Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Eur J Neurosci ; 59(10): 2450-2464, 2024 May.
Article in English | MEDLINE | ID: mdl-38480476

ABSTRACT

Amphetamine (AMPH) exposure induces behavioural and neurochemical sensitization observed in rodents as hyperlocomotion and increased dopamine release in response to a subsequent dose. Brain Angiotensin II modulates dopaminergic neurotransmission through its AT1 receptors (AT1-R), positively regulating striatal dopamine synthesis and release. This work aims to evaluate the AT1-R role in the development and maintenance of AMPH-induced sensitization. Also, the AT1-R involvement in striatal dopamine reuptake was analysed. The sensitization protocol consisted of daily AMPH administration for 5 days and tested 21 days after withdrawal. An AT1-R antagonist, candesartan, was administered before or after AMPH exposure to evaluate the participation of AT1-R in the development and maintenance of sensitization, respectively. Sensitization was evaluated by locomotor activity and c-Fos immunostaining. Changes in dopamine reuptake kinetics were evaluated 1 day after AT1-R blockade withdrawal treatment, with or without the addition of AMPH in vitro. The social interaction test was performed as another behavioural output. Repeated AMPH exposure induced behavioural and neurochemical sensitization, which was prevented and reversed by candesartan. The AT1-R blockade increased the dopamine reuptake kinetics. Neither the AMPH administration nor the AT1-R blockade altered the performance of social interaction. Our results highlight the AT1-R's crucial role in AMPH sensitization. The enhancement of dopamine reuptake kinetics induced by the AT1-R blockade might attenuate the neuroadaptive changes that lead to AMPH sensitization and its self-perpetuation. Therefore, AT1-R is a prominent candidate as a target for pharmacological treatment of pathologies related to dopamine imbalance, including drug addiction and schizophrenia.


Subject(s)
Amphetamine , Angiotensin II Type 1 Receptor Blockers , Angiotensin II , Benzimidazoles , Biphenyl Compounds , Corpus Striatum , Dopamine , Animals , Amphetamine/pharmacology , Male , Dopamine/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Angiotensin II/pharmacology , Biphenyl Compounds/pharmacology , Benzimidazoles/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Rats, Wistar , Rats , Receptor, Angiotensin, Type 1/metabolism , Tetrazoles/pharmacology , Central Nervous System Stimulants/pharmacology , Social Interaction/drug effects , Motor Activity/drug effects , Proto-Oncogene Proteins c-fos/metabolism
2.
Eur J Neurosci ; 54(5): 5705-5716, 2021 09.
Article in English | MEDLINE | ID: mdl-32320503

ABSTRACT

Astrocytes play an essential role in the genesis, maturation and regulation of the neurovascular unit. Multiple evidence support that astrocyte reactivity has a close relationship to neurovascular unit dysfunction, oxidative stress and inflammation, providing a suitable scenario for the development of mental disorders. Ketamine has been proposed as a single-use antidepressant treatment in major depression, and its antidepressant effects have been associated with anti-inflammatory properties. However, Ketamine long-lasting effects over the neurovascular unit components remain unclear. Angiotensin II AT1 receptor (AT1 -R) blockers have anti-inflammatory, antioxidant and neuroprotective effects. The present work aims to distinguish the acute and long-term Ketamine effects over astrocytes response extended to other neurovascular unit components, and the involvement of AT1 -R, in prefrontal cortex and ventral tegmental area. Male Wistar rats were administered with AT1 -R antagonist Candesartan/Vehicle (days 1-10) and Ketamine/Saline (days 6-10). After 14 days drug-free, at basal conditions or after Ketamine Challenge, the brains were processed for oxidative stress analysis, cresyl violet staining and immunohistochemistry for glial, neuronal activation and vascular markers. Repeated Ketamine administration induced long-lasting region-dependent astrocyte reactivity and morphological alterations, and neuroadaptative changes observed as exacerbated oxidative stress and neuronal activation, prevented by the AT1 -R blockade. Ketamine Challenge decreased microglial and astrocyte reactivity and augmented cellular apoptosis, independently of previous treatment. Overall, AT1 -R is involved in the development of neuroadaptative changes induced by repeated Ketamine administration but does not interfere with the acute effects supporting the potential use of AT1 -R blockers as a Ketamine complementary therapy in mental disorders.


Subject(s)
Astrocytes , Ketamine , Angiotensin II Type 1 Receptor Blockers , Animals , Ketamine/toxicity , Male , Oxidative Stress , Rats , Rats, Wistar
3.
Eur J Neurosci ; 51(4): 1026-1041, 2020 02.
Article in English | MEDLINE | ID: mdl-31646669

ABSTRACT

Amphetamine-induced neuroadaptations involve vascular damage, neuroinflammation, a hypo-functioning prefrontal cortex (PFC), and cognitive alterations. Brain angiotensin II, through angiotensin type 1 receptor (AT1 -R), mediates oxidative/inflammatory responses, promoting endothelial dysfunction, neuronal oxidative damage and glial reactivity. The present work aims to unmask the role of AT1 -R in the development of amphetamine-induced changes over glial and vascular components within PFC and hippocampus. Attention deficit was evaluated as a behavioral neuroadaptation induced by amphetamine. Brain microvessels were isolated to further evaluate vascular alterations after amphetamine exposure. Male Wistar rats were administered with AT1 -R antagonist, candesartan, followed by repeated amphetamine. After one week drug-off period, animals received a saline or amphetamine challenge and were evaluated in behavioral tests. Afterward, their brains were processed for cresyl violet staining, CD11b (microglia marker), GFAP (astrocyte marker) or von Willebrand factor (vascular marker) immunohistochemistry, and oxidative/cellular stress determinations in brain microvessels. Statistical analysis was performed by using factorial ANOVA followed by Bonferroni or Tukey tests. Repeated amphetamine administration increased astroglial and microglial markers immunoreactivity, increased apoptotic cells, and promoted vascular network rearrangement at the PFC concomitantly with an attention deficit. Although the amphetamine challenge improved the attentional performance, it triggers detrimental effects probably because of the exacerbated malondialdehyde levels and increased heat shock protein 70 expression in microvessels. All observed amphetamine-induced alterations were prevented by the AT1 -R blockade. Our results support the AT1 -R involvement in the development of oxidative/inflammatory conditions triggered by amphetamine exposure, affecting cortical areas and increasing vascular susceptibility to future challenges.


Subject(s)
Amphetamine , Receptor, Angiotensin, Type 1 , Amphetamine/toxicity , Angiotensin II , Animals , Brain/metabolism , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism
4.
Neuropsychobiology ; 79(3): 191-197, 2020.
Article in English | MEDLINE | ID: mdl-31927553

ABSTRACT

INTRODUCTION: In recent years, an important number of studies have emphasized the psychopharmacological actions of cycloleucine (1-aminocyclopentanecarboxylic acid) acting on the NR1 subunit (glycine allosteric site) of NMDA (N-methyl-D-aspartic acid) receptor. We studied the effects of its injection in an anxiety test. METHODS: The elevated plus maze test was used. Male rats bilaterally cannulated into the nucleus accumbens septi (NAS) were employed. Rats were divided into 5 groups that received either 1 µL injections of saline or cycloleucine (0.5, 1, 2, or 4 µg) 15 min before testing. RESULTS: Time spent in the open arm was significantly increased by cycloleucine treatment with all doses (1 and 2 µg, p < 0.05; 0.5 and 4 µg, p < 0.01), like number of extreme arrivals (0.5 and 1 µg, p < 0.05; 2 µg, p < 0.01; and 4 µg, p < 0.001). Open arm entries were increased by the highest dose only (4 µg, p < 0.01). DISCUSSION/CONCLUSION: Present results show no difference between all doses in the time spent in the open arm, suggesting an indirect, noncompetitive action of the drug. The increase in extreme arrivals and open arm entries suggests a dose influence in these parameters. We conclude that cycloleucine influence on the NMDA receptors within NAS leads to anxiolytic-like effects and behavioral disinhibition, which once more confirms the involvement of NAS in anxiety processing.


Subject(s)
Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Cycloleucine/pharmacology , Elevated Plus Maze Test , Nucleus Accumbens/drug effects , Psychomotor Performance/drug effects , Animals , Anti-Anxiety Agents/administration & dosage , Cycloleucine/administration & dosage , Rats
5.
Eur J Neurosci ; 45(12): 1586-1593, 2017 06.
Article in English | MEDLINE | ID: mdl-28449313

ABSTRACT

The use of psychostimulants, such as amphetamine (Amph), is associated with inflammatory processes, involving glia and vasculature alterations. Brain Angiotensin II (Ang II), through AT1 -receptors (AT1 -R), modulates neurotransmission and plays a crucial role in inflammatory responses in brain vasculature and glia. Our aim for the present work was to evaluate the role of AT1 -R in long-term alterations induced by repeated exposure to Amph. Astrocyte reactivity, neuronal survival and brain microvascular network were analysed at the somatosensory cortex. Thermal nociception was evaluated as a physiological outcome of this brain area. Male Wistar rats (250-320 g) were administered with AT1 -R antagonist Candesartan/vehicle (3 mg/kg p.o., days 1-5) and Amph/saline (2.5 mg/kg i.p., days 6-10). The four experimental groups were: Veh-Sal, CV-Sal, Veh-Amph, CV-Amph. On day 17, the animals were sacrificed and their brains were processed for Nissl staining and immunohistochemistry against glial fibrillary acidic protein (GFAP) and von Willebrand factor. In another group of animals, thermal nociception was evaluated using hot plate test, in the four experimental groups, on day 17. Data were analysed with two-way anova followed by Bonferroni test. Our results indicate that Amph exposure induces an increase in: neuronal apoptosis, astrocyte reactivity and microvascular network, evaluated as an augmented occupied area by vessels, branching points and their tortuosity. Moreover, Amph exposure decreased the thermal nociception threshold. Pretreatment with the AT1 -R blocker prevented the described alterations induced by this psychostimulant. The decreased thermal nociception and the structural changes in somatosensory cortex could be considered as extended neuroadaptative responses to Amph, involving AT1 -R activation.


Subject(s)
Amphetamine/pharmacology , Central Nervous System Stimulants/pharmacology , Nociception , Receptor, Angiotensin, Type 1/metabolism , Somatosensory Cortex/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Astrocytes/metabolism , Benzimidazoles/pharmacology , Biphenyl Compounds , Glial Fibrillary Acidic Protein/metabolism , Hot Temperature , Male , Microvessels/drug effects , Microvessels/metabolism , Neurons/metabolism , Rats , Rats, Wistar , Somatosensory Cortex/drug effects , Somatosensory Cortex/physiology , Tetrazoles/pharmacology , von Willebrand Factor/metabolism
6.
Med Biol Eng Comput ; 60(10): 2995-3007, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36018532

ABSTRACT

Computerized techniques for image analysis are critical for progress in cell biology. The complexity of the data in current methods eliminates the need for manual image analysis and usually requires the application of multiple algorithms sequentially to the images. Our aim was to develop a software for immunohistochemical analysis of brain dopaminergic neurons combining several computational approaches to automatically analyze and quantify their number in the substantia nigra after a neurotoxic injury. For this purpose, we used a Parkinson's disease animal model to test our application. The dopaminergic neurotoxin, 6-hydroxydopamine, was administered in adult male rats to damage dopaminergic neurons in substantia nigra and to induce hemiparkinsonism. The lesion was corroborated by behavioral evaluation in response to apomorphine and amphetamine. The animals were euthanized and their brains processed for tyrosine hydroxylase immunohistochemistry for dopamine neuron identification. Neurons positive for tyrosine hydroxylase were evaluated in substantia nigra by light microscopy. The images were used to show quantification applicability. To test our software counting accuracy and validity, automatic dopamine neuron number was correlated with the data obtained by three independent observers. Several parameters were used to depict neuronal function in dataset images from control and lesioned brains. In conclusion, we could perform an automated quantification of dopaminergic neurons and corroborate the validity and accuracy of a freely available software.


Subject(s)
Dopaminergic Neurons , Tyrosine 3-Monooxygenase , Animals , Dopaminergic Neurons/metabolism , Male , Oxidopamine/toxicity , Rats , Software , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
7.
Behav Brain Res ; 425: 113809, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35218792

ABSTRACT

Schizophrenia is a chronic disease affecting 1% worldwide population, of which 30% are refractory to the available treatments: thus, searching for new pharmacological targets is imperative. The acute and repeated ketamine administration are validated preclinical models that recreate the behavioral and neurochemical features of this pathology, including the parvalbumin-expressing interneurons dysfunction. Angiotensin II, through AT1 receptors (AT1-R), modulates the dopaminergic and GABAergic neurotransmission. We evaluated the AT1-R role in the long-term neuronal activation and behavioral alterations induced by repeated ketamine administration. Adult male Wistar rats received AT1-R antagonist candesartan/vehicle (days 1-10) and ketamine/saline (days 6-10). After 14 days of drug-free, neuronal activation and behavioral analysis were performed. Locomotor activity, social interaction and novel object recognition tests were assessed at basal conditions or after ketamine challenge. Immunostaining for c-Fos, GAD67 and parvalbumin were assessed after ketamine challenge in cingulate, insular, piriform, perirhinal, and entorhinal cortices, striatum, and hippocampus. Additionally, to evaluate the AT1-R involvement in acute ketamine psychotomimetic effects, the same behavioral tests were performed after 6 days of daily-candesartan and a single-ketamine administration. We found that ketamine-induced long-lasting schizophrenia-like behavioral alterations, and regional-dependent neuronal activation changes, involving the GABAergic neurotransmission system and the parvalbumin-expressing interneurons, were AT1-R-dependent. The AT1-R were not involved in the acute ketamine psychotomimetic effects. These results add new evidence to the wide spectrum of action of ketamine and strengthen the AT1-R involvement in endurable alterations induced by psychostimulants administration, previously proposed by our group, as well as their preponderant role in the development of psychiatric pathologies.


Subject(s)
Ketamine , Receptor, Angiotensin, Type 1 , Schizophrenia , Angiotensin II , Animals , Male , Parvalbumins , Rats , Rats, Wistar , Schizophrenia/chemically induced
8.
Front Pharmacol ; 12: 647747, 2021.
Article in English | MEDLINE | ID: mdl-34012397

ABSTRACT

Background: Amphetamine (AMPH) alters neurons, glia and microvessels, which affects neurovascular unit coupling, leading to disruption in brain functions such as attention and working memory. Oxidative stress plays a crucial role in these alterations. The angiotensin type I receptors (AT1-R) mediate deleterious effects, such as oxidative/inflammatory responses, endothelial dysfunction, neuronal oxidative damage, alterations that overlap with those observed from AMPH exposure. Aims: The aim of this study was to evaluate the AT1-R role in AMPH-induced oxidative stress and glial and vascular alterations in the prefrontal cortex (PFC). Furthermore, we aimed to evaluate the involvement of AT1-R in the AMPH-induced short-term memory and working memory deficit. Methods: Male Wistar rats were repeatedly administered with the AT1-R blocker candesartan (CAND) and AMPH. Acute oxidative stress in the PFC was evaluated immediately after the last AMPH administration by determining lipid and protein peroxidation. After 21 off-drug days, long-lasting alterations in the glia, microvessel architecture and to cognitive tasks were evaluated by GFAP, CD11b and von Willebrand immunostaining and by short-term and working memory assessment. Results: AMPH induced acute oxidative stress, long-lasting glial reactivity in the PFC and a working memory deficit that were prevented by AT1-R blockade pretreatment. Moreover, AMPH induces transient angiogenesis in PFC via AT1-R. AMPH did not affect short-term memory. Conclusion: Our results support the protective role of AT1-R blockade in AMPH-induced oxidative stress, transient angiogenesis and long-lasting glial activation, preserving working memory performance.

9.
Neuroendocrinology ; 88(4): 276-86, 2008.
Article in English | MEDLINE | ID: mdl-18679017

ABSTRACT

We studied the effect of ovariectomy and estrogen replacement on expression of adrenal angiotensin II AT1 and AT2 receptors, aldosterone content, catecholamine synthesis, and the transcription factor Fos-related antigen 2 (Fra-2). Ovariectomy increased AT1 receptor expression in the adrenal zona glomerulosa and medulla, and decreased adrenomedullary catecholamine content and Fra-2 expression when compared to intact female rats. In the zona glomerulosa, estrogen replacement normalized AT1 receptor expression, decreased AT1B receptor mRNA, and increased AT2 receptor expression and mRNA. Estrogen treatment decreased adrenal aldosterone content. In the adrenal medulla, the effects of estrogen replacement were: normalized AT1 receptor expression, increased AT2 receptor expression, AT2 receptor mRNA, and tyrosine hydroxylase mRNA, and normalized Fra-2 expression and catecholamine content. We demonstrate that the constitutive adrenal expression of AT1 receptors, catecholamine synthesis and Fra-2 expression are partially under the control of reproductive hormones. Our results suggest that estrogen treatment decreases aldosterone production through AT1 receptor downregulation and AT2 receptor upregulation. AT2 receptor upregulation and modulation of Fra-2 expression may participate in the estrogen-dependent normalization of adrenomedullary catecholamine synthesis in ovariectomized rats. The AT2 receptor upregulation and the decrease in AT1 receptor function and in the production of the fluid-retentive, pro-inflammatory hormone aldosterone partially explain the protective effects of estrogen therapy.


Subject(s)
Adrenal Medulla/metabolism , Aldosterone/metabolism , Estrogens/pharmacology , Fos-Related Antigen-2/metabolism , Ovariectomy , Receptor, Angiotensin, Type 2/metabolism , Zona Glomerulosa/metabolism , Adrenal Medulla/drug effects , Animals , Catecholamines/metabolism , Estrogen Replacement Therapy , Female , Fos-Related Antigen-2/drug effects , Models, Animal , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/drug effects , Tyrosine 3-Monooxygenase/metabolism , Zona Glomerulosa/drug effects
10.
J Basic Clin Physiol Pharmacol ; 29(3): 241-246, 2018 Jun 27.
Article in English | MEDLINE | ID: mdl-29902911

ABSTRACT

BACKGROUND: In previous studies, we have observed that specific N-methyl-d-aspartic acid (NMDA) antagonists and non-NMDA antagonists injected within the nucleus accumbens septi (NAS) induced an anxiolytic-like effect in the plus maze test in rats. In the present study, the effect of intracanalicular blockade of NMDA receptors using dizocilpine in the plus maze was studied in male rats bilaterally cannulated NAS. METHODS: Rats were divided into five groups that received either 1 µL injections of saline or dizocilpine (MK-801, [5R,10S]-[+]-5-methyl-10,11-dihydro-5H-dibenzo [a,d] cyclohepten-5,10-imine) in different doses (0.5, 1, 2, or 4 µg) 15 min before testing. RESULTS: Time spent in the open arm increased under dizocilpine treatment with the two higher doses (2 and 4 µg, p<0.05), extreme arrivals were increased by the three higher doses (1 µg, p<0.05; 2 and 4 µg, p<0.01), and open arm entries by the three higher doses (1, 2, and 4 µg, p<0.05). A dose-effect relationship was observed in all cases. CONCLUSIONS: We conclude that dizocilpine-glutamatergic blockade in the accumbens lead to an anxiolytic-like effect and a behavioral disinhibition related to an increase in some motoric parameters, showing specific behavioral patterns.


Subject(s)
Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Maze Learning/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Behavior, Animal/drug effects , Dizocilpine Maleate/administration & dosage , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/administration & dosage , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
11.
Eur J Pharmacol ; 568(1-3): 186-91, 2007 Jul 30.
Article in English | MEDLINE | ID: mdl-17537427

ABSTRACT

Excessive grooming behaviour is induced by intracerebroventricular injections of the neuropeptide glutamic acid isoleucine amide (neuropeptide-EI), via the activation of A-10 dopaminergic neurons and the noradrenergic system. Our object was to study the latter system involved in these behaviours, using male Wistar rats weighing 250-300 g with i.c.v. implants. The results show that all the adrenoceptor antagonists "per se" do not affect excessive grooming behaviour or motor activity. Intracerebroventricular administration of propranolol, a general beta-adrenoceptor antagonist, before neuropeptide-EI, inhibited the induced excessive grooming behaviour in a dose dependent manner. Metoprolol, a beta(1)-adrenoceptor antagonist, also blocked this behaviour. However, intracerebroventricular injections of phentolamine, an alpha-adrenoceptor antagonist, and ((+/-)-1-[2,3-(Dihydro-7-methyl-1H-inden-4-yl)oxy]-3-[(1-methylethyl)amino]-2-butanol), a beta(2)-adrenoceptor antagonist, had no effect on the behaviour induced by neuropeptide-EI induced behaviour for any of the doses tested. On the other hand, isoproterenol, a general beta-adrenoceptor agonist and dobutamine, a beta(1)-adrenoceptor agonist, both elicited similar behaviours as those induced by neuropeptide-EI. These results support the hypothesis that a relationship exists between neuropeptide-EI and beta-adrenoceptors, more specifically the beta(1)-adrenoceptor, as found with other similar endogenous peptides such as neurotensin, cholecystin, substance P and alpha-melanocyte stimulating hormone. Hence, neuropeptide-EI could probably be exerting a neuromodulating effect on the central nervous system.


Subject(s)
Grooming/drug effects , Motor Activity/drug effects , Oligopeptides/pharmacology , Receptors, Adrenergic, beta-1/metabolism , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-1 Receptor Agonists , Adrenergic beta-1 Receptor Antagonists , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Behavior, Animal/drug effects , Dobutamine/pharmacology , Isoproterenol/pharmacology , Male , Metoprolol/pharmacology , Phentolamine/pharmacology , Propranolol/pharmacology , Rats , Rats, Wistar
12.
Protein Pept Lett ; 24(9): 817-826, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-28758591

ABSTRACT

BACKGROUND: The functioning of the central nervous system is complex and it implies tight and coordinated interactions among multiple components. Neurotransmitters systems imbalance is a hallmark in the central nervous system (CNS) disorders. These pathologies profoundly impact the social, cultural, and economic perspective worldwide. The etiopathology of CNS illnesses is still poorly understood, making their treatment difficult. Brain angiotensin II (Ang II), through its AT1 receptors, modulates dopaminergic, glutamatergic and GABAergic neurotransmission, which are responsible for movement control, cognition, emotions and stress responses. Alterations of these functions, concomitant with modified brain reninangiotensin system (RAS) components, have been described in CNS pathologies like depression, Parkinson, Alzheimer, and schizophrenia. In this sense, altered functionality of angiotensin I converting enzyme and AT1 receptors, is associated with augmented susceptibility to the occurrence of these pathologies. Moreover, some epidemiological data showed lower incidence of Alzheimer disease in hypertensive patients under treatment targeting RAS; meanwhile preclinical studies relate RAS with Parkinson and depression. Little is known about schizophrenia and RAS; however, Ang II is closely related to dopamine and glutamate pathways, which are mainly altered in this pathology. CONCLUSION: The available evidences, together with the results obtained by our group, open the possibility to postulate brain Ang II as a possible therapeutic target to treat the above-mentioned CNS disorders.


Subject(s)
Angiotensin II/metabolism , Mental Disorders/drug therapy , Animals , Chronic Disease , Humans , Mental Disorders/metabolism , Molecular Targeted Therapy , Peptidyl-Dipeptidase A/metabolism , Receptor, Angiotensin, Type 1/metabolism
13.
Psychopharmacology (Berl) ; 233(5): 795-807, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26613735

ABSTRACT

RATIONALE: Angiotensin II, by activation of its brain AT1-receptors, plays an active role as neuromodulator in dopaminergic transmission. These receptors participate in the development of amphetamine-induced behavioral and dopamine release sensitization. Dopamine is involved in cognitive processes and provides connectivity between brain areas related to these processes. Amphetamine by its mimetic activity over dopamine neurotransmission elicits differential responses after acute administration or after re-exposure following long-term withdrawal periods in different cognitive processes. OBJECTIVE: The purpose of this study is to evaluate the AT1-receptor involvement in the acute and long-term amphetamine-induced alterations in long-term memory and in cellular-related events. METHODS: Male Wistar rats (250-300 g) were used in this study. Acute effects: Amphetamine (0.5/2.5 mg/kg i.p.) was administered after post-training in the inhibitory avoidance (IA) response. The AT1-receptor blocker Losartan was administered i.c.v. before a single dose of amphetamine (0.5 mg/kg i.p.). Long-term effects: The AT1-receptors blocker Candesartan (3 mg/kg p.o.) was administered for 5 days followed by 5 consecutive days of amphetamine (2.5 mg/kg/day, i.p.). The neuroadaptive changes were evidenced after 1 week of withdrawal by an amphetamine challenge (0.5 mg/kg i.p.). The IA response, the neuronal activation pattern, and the hippocampal synaptic transmission were evaluated. RESULTS: The impairing effect in the IA response of post-training acute amphetamine was partially prevented by Losartan. The long-term changes induced by repeated amphetamine (resistance to acute amphetamine interference in the IA response, neurochemical altered response, and increased hippocampal synaptic transmission) were prevented by AT1-receptors blockade. CONCLUSIONS: AT1-receptors are involved in the acute alterations and in the neuroadaptations induced by repeated amphetamine associated with neurocognitive processes.


Subject(s)
Amphetamines/toxicity , Central Nervous System Stimulants/toxicity , Cognition Disorders/chemically induced , Receptor, Angiotensin, Type 1/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Avoidance Learning , Behavior, Animal/drug effects , Benzimidazoles/pharmacology , Biphenyl Compounds , Cognition Disorders/physiopathology , Hippocampus/drug effects , Injections, Intraventricular , Losartan/pharmacology , Male , Memory, Long-Term/drug effects , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Tetrazoles/pharmacology
14.
Article in Spanish | BINACIS | ID: biblio-1379404

ABSTRACT

La inteligencia artificial tiene el potencial de transformar la forma en que se brinda la atención médica. Puede respaldar mejoras en los resultados y aumentar la productividad y la eficiencia de la prestación de los servicios. En servicios de las diferentes especialidades los avances realizados a nivel hardware deben desarrollarse en paralelo con los métodos de aprendizaje automático, aspectos que la inteligencia artificial contribuye para promover un cambio de paradigma significativo en las más diversas áreas de la medicina. Es importante en la educación médica como eje para el conocimiento y en la toma de decisiones que pueden mejorar el desempeño de los profesionales. Los estudiantes de medicina de nueva generación pueden adaptarse perfectamente a los nuevos métodos digitalizados en un contexto médico globalizado, incluida la inteligencia artificial. Por ello es importante tener como objetivos a implementar en los planes de estudio e introducir programas educativos representativos de esta tecnología. Es fundamental que todas las áreas del Sistema de Salud tengan confianza en los sistemas informáticos específicamente en el aprendizaje profundo, no solo por la información concreta y objetiva que de él se deriva sino también por la posibilidad de predecir eventos futuros, brindando alta certeza en cuanto al diagnóstico y tratamiento de enfermedades.


Artificial intelligence has the potential to transform the way healthcare is delivered. You can support improved results and increase the productivity and efficiency of service delivery. In the services of the different specialties, the advances made at the hardware level must be developed in parallel with the methods of machine learning, aspects that artificial intelligence contributes to promote a significant paradigm shift in the most diverse areas of medicine. It is important in medical education as an axis for knowledge and in making decisions that can improve the performance of professionals. New generation medical students can perfectly adapt to new digitized methods in a globalized medical context, including artificial intelligence. For this reason, it is important to have as objectives to be implemented in the study plans and to introduce educational programs that are representative of this technology. It is essential that all areas of the Health System have confidence in computer systems specifically in deep learning, not only because of the concrete and objective information that is derived from it but also because of the possibility of predicting future events, providing high certainty regarding to the diagnosis and treatment of diseases.


Subject(s)
Technology , Artificial Intelligence , Education, Medical , Medical Care , Diagnosis
15.
Regul Pept ; 124(1-3): 7-17, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15544836

ABSTRACT

We studied renal AT1 and AT2 receptors in male, female, ovariectomized and ovariectomized-estrogen-treated Wistar-Hanover and Wistar-Kyoto rats. AT1 receptors and AT1A receptor mRNA predominated, with no significant differences between males and females. AT2 receptor expression was restricted in female rats to the capsule, the transition zone between outer and inner medulla, the endothelium lining the papilla, and arcuate arteries and veins. There were no AT2 receptors in male rats, while male mice express substantial numbers of estrogen-dependent AT2 receptors. Arcuate arteries and veins expressed AT1B mRNA in males and females, and AT2 mRNA in females only. AT1 receptor and AT2 receptor expression were estrogen-dependent, with increases in AT1 and AT2 receptor expression after estrogen treatment in ovariectomized rats. Estrogen treatment increased prostaglandin E2 (PGE2) and cGMP concentrations in the renal medulla, and eNOS expression in cortical arteries. In rodents, expression of renal Angiotensin II receptor types is estrogen-dependent, with significant species, strain and area differences. Our results support an important role for AT2 receptors in the regulation of renal function and in the protective effects of estrogen in the kidney.


Subject(s)
Estrogens/pharmacology , Kidney/drug effects , Kidney/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Animals , Autoradiography , Cyclic GMP/metabolism , Female , Gene Expression Regulation , Immunohistochemistry , In Situ Hybridization , Kidney/blood supply , Male , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type III , Ovariectomy , RNA, Messenger/genetics , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Sex Characteristics , Up-Regulation
16.
Regul Pept ; 128(3): 227-38, 2005 Jun 30.
Article in English | MEDLINE | ID: mdl-15837532

ABSTRACT

The brain and the peripheral (hormonal) angiotensin II systems are stimulated during stress. Activation of brain angiotensin II AT(1) receptors is required for the stress-induced hormone secretion, including CRH, ACTH, corticoids and vasopressin, and for stimulation of the central sympathetic activity. Long-term peripheral administration of the angiotensin II AT(1) antagonist candesartan blocks not only peripheral but also brain AT(1) receptors, prevents the hormonal and sympathoadrenal response to isolation stress and prevents the formation of stress-induced gastric ulcers. The mechanisms responsible for the prevention of stress-induced ulcers by the AT(1) receptor antagonist include protection from the stress-induced ischemia and inflammation (neutrophil infiltration and increase in ICAM-1 and TNF-alpha) in the gastric mucosa and a partial blockade of the stress-induced sympathoadrenal stimulation, while the protective effect of the glucocorticoid release during stress is maintained. AT(1) receptor antagonism prevents the stress-induced decrease in cortical CRH(1) and benzodiazepine binding and is anxiolytic. Blockade of brain angiotensin II AT(1) receptors offers a novel therapeutic opportunity for the treatment of anxiety and other stress-related disorders.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Anti-Anxiety Agents/therapeutic use , Receptor, Angiotensin, Type 1/physiology , Stress, Psychological/prevention & control , Angiotensin II/physiology , Brain/physiology , Brain/physiopathology , Humans , Hypertension/physiopathology
17.
Ann N Y Acad Sci ; 1018: 131-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15240361

ABSTRACT

A 2-week pretreatment with an Angiotensin II AT(1) antagonist prevented the adrenomedullary and hormonal response to isolation stress. We studied the effect of life-long treatment with the AT(1) receptor antagonist candesartan, 10 mg/kg/day, or vehicle administered orally in the drinking water from 8 weeks of age on the response to stress of stress-sensitive spontaneously hypertensive rats (SHRs) and their normotensive controls, the Wistar Kyoto (WKY). Rats were submitted to 24-h isolation stress at different times during the treatment. Treatment with candesartan extended the lifespan of SHRs. AT(1) receptor blockade retained its capacity to blunt the response to isolation stress over a long period of treatment. The AT(1) antagonist inhibited epinephrine release in SHR but not in WKY rats during the first 3 months, corticosterone release in SHR and WKY rats during 10 months, and vasopressin release in SHR rats during 18 months of treatment when rats were submitted to isolation stress. There were no changes in vasopressin release in WKY rats during stress or after AT(1) receptor blockade. We conclude that the blockade of the stress response by the AT(1) receptor antagonist is long lasting and differs between stress-prone SHR and WKY rats and that the specific components of the stress response (sympathoadrenal activity, hypothalamic-pituitary-adrenal axis activation, and vasopressin release) react differently to AT(1) receptor blockade. The long-term protective effects of AT(1) receptor blockade can be important in animals vulnerable to stress and, in conjunction with the normalization of blood pressure, can prolong lifespan through end-organ protection.


Subject(s)
Angiotensin II/antagonists & inhibitors , Angiotensin Receptor Antagonists , Catecholamines/metabolism , Glucocorticoids/metabolism , Stress, Physiological/metabolism , Vasopressins/metabolism , Angiotensin II/metabolism , Animals , Male , Rats , Rats, Inbred SHR , Receptors, Angiotensin/metabolism
18.
Brain Res ; 1028(1): 9-18, 2004 Nov 26.
Article in English | MEDLINE | ID: mdl-15518636

ABSTRACT

Peripheral and brain angiotensin II AT(1) receptor blockade decreases high blood pressure, stress, and neuronal injury. To clarify the effects of long-term brain Ang II receptor blockade, the AT(1) blocker, candesartan, was orally administered to spontaneously hypertensive rats (SHRs) for 40 days, followed by intraventricular injection of 25 ng of Ang II. Before Ang II injection, AT(1) receptor blockade normalized blood pressure and decreased plasma adrenocorticotropic hormone (ACTH) and corticosterone. After central administration of excess Ang II, the reduction of ACTH and corticosterone release induced by AT(1) receptor blockade no longer occurred. Central Ang II administration to vehicle-treated SHRs further increased blood pressure, provoked drinking, increased tyrosine hydroxylase (TH) mRNA expression in the locus coeruleus, and stimulated sympathoadrenal catecholamine release. Pretreatment with the AT(1) receptor antagonist eliminated Ang II-induced increases in blood pressure, water intake, and sympathoadrenal catecholamine release; inhibited peripheral and brain AT(1) receptors; increased AT(2) receptor binding in the locus coeruleus, inferior olive, and adrenal cortex; and decreased AT(2) receptor binding in the adrenal medulla. Inhibition of brain AT(1) receptors correlated with decreased TH transcription in the locus coeruleus, indicating a decreased central sympathetic drive. This, and the diminished adrenomedullary AT(1) and AT(2) receptor stimulation, result in decreased sympathoadrenomedullary stimulation. Oral administration of AT(1) antagonists can effectively block central actions of Ang II, regulating blood pressure and reaction to stress, and selectively and differentially modulating sympathoadrenal response and the hypothalamic-pituitary-adrenal stimulation produced by brain Ang II--effects of potential therapeutic importance.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II/administration & dosage , Benzimidazoles/administration & dosage , Blood Pressure/drug effects , Locus Coeruleus/drug effects , Receptor, Angiotensin, Type 1/drug effects , Tetrazoles/administration & dosage , Administration, Oral , Adrenal Glands/drug effects , Adrenocorticotropic Hormone/blood , Analysis of Variance , Animals , Biphenyl Compounds , Catechols/blood , Corticosterone/blood , Drug Administration Schedule , Drug Interactions , Injections, Intraventricular , Locus Coeruleus/metabolism , Male , RNA, Messenger/analysis , Rats , Rats, Inbred SHR , Receptor, Angiotensin, Type 1/physiology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
Behav Brain Res ; 272: 314-23, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25046593

ABSTRACT

A single or repeated exposure to psychostimulants induces long-lasting neuroadaptative changes. Different neurotransmitter systems are involved in these responses including the neuropeptide angiotensin II. Our study tested the hypothesis that the neuroadaptative changes induced by amphetamine produce alterations in brain RAS components that are involved in the expression of the locomotor sensitization to the psychostimulant drug. Wistar male rats, pretreated with amphetamine were used 7 or 21 days later to study AT1 receptors by immunohistochemistry and western blot and also angiotensinogen mRNA and protein in caudate putamen and nucleus accumbens. A second group of animals was used to explore the possible role of Ang II AT1 receptors in the expression of behavioral sensitization. In these animals treated in the same way, bearing intra-cerebral cannula, the locomotor activity was tested 21 days later, after an amphetamine challenge injection and the animals received an AT1 blocker, losartan, or saline 5min before the amphetamine challenge. An increase of AT1 receptor density induced by amphetamine was found in both studied areas and a decrease in angiotensinogen mRNA and protein only in CPu at 21 days after treatment; meanwhile, no changes were established in NAcc. Finally, the increased locomotor activity induced by amphetamine challenge was blunted by losartan administration in CPu. No differences were detected in the behavioral sensitization when the AT1 blocker was injected in NAcc. Our results support the hypothesis of a key role of brain RAS in the neuroadaptative changes induced by amphetamine.


Subject(s)
Amphetamine/pharmacology , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Renin-Angiotensin System/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensinogen/metabolism , Animals , Brain/physiology , Losartan/pharmacology , Male , Motor Activity/drug effects , Motor Activity/physiology , RNA, Messenger/metabolism , Random Allocation , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/physiology
20.
Biomed Res Int ; 2014: 183248, 2014.
Article in English | MEDLINE | ID: mdl-25003108

ABSTRACT

Central nucleus of the amygdala (CeA) is one of the most important regulatory centres for the emotional processes. Among the different neurotransmitter systems present in this nucleus, AT1 receptors have been also found, but their role in the generation and modulation of emotions is not fully understood. The present work evaluated the effect of intra-amygdalar injection of losartan (AT1 receptor antagonist) and angiotensin II (Ang II) in the anxiety state induced by fear-potentiated plus maze in male Wistar rats. Fear in the elevated plus maze can be potentiated by prior inescapable footshock stress. The decrease in the time spent in the open arms induced by the inescapable footshock was totally prevented by losartan (4 pmol) administration in CeA. It was also found that Ang II (48 fmol) administration decreased the time spent in the open arms in animals with or without previous footshock exposure. The locomotor activity and grooming behaviour were also evaluated. The results obtained from the different parameters analyzed allowed us to conclude that the Ang II AT1 receptors in CeA are involved in the anxiety state induced by stress in the fear-potentiated plus-maze behaviour.


Subject(s)
Angiotensin II/pharmacology , Central Amygdaloid Nucleus/physiology , Fear/drug effects , Receptor, Angiotensin, Type 1/metabolism , Animals , Behavior, Animal/drug effects , Central Amygdaloid Nucleus/drug effects , Grooming/drug effects , Male , Maze Learning/drug effects , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL