagonists at 5-HT1B (cyanopindolol), 5-HT1D (BRL 15,572) and 5-HT2 (ketanserin) receptors. We also used anti-5-HT1B and 5-HT1D receptor antibodies to search for the expression of protein of these receptor subtypes. From the CR curves, the relative intrinsic activity and potency of these agonists were determined. The ranking order for the intrinsic activity was 5-HT > or = sumatriptan > zolmitriptan > or = naratriptan, whereas that for the potency was zolmitriptan > or = 5-HT > or = sumatriptan > naratriptan. Our results also show that the human basilar artery seems to have a mixed population of 5-HT1B/1D receptors mediating the contractile response to triptans, which is also suggested by the expression of both receptor subtypes. There is also a population of 5-HT2 receptors for which the antimigraine drugs used have no apparent affinity. From this study, one can conclude that the second generation triptans have lower contractile capacity than sumatriptan, suggesting that they have a better cerebrovascular safety profile.


Subject(s)
Basilar Artery/drug effects , Oxazolidinones/pharmacology , Piperidines/pharmacology , Serotonin/pharmacology , Sumatriptan/pharmacology , Tryptamines/pharmacology , Vasoconstrictor Agents/pharmacology , Basilar Artery/physiology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Receptor, Serotonin, 5-HT1B/metabolism , Receptor, Serotonin, 5-HT1D/metabolism , Serotonin Receptor Agonists/pharmacology , Vasoconstriction/drug effects
20.
Ann N Y Acad Sci ; 1074: 466-77, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17105945

ABSTRACT

To assess whether chronic heroin abuse may generate vascular central nervous deficits, we studied the profile of vascular alterations in 17 heroin addicts (14 males mean age 31 years, range 23-39 years and 3 females mean age 33 years, range 30-35 years) before and, in one of them, 10 weeks after an ultra-rapid heroin detoxification. Using the functional technique of single-photon emission tomography (SPET) with 740 MBq of (99m)Tc-hexametazine (HMPAO) and computational brain-mapping techniques by means of a Talairach analysis, we determined the pattern of vascular brain alterations associated with chronic heroin abuse. Compared with controls, subjects who had used heroin chronically showed a decrease of global brain perfusion that was more significant in the frontal cortex-mainly in orbito-frontal regions, as well as in the occipital and temporal lobes. All patients showed marked asymmetric perfusion of the basal ganglia and the majority of them showed also an asymmetric perfusion of cerebellum. In addition, there were small activated areas dispersed in the occipital lobe (3 of 17) and apex region (4 of 17). In conclusion, decreased perfusion in heroin addicts was found in regions involved in the control of attention, motor speed, memory and visual-spatial processing. The prefrontal cortex is involved in decision making and inhibitory control, processes disturbed in heroin addicts who have stopped heroin consumption. A reduction in regional perfusion may reflect ongoing subtle neurocognitive deficits, which are consistent with the maintenance of asymmetry of the basal nuclei.


Subject(s)
Brain/blood supply , Brain/ultrastructure , Heroin Dependence/pathology , Heroin/adverse effects , Tomography, Emission-Computed, Single-Photon/methods , Adult , Cohort Studies , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...