Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Eur J Clin Invest ; 52(4): e13694, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34694635

RESUMO

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.


Assuntos
Astrócitos/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Metanfetamina/farmacologia , Sesquiterpenos/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
BMC Med Educ ; 22(1): 624, 2022 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-35978358

RESUMO

BACKGROUND: Physical activity is a major determinant of physical and mental health. International recommendations identify health professionals as pivotal agents to tackle physical inactivity. This study sought to characterize medical doctors' clinical practices concerning the promotion of patients' physical activity, while also exploring potential predictors of the frequency and content of these practices, including doctors' physical activity level and sedentary behaviours. METHODS: A cross-sectional study assessed physical activity promotion in clinical practice with a self-report questionnaire delivered through the national medical prescription software (naturalistic survey). Physical activity and sedentary behaviours were estimated using the International Physical Activity Questionnaire (short form). Indicators of medical doctors' attitudes, knowledge, confidence, barriers, and previous training concerning physical activity promotion targeting their patients were also assessed. Multiple regression analysis was performed to identify predictors of physical activity promotion frequency by medical doctors, including sociodemographic, attitudes and knowledge-related variables, and physical activity behaviours as independent variables. RESULTS: A total of 961 medical doctors working in the Portuguese National Health System participated (59% women, mean age 44 ± 13 years) in the study. The majority of the participants (84.6%) reported to frequently promote patients' physical activity. Five predictors of physical activity promotion frequency emerged from the multiple regression analysis, explaining 17.4% of the dependent variable (p < 0.001): working in primary healthcare settings (p = 0.037), having a medical specialty (p = 0.030), attributing a high degree of relevance to patients' physical activity promotion in healthcare settings (p < 0.001), being approached by patients to address physical activity (p < 0.001), and having higher levels of physical activity (p = 0.001). CONCLUSIONS: The sample of medical doctors approached reported a high level of engagement with physical activity promotion. Physical activity promotion frequency seems to be influenced by the clinical practice setting, medical career position and specialty, attitudes towards physical activity, and perception of patients´ interest on the topic, as well as medical doctors' own physical activity levels.


Assuntos
Médicos , Adulto , Atitude do Pessoal de Saúde , Estudos Transversais , Exercício Físico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Médicos/psicologia , Autorrelato , Inquéritos e Questionários
3.
Int J Mol Sci ; 23(22)2022 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-36430321

RESUMO

Methylmercury (MeHg) is highly toxic to the human brain. Although much is known about MeHg neurotoxic effects, less is known about how chronic MeHg affects hippocampal amino acids and other neurochemical markers in adult mice. In this study, we evaluated the MeHg effects on systemic lipids and inflammation, hippocampal oxidative stress, amino acid levels, neuroinflammation, and behavior in adult male mice. Challenged mice received MeHg in drinking water (2 mg/L) for 30 days. We assessed weight gain, total plasma cholesterol (TC), triglycerides (TG), endotoxin, and TNF levels. Hippocampal myeloperoxidase (MPO), malondialdehyde (MDA), acetylcholinesterase (AChE), amino acid levels, and cytokine transcripts were evaluated. Mice underwent open field, object recognition, Y, and Barnes maze tests. MeHg-intoxicated mice had higher weight gain and increased the TG and TC plasma levels. Elevated circulating TNF and LPS confirmed systemic inflammation. Higher levels of MPO and MDA and a reduction in IL-4 transcripts were found in the hippocampus. MeHg-intoxication led to increased GABA and glycine, reduced hippocampal taurine levels, delayed acquisition in the Barnes maze, and poor locomotor activity. No significant changes were found in AChE activity and object recognition. Altogether, our findings highlight chronic MeHg-induced effects that may have long-term mental health consequences in prolonged exposed human populations.


Assuntos
Compostos de Metilmercúrio , Animais , Humanos , Masculino , Camundongos , Acetilcolinesterase/metabolismo , Aminoácidos , Hipocampo/metabolismo , Inflamação/induzido quimicamente , Compostos de Metilmercúrio/toxicidade , Compostos de Metilmercúrio/metabolismo , Aumento de Peso , Camundongos Endogâmicos C57BL
4.
Int J Mol Sci ; 22(6)2021 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-33803585

RESUMO

Methylmercury (MeHg) toxicity is a major environmental concern. In the aquatic reservoir, MeHg bioaccumulates along the food chain until it is consumed by riverine populations. There has been much interest in the neurotoxicity of MeHg due to recent environmental disasters. Studies have also addressed the implications of long-term MeHg exposure for humans. The central nervous system is particularly susceptible to the deleterious effects of MeHg, as evidenced by clinical symptoms and histopathological changes in poisoned humans. In vitro and in vivo studies have been crucial in deciphering the molecular mechanisms underlying MeHg-induced neurotoxicity. A collection of cellular and molecular alterations including cytokine release, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate dyshomeostasis, and cell death mechanisms are important consequences of brain cells exposure to MeHg. The purpose of this review is to organize an overview of the mercury cycle and MeHg poisoning events and to summarize data from cellular, animal, and human studies focusing on MeHg effects in neurons and glial cells. This review proposes an up-to-date compendium that will serve as a starting point for further studies and a consultation reference of published studies.


Assuntos
Encéfalo/patologia , Inflamação/patologia , Compostos de Metilmercúrio/toxicidade , Síndromes Neurotóxicas/patologia , Animais , Bioacumulação , Encéfalo/efeitos dos fármacos , Humanos , Compostos de Metilmercúrio/farmacocinética , Microbiota/efeitos dos fármacos , Síndromes Neurotóxicas/microbiologia , Síndromes Neurotóxicas/prevenção & controle , Síndromes Neurotóxicas/terapia
5.
Eur J Neurosci ; 51(6): 1377-1387, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31454441

RESUMO

Microglia cells exert a critical role in brain development, mainly supported by their immune functions, which predicts an impact on the genesis of psychiatric disorders. In fact, microglia stress during gestation is, for instance, associated with chronic anxiety and cognitive deficits accompanied by long-lasting, region- and sex-specific changes in microglia morphology. We recently reported that the pattern of microglia morphologic plasticity, which is sex-determined, impacts on anxious-like behaviour and cognition. We also reported that the pharmacologic blockade of adenosine A2A receptors (A2A R) is able to reshape microglia morphology, in a sex-specific manner and with behavioural sequelae. In order to better understand the role of A2A R in the sex differentiation of microglia, we now compared their morphology in wild-type and A2A R knockout male and female C57BL/6 mice in two cardinal brain regions implicated in anxiety-like behaviour and cognition, the prefrontal cortex (PFC) and the dorsal hippocampus (dHIP). We report interregional differences between PFC and dHIP in a sex-specific manner: while males presented more complex microglia in the dHIP, microglia from females had a more complex morphology in the PFC. Surprisingly, the genetic deletion of A2A R did not alter these sex differences, but promoted the exclusive remodelling (increase in complexity) in PFC microglia from females. These findings further support the existence of a heterogeneous microglial network, distinct between sexes and brain regions, and help characterizing the role of A2A R in the sex- and brain region-specific morphologic differentiation of microglia.


Assuntos
Microglia , Receptor A2A de Adenosina , Caracteres Sexuais , Adenosina , Animais , Encéfalo/metabolismo , Feminino , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo
6.
Mol Pharm ; 17(6): 2145-2154, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32339462

RESUMO

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.


Assuntos
Caveolina 1/metabolismo , Indóis/química , Indóis/uso terapêutico , Fotoquimioterapia/métodos , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/terapia , Idoso , Western Blotting , Caveolina 1/genética , Linhagem Celular Tumoral , Endocitose/efeitos dos fármacos , Feminino , Galectina 1/genética , Galectina 1/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Técnicas In Vitro , Isoindóis , Masculino , Microscopia de Fluorescência
7.
J Cell Physiol ; 234(11): 19750-19760, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30937892

RESUMO

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor, with an average life expectancy of 12-15 months. GBM is highly infiltrated by microglial cells (MG) promoting tumor growth and invasiveness. Moreover, microglia activation and subsequent neuroinflammation seem to be involved in blood-brain barrier (BBB) dysfunction commonly observed in several central nervous system diseases, including brain tumors. Nevertheless, how the crosstalk between microglia and tumor cells interferes with BBB function is far from being clarified. Herein, we evaluated the effects of reciprocal interactions between MG and GBM cells in the barrier properties of brain endothelial cells (ECs), using an in vitro approach. The exposure of ECs to the inflammatory microenvironment mediated by MG-GBM crosstalk induced a decrease in the transendothelial electric resistance and an increase in permeability across the ECs (macromolecular flux of 4 kDa-fluorescein isothiocyanate and 70 kDa-Rhodamine B isothiocyanate-Dextran). These effects were accompanied by a downregulation of the intercellular junction proteins, ß-catenin and zonula occludens. Moreover, the dynamic interaction between microglia and tumor cells triggered the release of interleukin-6 (IL-6) by microglia and subsequent activation of the downstream Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) pathway. Interestingly, the depletion of IL-6 or the blockade of the JAK/STAT3 signaling with AG490 were able to prevent the EC hyperpermeability. Overall, we demonstrated that IL-6 released during MG-GBM crosstalk leads to barrier dysfunction through the activation of the JAK/STAT3 pathway in ECs and downregulation of intercellular junction proteins. These results provide new insights into the mechanisms underlying the disruption of BBB permeability in GBM.


Assuntos
Glioblastoma/genética , Interleucina-6/genética , Janus Quinase 2/genética , Fator de Transcrição STAT3/genética , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Proliferação de Células/genética , Técnicas de Cocultura , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Glioblastoma/patologia , Glucose-6-Fosfato Isomerase , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Microglia/metabolismo , Microglia/patologia , Permeabilidade , Transdução de Sinais/genética , Microambiente Tumoral/genética , beta Catenina/genética , beta Catenina/metabolismo
8.
Glia ; 67(1): 182-192, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30461068

RESUMO

Epidemiologic studies have provided compelling evidence that prenatal stress, through excessive maternal glucocorticoids exposure, is associated with psychiatric disorders later in life. We have recently reported that anxiety associated with prenatal exposure to dexamethasone (DEX, a synthetic glucocorticoid) correlates with a gender-specific remodeling of microglia in the medial prefrontal cortex (mPFC), a core brain region in anxiety-related disorders. Gender differences in microglia morphology, the higher prevalence of anxiety in women and the negative impact of anxiety in cognition, led us to specifically evaluate cognitive behavior and associated circuits (namely mPFC-dorsal hippocampus, dHIP), as well as microglia morphology in female rats prenatally exposed to dexamethasone (in utero DEX, iuDEX). We report that iuDEX impaired recognition memory and deteriorated neuronal synchronization between mPFC and dHIP. These functional deficits are paralleled by microglia hyper-ramification in the dHIP and decreased ramification in the mPFC, showing a heterogeneous remodeling of microglia morphology, both postnatally and at adulthood in different brain regions, that differently affect mood and cognition. The chronic blockade of adenosine A2A receptors (A2A R), which are core regulators of microglia morphology and physiology, ameliorated the cognitive deficits, but not the anxiety-like behavior. Notably, A2A R blockade rectified both microglia morphology in the dHIP and the lack of mPFC-dHIP synchronization, further heralding their role in cognitive function.


Assuntos
Ansiedade/metabolismo , Disfunção Cognitiva/metabolismo , Microglia/metabolismo , Receptor A2A de Adenosina/metabolismo , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Ansiedade/induzido quimicamente , Ansiedade/psicologia , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/psicologia , Dexametasona/toxicidade , Feminino , Glucocorticoides/toxicidade , Masculino , Microglia/efeitos dos fármacos , Gravidez , Ratos , Ratos Wistar
9.
Analyst ; 144(6): 2062-2079, 2019 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-30724915

RESUMO

trans-Resveratrol (RSV) is a plant-derived polyphenol endowed with a broad spectrum of promising therapeutic activities. The applicability of RSV in vivo has, however, had limited success so far, largely due to its inefficient systemic delivery resulting from its low water solubility. Layer-by-Layer (LbL) nanotechnology constitutes an innovative formulation strategy to address this concern, and is based on the design of tunable onion-like multilayered nanoarchitectures on the surface of low solubility drug nanocores, such as RSV. The purpose of this study was the investigation of the bioavailability of an LbL nanoformulation composed of 5.5 bilayers of polyallylamine hydrochloride (PAH) and dextran sulfate (DS) (LbL NPs) by pharmacokinetic studies following oral dosing to Wistar rats (20 mg kg-1). The systemic exposure of LbL NPs was compared to the respective nanoformulation without LbL coatings (RSV nanocores) and the free RSV suspension. The results demonstrated that both LbL NPs and RSV nanocores significantly enhanced, respectively, 1.76-fold and 2.74-fold the systemic exposure of RSV compared to the free RSV suspension, emphasizing their biopharmaceutical advantage. Surprisingly, besides the modified drug release potential of the LbL NPs, these exhibited a slightly lower systemic exposure (0.36-fold) in comparison with non-LbL modified RSV nanocores. These results were justified only by the electrostatic interactions composition of the LbL shell composition, requiring further research towards the application of stronger interactions. For this study, due to the key role of the bioanalytical method in the in vivo data acquisition, a rapid, selective, and sensitive HPLC-DAD method has been successfully optimized and fully validated to confidently quantify RSV levels in the rat plasma matrix, together with the optimization of the sample preparation procedure. Moreover, the chemical stability of RSV was evaluated for 24 h in simulated gastric and intestinal fluids with enzymes. Overall, our findings suggest that LbL NPs should be given great attention, representing a potential drug delivery system for RSV in view of the application of RSV not solely as a supplement but also as a therapeutic drug.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacocinética , Sistemas de Liberação de Medicamentos , Nanopartículas/administração & dosagem , Nanopartículas/química , Resveratrol/administração & dosagem , Resveratrol/farmacocinética , Administração Oral , Animais , Disponibilidade Biológica , Masculino , Ratos , Ratos Wistar , Distribuição Tecidual
10.
Brain Behav Immun ; 68: 169-182, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29061363

RESUMO

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.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Privilégio Imunológico/fisiologia , Metilfenidato/farmacologia , Animais , Ansiedade/metabolismo , Atenção/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Privilégio Imunológico/imunologia , Masculino , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY
11.
BMC Pediatr ; 18(1): 212, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29970045

RESUMO

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.


Assuntos
Composição Corporal , Densidade Óssea , Treinamento Resistido , Natação/fisiologia , Voleibol/fisiologia , Absorciometria de Fóton , Adolescente , Tamanho Corporal , Dieta , Feminino , Humanos , Extremidade Inferior/fisiologia , Dobras Cutâneas , Extremidade Superior/fisiologia
12.
Brain Behav Immun ; 62: 306-317, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28237710

RESUMO

Methamphetamine (METH) is a highly addictive psychostimulant drug that can lead to neurological and psychiatric abnormalities. Several studies have explored the central impact of METH use, but the mechanism(s) underlying blood-brain barrier (BBB) dysfunction and associated neuroinflammatory processes after chronic METH consumption are still unclear. Important findings in the field are mainly based on in vitro approaches and animal studies using an acute METH paradigm, and not much is known about the neurovascular alterations under a chronic drug use. Thus, the present study aimed to fill this crucial gap by exploring the effect of METH-self administration on BBB function and neuroinflammatory responses. Herein, we observed an increase of BBB permeability characterized by Evans blue and albumin extravasation in the rat hippocampus and striatum triggered by extended-access METH self-administration followed by forced abstinence. Also, there was a clear structural alteration of blood vessels showed by the down-regulation of collagen IV staining, which is an important protein of the endothelial basement membrane, together with a decrease of intercellular junction protein levels, namely claudin-5, occludin and vascular endothelial-cadherin. Additionally, we observed an up-regulation of vascular cell and intercellular adhesion molecule, concomitant with the presence of T cell antigen CD4 and tissue macrophage marker CD169 in the brain parenchyma. Rats trained to self-administer METH also presented a neuroinflammatory profile characterized by microglial activation, astrogliosis and increased pro-inflammatory mediators, namely tumor necrosis factor-alpha, interleukine-1 beta, and matrix metalloproteinase-9. Overall, our data provide new insights into METH abuse consequences, with a special focus on neurovascular dysfunction and neuroinflammatory response, which may help to find novel approaches to prevent or diminish brain dysfunction triggered by this overwhelming illicit drug.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Corpo Estriado/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Inflamação/etiologia , Metanfetamina/administração & dosagem , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Claudina-5/metabolismo , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Ocludina/metabolismo , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Wistar , Autoadministração
13.
Cell Mol Life Sci ; 73(24): 4701-4716, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27376435

RESUMO

Methylphenidate (MPH) is an amphetamine-like stimulant commonly prescribed for attention deficit hyperactivity disorder. Despite its widespread use, the cellular/molecular effects of MPH remain elusive. Here, we report a novel direct role of MPH on the regulation of macromolecular flux through human brain endothelial cells (ECs). MPH significantly increased caveolae-mediated transcytosis of horseradish peroxidase through ECs without affecting paracellular permeability. Using FRET-based live cell imaging, together with pharmacological inhibitors and lentiviral-mediated shRNA knockdown, we demonstrate that MPH promoted ROS generation via activation of Rac1-dependent NADPH oxidase (NOX) and c-Src activation at the plasma membrane. c-Src in turn was shown to mediate the phosphorylation of caveolin-1 (Cav1) on Tyr14 leading to enhanced caveolae formation and transendothelial transport. Accordingly, the inhibition of Cav1 phosphorylation by overexpression of a phosphodefective Cav1Y14F mutant or knocking down Cav1 expression abrogated MPH-induced transcytosis. In addition, both vitamin C and inhibition of NOX blocked MPH-triggered vesicular transport. This study, therefore, identifies Rac1/NOX/c-Src-dependent signaling in MPH-induced increase in transendothelial permeability of brain endothelial cell monolayers via caveolae-mediated transcytosis.


Assuntos
Cavéolas/metabolismo , Caveolina 1/metabolismo , Células Endoteliais/metabolismo , Metilfenidato/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transcitose/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo , Transporte Biológico/efeitos dos fármacos , Encéfalo/citologia , Proteína Tirosina Quinase CSK , Permeabilidade Capilar/efeitos dos fármacos , Cavéolas/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Modelos Biológicos , NADPH Oxidases/metabolismo , Oxidantes/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Vesículas Transportadoras/efeitos dos fármacos , Vesículas Transportadoras/metabolismo , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
14.
J Neurochem ; 138(4): 598-609, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27221633

RESUMO

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 .


Assuntos
Astrócitos/metabolismo , Corpo Estriado/metabolismo , Neostriado/metabolismo , Doença de Parkinson/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Animais , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo , Receptor para Produtos Finais de Glicação Avançada/genética
15.
Mediators Inflamm ; 2014: 538737, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24817793

RESUMO

This study aimed to evaluate the efficacy of sitagliptin, a dipeptidyl peptidase IV (DPP-IV) inhibitor, in preventing the deleterious effects of diabetes on the kidney in an animal model of type 2 diabetes mellitus; the Zucker diabetic fatty (ZDF) rat: 20-week-old rats were treated with sitagliptin (10 mg/kg bw/day) during 6 weeks. Glycaemia and blood HbA1c levels were monitored, as well as kidney function and lesions. Kidney mRNA and/or protein content/distribution of DPP-IV, GLP-1, GLP-1R, TNF-α, IL-1ß, BAX, Bcl-2, and Bid were evaluated by RT-PCR and/or western blotting/immunohistochemistry. Sitagliptin treatment improved glycaemic control, as reflected by the significantly reduced levels of glycaemia and HbA1c (by about 22.5% and 1.2%, resp.) and ameliorated tubulointerstitial and glomerular lesions. Sitagliptin prevented the diabetes-induced increase in DPP-IV levels and the decrease in GLP-1 levels in kidney. Sitagliptin increased colocalization of GLP-1 and GLP-1R in the diabetic kidney. Sitagliptin also decreased IL-1ß and TNF-α levels, as well as, prevented the increase of BAX/Bcl-2 ratio, Bid protein levels, and TUNEL-positive cells which indicates protective effects against inflammation and proapoptotic state in the kidney of diabetic rats, respectively. In conclusion, sitagliptin might have a major role in preventing diabetic nephropathy evolution due to anti-inflammatory and antiapoptotic properties.


Assuntos
Apoptose/efeitos dos fármacos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Inflamação/prevenção & controle , Pirazinas/uso terapêutico , Triazóis/uso terapêutico , Animais , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Inflamação/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Ratos , Ratos Zucker , Fosfato de Sitagliptina , Fator de Necrose Tumoral alfa/metabolismo
16.
Pharmacology ; 91(3-4): 153-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23392332

RESUMO

Tramadol is a central-acting analgesic associated with nausea and vomiting. Clinical studies have demonstrated that glucocorticoids have analgesic and antiemetic effects when administered perioperatively. The aim of this study is to test the hypothesis that coadministration of tramadol and dexamethasone decreases both postoperative pain and tramadol requirement by patient-controlled analgesia (PCA). Forty female patients undergoing thyroidectomy under general anesthesia were enrolled in a double-blind randomized controlled study and allocated to receive dexamethasone 4 mg i.v. (dexamethasone group, n = 20) or saline (control group, n = 20). At 0, 1, 2, 4 and 22 h of PCA, tramadol consumption and pain were evaluated. Although pain (numerical rating scale 0-10) was significantly lower in the dexamethasone group compared to the control group (2.9 ± 1.4 vs. 3.8 ± 1.2, p = 0.02) at the beginning of PCA, tramadol demand was not significantly different. Although the results herein show a possible beneficial effect of a preoperative single low dose of dexamethasone on postoperative pain, the hypothesis that this corticosteroid decreases tramadol requirement is not supported.


Assuntos
Analgésicos Opioides/administração & dosagem , Antieméticos/administração & dosagem , Dexametasona/administração & dosagem , Dor Pós-Operatória/tratamento farmacológico , Tramadol/administração & dosagem , Adulto , Método Duplo-Cego , Quimioterapia Combinada , Feminino , Humanos , Pessoa de Meia-Idade , Náusea/prevenção & controle , Tireoidectomia , Vômito/prevenção & controle
17.
Antioxidants (Basel) ; 12(4)2023 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-37107312

RESUMO

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.

18.
World J Emerg Med ; 14(6): 454-461, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37969211

RESUMO

BACKGROUND: Long QT syndrome (LQTS) is a heterogeneous syndrome that may be congenital or, more frequently, acquired. The real-world prevalence of acquired LQTS (aLQTS) in the emergency department (ED) remains to be determined. The aim of this study was to determine prevalence of aLQTS and its impact on symptoms on ED admissions. METHODS: Electrocardiograms (ECG) of 5,056 consecutively patients admitted in the ED of a tertiary hospital between January 28th and March 17th of 2020 were reviewed. All patients with aLQTS were included. Clinical data with a focus on QT prolonging drugs and clinical factors were recorded. Statistical comparison was made between the groups with and without corrected QT (QTc) interval greater than 500 ms (value that is considered severely increased). RESULTS: A total of 383 ECGs with prolonged QTc were recognized, corresponding to a prevalence of aLQTS at admission of 7.82%. Patients with aLQTS were more commonly men (53.3%) with an age of (73.49±14.79) years old and QTc interval of (505.3±32.4) ms. Only 20.4% of these patients with aLQTS were symptomatic. No ventricular arrhythmias were recorded. Patients with QT interval greater than 500 ms were more frequently female (59.5%; P<0.001) and were more frequently on QT prolonging drugs (77.3%; P=0.025). Main contributing factor was intake of antibiotics (odds ratio [OR] 4.680) followed by female gender (OR 2.473) and intake of antipsychotics (OR 1.925). CONCLUSION: aLQTS is particularly prevalent in the ED. Female patients on antibiotics and antipsychotics are at particularly high risk. Efforts must be made to avoid, detect and treat aLQTS as early as possible.

20.
J Neurochem ; 123(6): 1041-53, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23061411

RESUMO

Methamphetamine (METH) is a psychostimulant drug that causes irreversible brain damage leading to several neurological and psychiatric abnormalities, including cognitive deficits. Neuropeptide Y (NPY) is abundant in the mammalian central nervous system (CNS) and has several important functions, being involved in learning and memory processing. It has been demonstrated that METH induces significant alteration in mice striatal NPY, Y(1) and Y(2) receptor mRNA levels. However, the impact of this drug on the hippocampal NPY system and its consequences remain unknown. Thus, in this study, we investigated the effect of METH intoxication on mouse hippocampal NPY levels, NPY receptors function, and memory performance. Results show that METH increased NPY, Y(2) and Y(5) receptor mRNA levels, as well as total NPY binding accounted by opposite up- and down-regulation of Y(2) and Y(1) functional binding, respectively. Moreover, METH-induced impairment in memory performance and AKT/mammalian target of rapamycin pathway were both prevented by the Y(2) receptor antagonist, BIIE0246. These findings demonstrate that METH interferes with the hippocampal NPY system, which seems to be associated with memory failure. Overall, we concluded that Y(2) receptors are involved in memory deficits induced by METH intoxication.


Assuntos
Estimulantes do Sistema Nervoso Central/toxicidade , Hipocampo/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Metanfetamina/toxicidade , Neuropeptídeo Y/antagonistas & inibidores , Neuropeptídeo Y/metabolismo , Animais , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Transtornos da Memória/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Receptores de Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA