Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Pediatr Res ; 95(7): 1783-1790, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38360979

RESUMO

BACKGROUND: Neonatal rats can manifest post-stroke mood disorders (PSMD) following middle cerebral artery occlusion (MCAO). We investigated whether cannabidiol (CBD) neuroprotection, previously demonstrated in neonatal rats after MCAO, includes prevention of PSMD development. METHODS: Seven-day-old Wistar rats (P7) underwent MCAO and received either vehicle or 5 mg/kg CBD treatment. Brain damage was quantified by MRI, and neurobehavioral and histological (TUNEL) studies were performed at P14 and P37. PSMD were assessed using the tail suspension test, forced swimming test, and open field tests. The dopaminergic system was evaluated by quantifying dopaminergic neurons (TH+) in the Ventral Tegmental Area (VTA), measuring brain dopamine (DA) concentration and DA transporter expression, and assessing the expression and function D2 receptors (D2R) through [35S]GTPγS binding. Animals without MCAO served as controls. RESULTS: CBD reduced MCAO-induced brain damage and improved motor performance. At P14, MCAO induced depressive-like behavior, characterized by reduced TH+ cell population and DA levels, which CBD did not prevent. However, CBD ameliorated hyperactivity observed at P37, preventing increased DA concentration by restoring D2R function. CONCLUSIONS: These findings confirm the development of PSMD following MCAO in neonatal rats and highlight CBD as a neuroprotective agent capable of long-term functional normalization of the dopaminergic system post-MCAO. IMPACT: MCAO in neonatal rats led to post-stroke mood disorders consisting in a depression-like picture in the medium term evolving towards long-term hyperactivity, associated with an alteration of the dopaminergic system. The administration of CBD after MCAO did not prevent the development of depressive-like behavior, but reduced long-term hyperactivity, normalizing dopamine receptor function. These data point to the importance of considering the development of depression-like symptoms after neonatal stroke, a well-known complication after stroke in adults. Our work confirms the interest of CBD as a possible treatment for neonatal stroke.


Assuntos
Animais Recém-Nascidos , Canabidiol , Dopamina , Transtornos do Humor , Ratos Wistar , Acidente Vascular Cerebral , Animais , Canabidiol/farmacologia , Canabidiol/uso terapêutico , Ratos , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/metabolismo , Transtornos do Humor/tratamento farmacológico , Transtornos do Humor/etiologia , Dopamina/metabolismo , Fármacos Neuroprotetores/farmacologia , Receptores de Dopamina D2/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Masculino , Modelos Animais de Doenças , Comportamento Animal/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo
2.
Pediatr Res ; 93(1): 78-88, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35428877

RESUMO

BACKGROUND: We aimed to characterize a preclinical model of intraventricular hemorrhage-induced brain damage (IVH-BD) in extremely low birth weight newborns (ELBWN), to identify potential therapeutic targets based on its pathophysiology. METHODS: IVH was induced in 1-day-old (P1) Wistar rats by left periventricular injection of clostridium collagenase (PVCC). At P6, P14, and P45 IVH-BD (area of damage, motor and cognitive deficits, Lactate/N-acetylaspartate ratio), white matter injury (WMI: ipsilateral hemisphere and corpus callosum atrophy, oligodendroglial population and myelin basic protein signal reduction), blood-brain barrier (BBB) dysfunction (occludin and Mfsd2a expression, Gadolinium leakage) and inflammation (TNFα, TLR4, NFkB, and MMP9 expression; immune cell infiltration), excitotoxicity (Glutamate/N-acetylaspartate), and oxidative stress (protein nitrosylation) were assessed. Sham animals were similarly studied. RESULTS: IVH-BD leads to long-term WMI, resulting in motor and cognitive impairment, thus reproducing IVH-BD features in ELBWN. BBB dysfunction with increased permeability was observed at P6 and P14, coincident with an increased inflammatory response with TLR4 overexpression, increased TNFα production, and increased immune cell infiltration, as well as increased excitotoxicity and oxidative stress. CONCLUSIONS: This model reproduced some key hallmarks of IVH-BD in ELBWN. Inflammation associated with BBB dysfunction appears as relevant therapeutic target to prevent IVH-BD-induced WMI. IMPACT: Paraventricular injection of clostridium collagenase (PVCC) to 1-day-old Wistar rats uniquely reproduced the neuroimaging, histologic and functional characteristics of intraventricular hemorrhage-induced brain damage (IVH-BD) in extremely low birth weight newborns (ELBWN). PVCC-induced IVH triggered a prolonged inflammatory response associated with blood-brain barrier increased permeability, which in turn facilitates the infiltration of inflammatory cells. Thus, PVCC led to white matter injury (WMI) resulting in long-term motor and cognitive impairment. This model offers a valuable tool to obtain further insight into the mechanisms of IVH-BD in ELBWN and proposes some key therapeutic targets.


Assuntos
Barreira Hematoencefálica , Lesões Encefálicas , Animais , Ratos , Fator de Necrose Tumoral alfa/metabolismo , Ratos Wistar , Peso ao Nascer , Receptor 4 Toll-Like/metabolismo , Hemorragia Cerebral/complicações , Lesões Encefálicas/etiologia , Inflamação/metabolismo , Colagenases/metabolismo , Colagenases/uso terapêutico
3.
Int J Mol Sci ; 24(4)2023 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-36834637

RESUMO

Post-stroke mood disorders (PSMD) affect disease prognosis in adults. Adult rodent models underlie the importance of the dopamine (DA) system in PSMD pathophysiology. There are no studies on PSMD after neonatal stroke. We induced neonatal stroke in 7-day-old (P7) rats by temporal left middle cerebral artery occlusion (MCAO). Performance in the tail suspension test (TST) at P14 and the forced swimming test (FST) and open field test (OFT) at P37 were studied to assess PSMD. DA neuron density in the ventral tegmental area, brain DA concentration and DA transporter (DAT) expression as well as D2 receptor (D2R) expression and G-protein functional coupling were also studied. MCAO animals revealed depressive-like symptoms at P14 associated with decreased DA concentration and reduced DA neuron population and DAT expression. At P37, MCAO rats showed hyperactive behavior associated with increased DA concentration, normalization of DA neuron density and decreased DAT expression. MCAO did not modify D2R expression but reduced D2R functionality at P37. MCAO-induced depressive-like symptoms were reversed by the DA reuptake inhibitor GBR-12909. In conclusion, MCAO in newborn rats induced depressive-like symptoms and hyperactive behavior in the medium and long term, respectively, that were associated with alterations in the DA system.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Dopamina , Dopamina , Ratos , Animais , Dopamina/metabolismo , Animais Recém-Nascidos , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Encéfalo/metabolismo , Receptores de Dopamina D2/metabolismo , Inibidores da Captação de Dopamina/farmacologia
4.
Int J Mol Sci ; 23(17)2022 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-36077095

RESUMO

Background: Cannabidiol (CBD) is a phytocannabinoid with potential in one of the most prevalent syndromes occurring at birth, the hypoxia of the neonate. CBD targets a variety of proteins, cannabinoid CB2 and serotonin 5HT1A receptors included. These two receptors may interact to form heteromers (CB2-5HT1A-Hets) that are also a target of CBD. Aims: We aimed to assess whether the expression and function of CB2-5HT1A-Hets is affected by CBD in animal models of hypoxia of the neonate and in glucose- and oxygen-deprived neurons. Methods: We developed a quantitation of signal transduction events in a heterologous system and in glucose/oxygen-deprived neurons. The expression of receptors was assessed by immuno-cyto and -histochemistry and, also, by using the only existing technique to visualize CB2-5HT1A-Hets fixed cultured cells and tissue sections (in situ proximity ligation PLA assay). Results: CBD and cannabigerol, which were used for comparative purposes, affected the structure of the heteromer, but in a qualitatively different way; CBD but not CBG increased the affinity of the CB2 and 5HT1A receptor-receptor interaction. Both cannabinoids regulated the effects of CB2 and 5HT1A receptor agonists. CBD was able to revert the upregulation of heteromers occurring when neurons were deprived of oxygen and glucose. CBD significantly reduced the increased expression of the CB2-5HT1A-Het in glucose/oxygen-deprived neurons. Importantly, in brain sections of a hypoxia/ischemia animal model, administration of CBD led to a significant reduction in the expression of CB2-5HT1A-Hets. Conclusions: Benefits of CBD in the hypoxia of the neonate are mediated by acting on CB2-5HT1A-Hets and by reducing the aberrant expression of the receptor-receptor complex in hypoxic-ischemic conditions. These results reinforce the potential of CBD for the therapy of the hypoxia of the neonate.


Assuntos
Canabidiol , Canabinoides , Animais , Canabidiol/farmacologia , Canabinoides/metabolismo , Canabinoides/farmacologia , Modelos Animais de Doenças , Glucose , Hipóxia , Neurônios/metabolismo , Oxigênio , Receptor CB1 de Canabinoide/genética , Receptor CB2 de Canabinoide/genética , Receptor 5-HT1A de Serotonina , Serotonina
5.
Eur J Pediatr ; 179(10): 1609-1618, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32367328

RESUMO

The relationship between right ventricular (RV) function and cerebral blood flow (CBF) velocity and cerebral oxygenation was assessed in neonates with hypoxic-ischemic encephalopathy (HIE) treated with therapeutic hypothermia (TH). Echocardiographic, transcranial Doppler, and hemodynamic data from 37 neonates with moderate-severe HIE + TH were reviewed. Twenty healthy newborns served as controls. Cardiac dysfunction in HIE + TH was characterized by a predominant RV dysfunction, with concomitantly reduced CBF velocity. A significant correlation was found between CBF velocity and tricuspid annular plane systolic excursion (TAPSE), RV output (RVO), and stroke volume (SVRV), as well as with left ventricular output and stroke volume. Brain oxygenation (rSO2) correlated significantly with RVO, SVRV, TAPSE, ejection fraction, and fractional shortening, whereas cerebral fractional tissue oxygen extraction (FTOEc) correlated with RVO, SVRV, RV myocardial performance index, and superior vena cava flow. CBF velocity and cerebral NIRS correlations were stronger with parameters of right ventricular performance.Conclusion: CBF velocity and brain oxygenation correlate predominantly with RV function in HIE + TH. This suggests a preferential contribution of RV performance to cerebral hemodynamics in this context. What is Known: • Neonates with hypoxic ischemic encephalopathy frequently exhibit alterations of cardiac function and cerebral blood flow. • These are considered organ-specific consequences of perinatal asphyxia. What is New: • We show that cerebral blood flow velocity and brain oxygenation are correlated predominantly with right ventricular function during therapeutic hypothermia. • This suggests a potential direct contribution of right ventricular performance to cerebral hemodynamics in this context.


Assuntos
Asfixia Neonatal , Hipóxia-Isquemia Encefálica , Disfunção Ventricular Direita , Velocidade do Fluxo Sanguíneo , Circulação Cerebrovascular , Humanos , Hipóxia-Isquemia Encefálica/diagnóstico por imagem , Hipóxia-Isquemia Encefálica/terapia , Recém-Nascido , Volume Sistólico , Veia Cava Superior , Função Ventricular Direita
6.
Pediatr Res ; 85(4): 539-545, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30675020

RESUMO

BACKGROUND: Newborn pigs offer theoretical advantages for studying newborn hypoxic-ischemic (HI) brain damage because of a development and structure similar to the human brain. However, the correlation between functional features and actual HI brain damage has not been reported. METHODS: Newborn pigs were examined daily for 3 days after a HI insult using amplitude-integrated EEG (aEEG), and a neurobehavioral score enriched with stress and social and object interaction-driven activity evaluation. Brain damage was then assessed using histologic, immunohistochemical, and proton magnetic resonance spectroscopy studies. Brain concentration of several neurotransmitters was determined by HPLC. RESULTS: HI insult led to aEEG amplitude decrease, muscle tone and activity impairment, eating disorders, poor environmental interaction, and increased motionless periods. Basal aEEG amplitude, muscle tone, and general behavior were the best predictive items for histological and biochemical (lactate/N-acetylaspartate ratio) brain damage. Hyperexcitable response to stress correlated inversely with brain damage. Motionless time, which correlated with brain damage severity, was inversely related to brain concentration of dopamine and norepinephrine. CONCLUSION: Standard neurologic examination of brain activity and motor and behavioral performance of newborn pigs is a valuable tool to assess HI brain damage, thus offering a powerful translational model for HI brain damage pathophysiology and management studies.


Assuntos
Eletroencefalografia/métodos , Hipóxia-Isquemia Encefálica/patologia , Exame Neurológico , Animais , Hipóxia-Isquemia Encefálica/fisiopatologia , Índice de Gravidade de Doença , Análise de Sobrevida , Suínos
7.
Sensors (Basel) ; 18(12)2018 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-30544689

RESUMO

This paper presents non-contact vital sign monitoring in neonates, based on image processing, where a standard color camera captures the plethysmographic signal and the heart and breathing rates are processed and estimated online. It is important that the measurements are taken in a non-invasive manner, which is imperceptible to the patient. Currently, many methods have been proposed for non-contact measurement. However, to the best of the authors' knowledge, it has not been possible to identify methods with low computational costs and a high tolerance to artifacts. With the aim of improving contactless measurement results, the proposed method based on the computer vision technique is enhanced to overcome the mentioned drawbacks. The camera is attached to an incubator in the Neonatal Intensive Care Unit and a single area in the neonate's diaphragm is monitored. Several factors are considered in the stages of image acquisition, as well as in the plethysmographic signal formation, pre-filtering and filtering. The pre-filter step uses numerical analysis techniques to reduce the signal offset. The proposed method decouples the breath rate from the frequency of sinus arrhythmia. This separation makes it possible to analyze independently any cardiac and respiratory dysrhythmias. Nine newborns were monitored with our proposed method. A Bland-Altman analysis of the data shows a close correlation of the heart rates measured with the two approaches (correlation coefficient of 0.94 for heart rate (HR) and 0.86 for breath rate (BR)) with an uncertainty of 4.2 bpm for HR and 4.9 for BR (k = 1). The comparison of our method and another non-contact method considered as a standard independent component analysis (ICA) showed lower central processing unit (CPU) usage for our method (75% less CPU usage).


Assuntos
Arritmia Sinusal/diagnóstico , Monitorização Fisiológica/métodos , Fotopletismografia/métodos , Arritmia Sinusal/diagnóstico por imagem , Arritmia Sinusal/fisiopatologia , Frequência Cardíaca/fisiologia , Humanos , Recém-Nascido , Taxa Respiratória/fisiologia , Processamento de Sinais Assistido por Computador/instrumentação
9.
Pediatr Res ; 82(1): 79-86, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28388598

RESUMO

BackgroundBrain hypoxic-ischemic (HI) damage induces distant inflammatory lung damage in newborn pigs. We aimed to investigate the effects of cannabidiol (CBD) on lung damage in this scenario.MethodsNewborn piglets received intravenous vehicle, CBD, or CBD+WAY100635 (5-HT1A receptor antagonist) after HI brain damage (carotid flow interruption and FiO2 0.10 for 30 min). Total lung compliance (TLC), oxygenation index (OI), and extravascular lung water content (EVLW) were monitored for 6 h. Histological damage, interleukin (IL)-1ß concentration, and oxidative stress were assessed in brain and lung tissue. Total protein content was determined in bronchoalveolar lavage fluid (BALF).ResultsCBD prevented HI-induced deleterious effects on TLC and OI and reduced lung histological damage, modulating inflammation (decreased leukocyte infiltration and IL-1 concentration) and reducing protein content in BALF and EVLW. These effects were related to CBD-induced anti-inflammatory changes in the brain. HI did not increase oxidative stress in the lungs. In the lungs, WAY100635 blunted the beneficial effects of CBD on histological damage, IL-1 concentration, and EVLW.ConclusionsCBD reduced brain HI-induced distant lung damage, with 5-HT1A receptor involvement in these effects. Whether the effects of CBD on the lungs were due to the anti-inflammatory effects on the brain or due to the direct effects on the lungs remains to be elucidated.


Assuntos
Canabidiol/farmacologia , Hipóxia-Isquemia Encefálica/patologia , Lesão Pulmonar/tratamento farmacológico , Pulmão/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar , Modelos Animais de Doenças , Hemodinâmica , Hipóxia/metabolismo , Inflamação/tratamento farmacológico , Interleucina-1beta/metabolismo , Pulmão/fisiopatologia , Masculino , Estresse Oxidativo , Oxigênio/metabolismo , Suínos
10.
Pediatr Res ; 80(5): 710-718, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27441365

RESUMO

BACKGROUND: Cannabidiol (CBD), a nonpsychoactive cannabinoid, has shown neuroprotective actions after neonatal hypoxia-ischemia (HI) in animals. We wanted to further explore the effects of CBD, alone and in conjunction with hypothermia, in a piglet model of global HI. METHODS: Fifty-five anesthetized newborn piglets were randomized to either controls (n = 7) or HI (n = 48) by ventilation with 8% O2 until mean arterial blood pressure reached 20 mmHg and/or base excess reached -20 mmol/l. After resuscitation piglets were randomized to either: vehicle (VEH), CBD 1mg/kg, VEH+hypothermia (H) or CBD 1mg/kg+H (each n = 12). Piglets were euthanized 9.5 h after HI and plasma, urine, cerebrospinal fluid, and brain tissue were sampled for analysis. RESULTS: HI induced global damage with significantly increased neuropathology score, S100B in cerebrospinal fluid, hippocampal proton magnetic resonance spectroscopy biomarkers, plasma troponin-T, and urinary neutrophil gelatinase-associated lipocalin. CBD alone did not have any significant effects on these parameters while CBD+H reduced urinary neutrophil gelatinase-associated lipocalin compared with VEH+H (P < 0.05). Both hypothermic groups had significantly lower glutamate/N-acetylaspartate ratios (P < 0.01) and plasma troponin-T (P<0.05) levels compared with normothermic groups. CONCLUSION: In contrast to previous studies, we do not find significant protective effects of CBD after HI in piglets. Evaluation of CBD in higher doses might be warranted.


Assuntos
Canabidiol/farmacologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Pressão Sanguínea , Peso Corporal , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/patologia , Inflamação , Rim/patologia , Espectroscopia de Ressonância Magnética , Miocárdio/patologia , Estresse Oxidativo , Oxigênio , Suínos
11.
Pediatr Res ; 79(3): 401-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25950454

RESUMO

BACKGROUND: We aimed to investigate whether neonatal hypoxic-ischemic (HI) brain injury induces inflammatory lung damage. METHODS: Thus, hypoxic (HYP, FiO2 10% for 30 min), ischemic (ISC, bilateral carotid flow interruption for 30 min), or HI event was performed in 1-2-d-old piglets. Dynamic compliance (Cdyn), oxygenation index (OI), and extravascular lung water (EVLW) were monitored for 6 h. Then, histologic damage was assessed in brain and lung (lung injury severity score). Total protein content (TPC) was determined in broncoalveolar lavage fluid (BALF), and IL-1ß concentration was measured in lung and brain tissues and blood. RESULTS: Piglets without hypoxia or ischemia served as controls (SHM). HI-induced brain damage was associated with decreased Cdyn, increased OI and EVLW, and histologic lung damage (interstitial leukocyte infiltration, congestive hyperemia, and interstitial edema). BALF TPC was increased, suggesting inflammatory damage. In agreement, tissue IL-1ß concentration increased in the brain and lung, in correspondence with increased IL-1ß serum concentration. Neither HYP nor ISC alone led to brain or lung damage. CONCLUSION: HI brain damage in newborn piglets led to inflammatory lung damage, suggesting an additional mechanism accounting for the development of lung dysfunction after neonatal HI encephalopathy.


Assuntos
Encéfalo/patologia , Hipóxia-Isquemia Encefálica/patologia , Inflamação , Lesão Pulmonar/patologia , Pulmão/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Hipóxia , Interleucina-1beta/metabolismo , Pulmão/fisiopatologia , Masculino , Suínos
12.
Epilepsia ; 55(6): 791-802, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24854329

RESUMO

To present a summary of current scientific evidence about the cannabinoid, cannabidiol (CBD) with regard to its relevance to epilepsy and other selected neuropsychiatric disorders. We summarize the presentations from a conference in which invited participants reviewed relevant aspects of the physiology, mechanisms of action, pharmacology, and data from studies with animal models and human subjects. Cannabis has been used to treat disease since ancient times. Δ(9) -Tetrahydrocannabinol (Δ(9) -THC) is the major psychoactive ingredient and CBD is the major nonpsychoactive ingredient in cannabis. Cannabis and Δ(9) -THC are anticonvulsant in most animal models but can be proconvulsant in some healthy animals. The psychotropic effects of Δ(9) -THC limit tolerability. CBD is anticonvulsant in many acute animal models, but there are limited data in chronic models. The antiepileptic mechanisms of CBD are not known, but may include effects on the equilibrative nucleoside transporter; the orphan G-protein-coupled receptor GPR55; the transient receptor potential of vanilloid type-1 channel; the 5-HT1a receptor; and the α3 and α1 glycine receptors. CBD has neuroprotective and antiinflammatory effects, and it appears to be well tolerated in humans, but small and methodologically limited studies of CBD in human epilepsy have been inconclusive. More recent anecdotal reports of high-ratio CBD:Δ(9) -THC medical marijuana have claimed efficacy, but studies were not controlled. CBD bears investigation in epilepsy and other neuropsychiatric disorders, including anxiety, schizophrenia, addiction, and neonatal hypoxic-ischemic encephalopathy. However, we lack data from well-powered double-blind randomized, controlled studies on the efficacy of pure CBD for any disorder. Initial dose-tolerability and double-blind randomized, controlled studies focusing on target intractable epilepsy populations such as patients with Dravet and Lennox-Gastaut syndromes are being planned. Trials in other treatment-resistant epilepsies may also be warranted. A PowerPoint slide summarizing this article is available for download in the Supporting Information section here.


Assuntos
Anticonvulsivantes/uso terapêutico , Canabidiol/uso terapêutico , Epilepsia/tratamento farmacológico , Animais , Anticonvulsivantes/farmacologia , Encéfalo/efeitos dos fármacos , Canabidiol/farmacologia , Canabinoides/farmacologia , Canabinoides/uso terapêutico , Epilepsias Mioclônicas/tratamento farmacológico , Humanos , Deficiência Intelectual/tratamento farmacológico , Síndrome de Lennox-Gastaut , Transtornos Mentais/tratamento farmacológico , Fitoterapia , Extratos Vegetais/uso terapêutico , Convulsões/tratamento farmacológico , Espasmos Infantis/tratamento farmacológico
13.
Eur J Pharmacol ; 972: 176554, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38582276

RESUMO

BACKGROUND: Currently there is no effective treatment for neonatal stroke, an acute neurologic syndrome with sequelae, due to focal ischemic, thrombotic, or hemorrhagic event occurring in the perinatal period. VCE-004.8, an aminoquinone exhibiting activity on CB2 and PPARγ receptors, is neuroprotective in adult mice models of acute and chronic brain damaging conditions. We hereby aimed to study VCE-004.8 neuroprotection in a rat model of neonatal stroke. METHODS: 7-day-old (P7) Wistar rats of both sexes were submitted to Middle Cerebral Artery Occlusion (MCAO), receiving i.p. 30 min after vehicle (MCAO + VEH) or VCE-004.8 5 mg/kg (MCAO + VCE). Non-occluded rats served as controls (SHAM). MCAO consequences were assessed at P14 by MRI, histological (TUNEL staining), biochemical (lactate/n-acetyl aspartate ratio by 1H-NMR spectroscopy) and motor studies (grasp test), and at P37 assessing myelination (MBP signal), hemiparesis and hyperlocomotion. Effects of VCE-004.8 on excitotoxicity (glutamate/n-acetyl aspartate, 1H-NMR), oxidative stress (protein nitrosylation, Oxyblot) and neuroinflammation (Toll-like receptor 4 and TNFa expression, Western blot) were assessed at P14. Therapeutic window was assessed by delaying drug administration for 12 or 18 h. RESULTS: Post-MCAO administration of VCE-004.8 reduced the volume of infarct and histological and biochemical brain damage, reducing hyperlocomotion, restoring motor performance and preserving myelination, in a manner linked to the modulation of excitotoxicity, oxidative stress and neuroinflammation. VCE-004.8 was still effective being administered 12-18 h post-insult. CONCLUSIONS: These data suggest that this drug could be effective for the treatment of stroke in newborns.


Assuntos
Animais Recém-Nascidos , Modelos Animais de Doenças , Fármacos Neuroprotetores , Estresse Oxidativo , Ratos Wistar , Animais , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Masculino , Ratos , Feminino , Estresse Oxidativo/efeitos dos fármacos , Acidente Vascular Cerebral/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/complicações , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia
14.
Neurotherapeutics ; 21(2): e00326, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38301326

RESUMO

Intraventricular hemorrhage (IVH) is an important cause of long-term disability in extremely preterm infants, with no current treatment. This study assessed the potential neuroprotective effects of cannabidiol (CBD) in an IVH model using immature rats. IVH was induced in 1-day-old (P1) Wistar rats by left periventricular injection of Clostridial collagenase. Some rats received CBD prenatally (10 â€‹mg/kg i.p. to the dam) and then 5 â€‹mg/kg i.p. 6, 30 and 54 â€‹h after IVH (IVH+CBD, n â€‹= â€‹30). Other IVH rats received vehicle (IVH+VEH, n â€‹= â€‹34) and vehicle-treated non-IVH rats served as controls (SHM, n â€‹= â€‹29). Rats were humanely killed at P6, P14 or P45. Brain damage (motor and memory performance, area of damage, Lactate/N-acetylaspartate ratio), white matter injury (ipsilateral hemisphere and corpus callosum volume, oligodendroglial cell density and myelin basic protein signal), blood-brain barrier (BBB) integrity (Mfsd2a, occludin and MMP9 expression, gadolinium leakage), inflammation (TLR4, NFκB and TNFα expression, infiltration of pro-inflammatory cells), excitotoxicity (Glutamate/N-acetylspartate ratio) and oxidative stress (protein nitrosylation) were then evaluated. CBD prevented the long-lasting motor and cognitive consequences of IVH, reduced brain damage in the short- and long-term, protected oligodendroglial cells preserving adequate myelination and maintained BBB integrity. The protective effects of CBD were associated with the modulation of inflammation, excitotoxicity and oxidative stress. In conclusion, in immature rats, CBD reduced IVH-induced brain damage and its short- and long-term consequences, showing robust and pleiotropic neuroprotective effects. CBD is a potential candidate to ameliorate IVH-induced immature brain damage.


Assuntos
Lesões Encefálicas , Canabidiol , Fármacos Neuroprotetores , Humanos , Recém-Nascido , Animais , Ratos , Barreira Hematoencefálica , Canabidiol/farmacologia , Canabidiol/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Recém-Nascido Prematuro/metabolismo , Ratos Wistar , Hemorragia Cerebral/complicações , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/metabolismo , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/etiologia , Inflamação , Modelos Animais de Doenças
15.
Br J Clin Pharmacol ; 75(2): 323-33, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22625422

RESUMO

Cannabidiol (CBD) is a phytocannabinoid with therapeutic properties for numerous disorders exerted through molecular mechanisms that are yet to be completely identified. CBD acts in some experimental models as an anti-inflammatory, anticonvulsant, anti-oxidant, anti-emetic, anxiolytic and antipsychotic agent, and is therefore a potential medicine for the treatment of neuroinflammation, epilepsy, oxidative injury, vomiting and nausea, anxiety and schizophrenia, respectively. The neuroprotective potential of CBD, based on the combination of its anti-inflammatory and anti-oxidant properties, is of particular interest and is presently under intense preclinical research in numerous neurodegenerative disorders. In fact, CBD combined with Δ(9)-tetrahydrocannabinol is already under clinical evaluation in patients with Huntington's disease to determine its potential as a disease-modifying therapy. The neuroprotective properties of CBD do not appear to be exerted by the activation of key targets within the endocannabinoid system for plant-derived cannabinoids like Δ(9)-tetrahydrocannabinol, i.e. CB(1) and CB(2) receptors, as CBD has negligible activity at these cannabinoid receptors, although certain activity at the CB(2) receptor has been documented in specific pathological conditions (i.e. damage of immature brain). Within the endocannabinoid system, CBD has been shown to have an inhibitory effect on the inactivation of endocannabinoids (i.e. inhibition of FAAH enzyme), thereby enhancing the action of these endogenous molecules on cannabinoid receptors, which is also noted in certain pathological conditions. CBD acts not only through the endocannabinoid system, but also causes direct or indirect activation of metabotropic receptors for serotonin or adenosine, and can target nuclear receptors of the PPAR family and also ion channels.


Assuntos
Canabidiol/farmacologia , Doença de Huntington/tratamento farmacológico , Isquemia/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Canabinoides/farmacologia , Humanos
16.
Arch Dis Child Fetal Neonatal Ed ; 108(3): 316-318, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-35288449

RESUMO

The use of dexmedetomidine (DEX) has been extended in preterm newborns, but the effects on cerebral activity and their relationship with haemodynamic changes has not been studied.We retrospectively studied the effects of DEX administered to 10 preterm newborns, assessing amplitude-integrated EEG (aEEG) parameters, brain regional SO2 (brSO2), heart rate, non-invasive mean blood pressure (MBP), transcutaneous oxygen saturation (SpO2), venous pCO2 and haemoglobin (Hb) values, in two 6-hour periods: one starting 6 hours before the beginning of DEX perfusion and the other 6 hours afterwards.DEX infusion led to brSO2 decrease not associated to heart rate, MBP, SpO2, Hb or pCO2 variation, which suggests that brSO2 decrease could be related to local vasoconstriction. DEX infusion led to prolongation of interburst interval and reduction of cycling. Such effects, not been described so far, should be considered in the assessment of aEEG traces after DEX administration to avoid misinterpretations regarding patient's prognosis. More studies are needed to assess the safety of DEX use in the newborn.


Assuntos
Dexmedetomidina , Recém-Nascido Prematuro , Lactente , Humanos , Recém-Nascido , Recém-Nascido Prematuro/fisiologia , Dexmedetomidina/efeitos adversos , Estudos Retrospectivos , Hemodinâmica , Frequência Cardíaca
17.
Biomed Pharmacother ; 162: 114715, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37075665

RESUMO

One of the main global causes of mortality and morbidity is traumatic brain injury (TBI). Neuroinflammation and brain-blood barrier (BBB) disruption play a pivotal role in the pathogenesis of acute and chronic TBI onset. The activation of the hypoxia pathway is a promising approach for CNS neurodegenerative diseases, including TBI. Herein, we have studied the efficacy of VCE-005.1, a betulinic acid hydroxamate, against acute neuroinflammation in vitro and on a TBI mouse model. The effect of VCE-005.1 on the HIF pathway in endothelial vascular cells was assessed by western blot, gene expression, in vitro angiogenesis, confocal analysis and MTT assays. In vivo angiogenesis was evaluated through a Matrigel plug model and a mouse model of TBI induced by a controlled cortical impact (CCI) was used to assess VCE-005.1 efficacy. VCE-005.1 stabilized HIF-1α through a mechanism that involved AMPK and stimulated the expression of HIF-dependent genes. VCE-005.1 protected vascular endothelial cells under prooxidant and pro-inflammatory conditions by enhancing TJ protein expression and induced angiogenesis both in vitro and in vivo. Furthermore, in CCI model, VCE-005.1 greatly improved locomotor coordination, increased neovascularization and preserved BBB integrity that paralleled with a large reduction of peripheral immune cells infiltration, recovering AMPK expression and reducing apoptosis in neuronal cells. Taken together, our results demonstrate that VCE-005.1 is a multitarget compound that shows anti-inflammatory and neuroprotective effects mainly by preventing BBB disruption and has the potential to be further developed pharmacologically in TBI and maybe other neurological conditions that concur with neuroinflammation and BBB disruption.


Assuntos
Ácido Betulínico , Lesões Encefálicas Traumáticas , Camundongos , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Doenças Neuroinflamatórias , Proteínas Quinases Ativadas por AMP/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Barreira Hematoencefálica/metabolismo , Hipóxia/patologia , Camundongos Endogâmicos C57BL
18.
Antioxidants (Basel) ; 12(10)2023 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-37891918

RESUMO

Oxidative stress (OS) constitutes a pivotal factor within the mechanisms underlying brain damage, for which the immature brain is particularly vulnerable. This vulnerability is caused by the abundance of immature oligodendrocytes in the immature brain, which are highly susceptible to OS-induced harm. Consequently, any injurious process involving OS within the immature brain can lead to long-term myelination impairment. Among the detrimental repercussions of OS, protein carbonylation stands out as a prominently deleterious consequence. Noteworthy elevation of protein carbonylation is observable across diverse models of neonatal brain injury, following both diffuse and focal hypoxic-ischemic insults, as well as intraventricular hemorrhage, in diverse animal species encompassing rodents and larger mammals, and at varying stages of brain development. In the immature brain, protein carbonylation manifests as a byproduct of reactive nitrogen species, bearing profound implications for cell injury, particularly in terms of inflammation amplification. Moreover, protein carbonylation appears as a therapeutic target for mitigating neonatal brain damage. The administration of a potent antioxidant, such as cannabidiol, yields substantial neuroprotective effects. These encompass the reduction in cerebral damage, restoration of neurobehavioral performance, and preservation of physiological myelination. Such effects are linked to the modulation of protein carbonylation. The assessment of protein carbonylation emerges as a reliable method for comprehending the intricate mechanisms underpinning damage and neuroprotection within neonatal brain injury.

19.
Transl Stroke Res ; 14(3): 397-408, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-35419730

RESUMO

There is an increasing evidence of the neuroprotective effects of hypoxia inducing factor prolyl-hydroxylase inhibitors (HIF-PHDi) after hypoxic-ischemic (HI) brain damage (HIBD). We studied the neuroprotective effects of betulinic hydroxamate (BAH), a novel B55α/PP2A activator that dephosphorylates and inhibits PHD2 activity, in a rat model of neonatal HIBD. Seven-day-old (P7) Wistar rats were exposed to hypoxia after left carotid artery electrocoagulation and then received vehicle (HI + VEH) or BAH 3 mg/kg i.p. 30 min post-insult. Brain damage was assessed by magnetic resonance imaging (MRI) and neurobehavioral studies testing motor and cognitive performance at P14 and P37, as well as immunohistochemical studies (TUNEL and myelin basic protein (MBP) signal) at P37. Mechanisms of damage were assessed at P14 determining excitotoxicity (glutamate/N-acetylaspartate ratio by H+-magnetic resonance spectroscopy), oxidative stress (protein nitrosylation by Oxyblot), and inflammation (cytokine and chemokine concentration). BAH reduced brain damage volume and cell death, preventing the development of motor and working memory deficits. BAH showed a robust protective effect on myelination, restoring MBP expression at P37. BAH modulated excitotoxicity, oxidative stress, and inflammation. Most neuroprotective effects were still present despite BAH administration was delayed for 12 h, whereas beneficial effects on motor strength at P14 and on cell death and myelination at P37 were preserved even when BAH administration was delayed for 24 h. In conclusion, BAH appears as an effective neuroprotective treatment for neonatal HIBD in a manner associated with the modulation of excitotoxicity, oxidative stress, and inflammation, showing a broad therapeutic window.


Assuntos
Lesões Encefálicas , Hipóxia-Isquemia Encefálica , Fármacos Neuroprotetores , Animais , Ratos , Animais Recém-Nascidos , Ratos Wistar , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ácido Betulínico , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia/tratamento farmacológico , Lesões Encefálicas/metabolismo , Isquemia/patologia , Encéfalo/metabolismo
20.
Front Pediatr ; 10: 880786, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35692972

RESUMO

Diastolic dysfunction often complicates myocardial ischemia with increased mortality rates. However, less is known about diastolic function after perinatal asphyxia in neonates with hypoxic-ischemic encephalopathy (HIE) during therapeutic hypothermia (TH) and rewarming. Aim: The aim of this study was to assess diastolic function with tissue Doppler imaging (TDI) in neonates with moderate-severe HIE during TH and rewarming. Method: Newborns at >36 weeks' gestation with moderate-severe HIE treated with TH were evaluated with targeted neonatal echocardiography (TNE), including TDI, within 24 h of TH initiation (T1), at 48-72 h of treatment (T2), and after rewarming (T3). These retrospective data were collected and compared with a control group of healthy babies at >36 weeks' gestation that was prospectively evaluated following the same protocol. Results: A total of 21 patients with HIE + TH and 15 controls were included in the study. Myocardial relaxation before the onset of biventricular filling was prolonged in the HIE + TH group during TH with significantly longer isovolumic relaxation time (IVRT') in the left ventricle (LV), the septum, and the right ventricle (RV). This was associated with slower RV early diastolic velocity (e') and prolonged filling on T1. Total isovolumic time (t-IVT; isovolumic contraction time [IVCT'] + IVRT') and myocardial performance index (MPI') were globally increased in asphyxiated neonates. All these differences persisted after correction for heart rate (HR) and normalized after rewarming. TDI parameters assessing late diastole (a' velocity or e'/a' and E/e' ratios) did not differ between groups. Conclusion: TDI evaluation in our study demonstrated a pattern of early diastolic dysfunction during TH that normalized after rewarming, whereas late diastole seemed to be preserved. Our data also suggest a possible involvement of impaired twist/untwist motion and dyssynchrony. More studies are needed to investigate the impact and therapeutic implication of diastolic dysfunction in these babies, as well as to clarify the role of TH in these findings.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA