Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
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
2.
J Neuroinflammation ; 15(1): 158, 2018 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-29793509

RESUMO

BACKGROUND: Because of their low levels of expression and the inadequacy of current research tools, CB2 cannabinoid receptors (CB2R) have been difficult to study, particularly in the brain. This receptor is especially relevant in the context of neuroinflammation, so novel tools are needed to unveil its pathophysiological role(s). METHODS: We have generated a transgenic mouse model in which the expression of enhanced green fluorescent protein (EGFP) is under the control of the cnr2 gene promoter through the insertion of an Internal Ribosomal Entry Site followed by the EGFP coding region immediately 3' of the cnr2 gene and crossed these mice with mice expressing five familial Alzheimer's disease (AD) mutations (5xFAD). RESULTS: Expression of EGFP in control mice was below the level of detection in all regions of the central nervous system (CNS) that we examined. CB2R-dependent-EGFP expression was detected in the CNS of 3-month-old AD mice in areas of intense inflammation and amyloid deposition; expression was coincident with the appearance of plaques in the cortex, hippocampus, brain stem, and thalamus. The expression of EGFP increased as a function of plaque formation and subsequent microgliosis and was restricted to microglial cells located in close proximity to neuritic plaques. AD mice with CB2R deletion exhibited decreased neuritic plaques with no changes in IL1ß expression. CONCLUSIONS: Using a novel reporter mouse line, we found no evidence for CB2R expression in the healthy CNS but clear up-regulation in the context of amyloid-triggered neuroinflammation. Data from CB2R null mice indicate that they play a complex role in the response to plaque formation.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/metabolismo , Regulação da Expressão Gênica/genética , Receptor CB2 de Canabinoide/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/metabolismo , Análise de Variância , Animais , Encéfalo/patologia , Antígeno CD11b/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Fragmentos de Peptídeos/metabolismo , Fosfopiruvato Hidratase/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Receptor CB2 de Canabinoide/genética
3.
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
4.
J Biol Chem ; 289(32): 21960-72, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-24942731

RESUMO

The G protein-coupled receptors CB2 (CB2R) and GPR55 are overexpressed in cancer cells and human tumors. Because a modulation of GPR55 activity by cannabinoids has been suggested, we analyzed whether this receptor participates in cannabinoid effects on cancer cells. Here we show that CB2R and GPR55 form heteromers in cancer cells, that these structures possess unique signaling properties, and that modulation of these heteromers can modify the antitumoral activity of cannabinoids in vivo. These findings unveil the existence of previously unknown signaling platforms that help explain the complex behavior of cannabinoids and may constitute new targets for therapeutic intervention in oncology.


Assuntos
Neoplasias/metabolismo , Receptor CB2 de Canabinoide/química , Receptor CB2 de Canabinoide/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Canabinoides/metabolismo , Canabinoides/farmacologia , Linhagem Celular Tumoral , Dronabinol/farmacologia , Feminino , Marcação de Genes , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/genética , Estrutura Quaternária de Proteína , RNA Interferente Pequeno/genética , Receptor CB2 de Canabinoide/genética , Receptores de Canabinoides , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
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
6.
Brain ; 134(Pt 1): 119-36, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20929960

RESUMO

Endocannabinoids act as neuromodulatory and neuroprotective cues by engaging type 1 cannabinoid receptors. These receptors are highly abundant in the basal ganglia and play a pivotal role in the control of motor behaviour. An early downregulation of type 1 cannabinoid receptors has been documented in the basal ganglia of patients with Huntington's disease and animal models. However, the pathophysiological impact of this loss of receptors in Huntington's disease is as yet unknown. Here, we generated a double-mutant mouse model that expresses human mutant huntingtin exon 1 in a type 1 cannabinoid receptor-null background, and found that receptor deletion aggravates the symptoms, neuropathology and molecular pathology of the disease. Moreover, pharmacological administration of the cannabinoid Δ(9)-tetrahydrocannabinol to mice expressing human mutant huntingtin exon 1 exerted a therapeutic effect and ameliorated those parameters. Experiments conducted in striatal cells show that the mutant huntingtin-dependent downregulation of the receptors involves the control of the type 1 cannabinoid receptor gene promoter by repressor element 1 silencing transcription factor and sensitizes cells to excitotoxic damage. We also provide in vitro and in vivo evidence that supports type 1 cannabinoid receptor control of striatal brain-derived neurotrophic factor expression and the decrease in brain-derived neurotrophic factor levels concomitant with type 1 cannabinoid receptor loss, which may contribute significantly to striatal damage in Huntington's disease. Altogether, these results support the notion that downregulation of type 1 cannabinoid receptors is a key pathogenic event in Huntington's disease, and suggest that activation of these receptors in patients with Huntington's disease may attenuate disease progression.


Assuntos
Corpo Estriado/metabolismo , Doença de Huntington/genética , Neurônios/metabolismo , Receptor CB1 de Canabinoide/genética , Análise de Variância , Animais , Western Blotting , Sobrevivência Celular , Dronabinol/farmacologia , Hormônio Liberador de Hormônio do Crescimento/análogos & derivados , Doença de Huntington/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Receptor CB1 de Canabinoide/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Teste de Desempenho do Rota-Rod
7.
Sci Rep ; 12(1): 7895, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35551497

RESUMO

Studies have shown that anodically grown TiO2 nanotubes (TNTs) exhibit excellent biocompatibility. However, TiO2 nanowires (TNWs) have received less attention. The objective of this study was to investigate the proliferation of osteoblast precursor cells on the surfaces of TNWs grown by electrochemical anodization of a Ti-35Nb-7Zr-5Ta (TNZT) alloy. TNT and flat TNZT surfaces were used as control samples. MC3T3-E1 cells were cultured on the surfaces of the samples for up to 5 days, and cell viability and proliferation were investigated using fluorescence microscopy, colorimetric assay, and scanning electron microscopy. The results showed lower cell proliferation rates on the TNW surface compared to control samples without significant differences in cell survival among experimental conditions. Contact angles measurements showed a good level of hydrophilicity for the TNWs, however, their relatively thin diameter and their high density may have affected cell proliferation. Although more research is necessary to understand all the parameters affecting biocompatibility, these TiO2 nanostructures may represent promising tools for the treatment of bone defects and regeneration of bone tissue, among other applications.


Assuntos
Nanotubos , Nanofios , Ligas/química , Proliferação de Células , Microscopia Eletrônica de Varredura , Nanotubos/química , Osteoblastos , Propriedades de Superfície , Titânio/química , Titânio/farmacologia
8.
J Neurosci Res ; 89(9): 1509-18, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21674569

RESUMO

We studied whether combinations of botanical extracts enriched in either Δ(9)-tetrahydrocannabinol (Δ(9)-THC) or cannabidiol (CBD), which are the main constituents of the cannabis-based medicine Sativex, provide neuroprotection in rat models of Huntington's disease (HD). We used rats intoxicated with 3-nitropropionate (3NP) that were given combinations of Δ(9)-THC- and CBD-enriched botanical extracts. The issue was also studied in malonate-lesioned rats. The administration of Δ(9)-THC- and CBD-enriched botanical extracts combined in a ratio of 1:1 as in Sativex attenuated 3NP-induced GABA deficiency, loss of Nissl-stained neurons, down-regulation of CB(1) receptor and IGF-1 expression, and up-regulation of calpain expression, whereas it completely reversed the reduction in superoxide dismutase-1 expression. Similar responses were generally found with other combinations of Δ(9)-THC- and CBD-enriched botanical extracts, suggesting that these effects are probably related to the antioxidant and CB(1) and CB(2) receptor-independent properties of both phytocannabinoids. In fact, selective antagonists for both receptor types, i.e., SR141716 and AM630, respectively, were unable to prevent the positive effects on calpain expression caused in 3NP-intoxicated rats by the 1:1 combination of Δ(9)-THC and CBD. Finally, this combination also reversed the up-regulation of proinflammatory markers such as inducible nitric oxide synthase observed in malonate-lesioned rats. In conclusion, this study provides preclinical evidence in support of a beneficial effect of the cannabis-based medicine Sativex as a neuroprotective agent capable of delaying disease progression in HD, a disorder that is currently poorly managed in the clinic, prompting an urgent need for clinical trials with agents showing positive results in preclinical studies.


Assuntos
Canabinoides/uso terapêutico , Núcleo Caudado/efeitos dos fármacos , Doença de Huntington/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Fitoterapia , Putamen/efeitos dos fármacos , Animais , Canabidiol/uso terapêutico , Núcleo Caudado/citologia , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Dronabinol/uso terapêutico , Combinação de Medicamentos , Doença de Huntington/patologia , Masculino , Neostriado/efeitos dos fármacos , Neostriado/patologia , Extratos Vegetais/uso terapêutico , Putamen/citologia , Ratos , Ratos Sprague-Dawley
9.
Pediatr Res ; 70(3): 272-7, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21654550

RESUMO

Newborn piglets exposed to acute hypoxia-ischemia (HI) received i.v. cannabidiol (HI + CBD) or vehicle (HI + VEH). In HI + VEH, 72 h post-HI brain activity as assessed by amplitude-integrated EEG (aEEG) had only recovered to 42 ± 9% of baseline, near-infrared spectroscopy (NIRS) parameters remained lower than normal, and neurobehavioral performance was abnormal (27.8 ± 2.3 points, normal 36). In the brain, there were fewer normal and more pyknotic neurons, while astrocytes were less numerous and swollen. Cerebrospinal fluid concentration of neuronal-specific enolase (NSE) and S100ß protein and brain tissue percentage of TNFα(+) cells were all higher. In contrast, in HI + CBD, aEEG had recovered to 86 ± 5%, NIRS parameters increased, and the neurobehavioral score normalized (34.3 ± 1.4 points). HI induced histological changes, and NSE and S100ß concentration and TNFα(+) cell increases were suppressed by CBD. In conclusion, post-HI administration of CBD protects neurons and astrocytes, leading to histological, functional, biochemical, and neurobehavioral improvements.


Assuntos
Encéfalo/patologia , Canabidiol/uso terapêutico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/patologia , Fármacos Neuroprotetores/uso terapêutico , Animais , Animais Recém-Nascidos , Comportamento Animal , Encéfalo/efeitos dos fármacos , Eletroencefalografia , Humanos , Hipóxia-Isquemia Encefálica/fisiopatologia , Hipóxia-Isquemia Encefálica/reabilitação , Testes Neuropsicológicos , Sus scrofa
10.
Brain ; 132(Pt 11): 3152-64, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19805493

RESUMO

Cannabinoid-derived drugs are promising agents for the development of novel neuroprotective strategies. Activation of neuronal CB(1) cannabinoid receptors attenuates excitotoxic glutamatergic neurotransmission, triggers prosurvival signalling pathways and palliates motor symptoms in animal models of neurodegenerative disorders. However, in Huntington's disease there is a very early downregulation of CB(1) receptors in striatal neurons that, together with the undesirable psychoactive effects triggered by CB(1) receptor activation, foster the search for alternative pharmacological treatments. Here, we show that CB(2) cannabinoid receptor expression increases in striatal microglia of Huntington's disease transgenic mouse models and patients. Genetic ablation of CB(2) receptors in R6/2 mice, that express human mutant huntingtin exon 1, enhanced microglial activation, aggravated disease symptomatology and reduced mice lifespan. Likewise, induction of striatal excitotoxicity in CB(2) receptor-deficient mice by quinolinic acid administration exacerbated brain oedema, microglial activation, proinflammatory-mediator state and medium-sized spiny neuron degeneration. Moreover, administration of CB(2) receptor-selective agonists to wild-type mice subjected to excitotoxicity reduced neuroinflammation, brain oedema, striatal neuronal loss and motor symptoms. Studies on ganciclovir-induced depletion of astroglial proliferation in transgenic mice expressing thymidine kinase under the control of the glial fibrillary acidic protein promoter excluded the participation of proliferating astroglia in CB(2) receptor-mediated actions. These findings support a pivotal role for CB(2) receptors in attenuating microglial activation and preventing neurodegeneration that may pave the way to new therapeutic strategies for neuroprotection in Huntington's disease as well as in other neurodegenerative disorders with a significant excitotoxic component.


Assuntos
Doença de Huntington , Microglia/metabolismo , Fármacos Neuroprotetores/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Animais , Antibacterianos/farmacologia , Biomarcadores/metabolismo , Corpo Estriado/citologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Corpo Estriado/patologia , Humanos , Proteína Huntingtina , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Doença de Huntington/fisiopatologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Minociclina/farmacologia , Degeneração Neural/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ácido Quinolínico/farmacologia , Receptor CB2 de Canabinoide/genética , Teste de Desempenho do Rota-Rod , Convulsões/fisiopatologia
11.
Front Pharmacol ; 10: 1131, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31611802

RESUMO

Neonatal hypoxia-ischemia (HI) is a risk factor for myelination disturbances, a key factor for cerebral palsy. Cannabidiol (CBD) protects neurons and glial cells after HI insult in newborn animals. We hereby aimed to study CBD's effects on long-lasting HI-induced myelination deficits in newborn rats. Thus, P7 Wistar rats received s.c. vehicle (HV) or cannabidiol (HC) after HI brain damage (left carotid artery electrocoagulation plus 10% O2 for 112 min). Controls were non-HI pups. At P37, neurobehavioral tests were performed and immunohistochemistry [quantifying mature oligodendrocyte (mOL) populations and myelin basic protein (MBP) density] and electron microscopy (determining axon number, size, and myelin thickness) studies were conducted in cortex (CX) and white matter (WM). Expression of brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) were analyzed by western blot at P14. HI reduced mOL or MBP in CX but not in WM. In both CX and WM, axon density and myelin thickness were reduced. MBP impairment correlated with functional deficits. CBD administration resulted in normal function associated with normal mOL and MBP, as well as normal axon density and myelin thickness in all areas. CBD's effects were not associated with increased BDNF or GDNF expression. In conclusion, HI injury in newborn rats resulted in long-lasting myelination disturbance, associated with functional impairment. CBD treatment preserved function and myelination, likely as a part of a general neuroprotective effect.

12.
Biochem Pharmacol ; 157: 202-209, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30195729

RESUMO

The search for novel therapies for the treatment of Alzheimer's disease is an urgent need, due to the current paucity of available pharmacological tools and the recent failures obtained in clinical trials. Among other strategies, the modulation of amyloid-triggered neuroinflammation by the endocannabinoid system seems of relevance. Previous data indicate that the enhancement of the endocannabinoid tone through the inhibition of the enzymes responsible for the degradation of their main endogenous ligands may render beneficial effects. Based on previously reported data, in which we described a paradoxical effect of the genetic deletion of the fatty acid amide hydrolase, we here aimed to expand our knowledge on the role of the endocannabinoid system in the context of Alzheimer's disease. To that end, we inhibited the production of interleukin-1ß, one of the main inflammatory cytokines involved in the neuroinflammation triggered by amyloid peptides, in a transgenic mouse model of this disease by using minocycline, a drug known to impair the synthesis of this cytokine. Our data suggest that interleukin-1ß may be instrumental in order to achieve the beneficial effects derived of fatty acid amide hydrolase genetic inactivation. This could be appreciated at the molecular (cytokine expression, amyloid production, plaque deposition) as well as behavioral levels (memory impairment). We here describe a previously unknown link between the endocannabinoid system and interleukin-1ß in the context of Alzheimer's disease that open new possibilities for the development of novel therapeutics.


Assuntos
Doença de Alzheimer/metabolismo , Amidoidrolases/genética , Interleucina-1beta/fisiologia , Doença de Alzheimer/genética , Animais , Citocinas/biossíntese , Citocinas/genética , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Microglia/citologia , Minociclina/farmacologia , Fenótipo
13.
CNS Neurol Disord Drug Targets ; 16(1): 102-108, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27686886

RESUMO

BACKGROUND: A relevant therapeutic time window (TTW) is an important criterion for considering the clinical relevance of a substance preventing newborn hypoxic-ischemic (HI) brain damage. OBJECTIVE: To test the TTW of the neuroprotective effects of cannabidol (CBD), a non-psychoactive cannabinoid in a model of newborn HI brain damage. METHOD: 9-10 day-old C57BL6 mice underwent a HI insult (10% oxygen for 90 min after left carotid artery electrocoagulation). Then, CBD 1 mg/kg or vehicle were administered s.c. 15 min, or 1, 3, 6, 12, 18 or 24 h after the end of the HI insult. Seven days later brain damage was assessed using T2W Magnetic Resonance Imaging scan (ipsilateral hemisphere volume loss, IVHL) and histological studies: Nissl staining (neuropathological score), TUNEL staining (apoptotic damage) and immunohistochemistry with glial fibrillary acidic protein (astrocyte viability) or ionized calcium binding adaptor molecule (microglial activation). RESULTS: CBD administered up to 18 h after HI reduced IHVL and neuropathological score by 60%, TUNEL+ count by 90% and astrocyte damage by 50%. In addition, CBD blunted the HI-induced increase in microglial population. When CBD administration was delayed 24 h, however, the neuroprotective effect was lost in terms of IHVL, apoptosis or astrogliosis reduction. CONCLUSION: CBD shows a TTW of 18 h when administered to HI newborn mice, which represents a broader TTW than reported for other neuroprotective treatments including hypothermia.


Assuntos
Canabidiol/uso terapêutico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Canabidiol/administração & dosagem , Relação Dose-Resposta a Droga , Gliose , Hipóxia-Isquemia Encefálica/patologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Injeções Subcutâneas , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/patologia , Fármacos Neuroprotetores/administração & dosagem
14.
J Med Chem ; 59(5): 1840-53, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26789378

RESUMO

The orphan G protein-coupled receptor GPR55 has been proposed as a novel receptor of the endocannabinoid system. However, the validity of this categorization is still under debate mainly because of the lack of potent and selective agonists and antagonists of GPR55. Binding assays are not yet available for GPR55 screening, and discrepancies in GPR55 mediated signaling pathways have been reported. In this context, we have designed and synthesized novel GPR55 ligands based on a chromenopyrazole scaffold. Appraisal of GPR55 activity was accomplished using a label-free cell-impedance-based assay in hGPR55-HEK293 cells. The real-time impedance responses provided an integrative assessment of the cellular consequence to GPR55 stimulation taking into account the different possible signaling pathways. Potent GPR55 partial agonists (14b, 18b, 19b, 20b, and 21-24) have been identified; one of them (14b) being selective versus classical cannabinoid receptors. Upon antagonist treatment, chromenopyrazoles 21-24 inhibited lysophosphatidylinositol (LPI) effect. One of these GPR55 antagonists (21) is fully selective versus classic cannabinoid receptors. Compared to LPI, the predicted physicochemical parameters of the new compounds suggest a clear pharmacokinetic improvement.


Assuntos
Ensaios Enzimáticos/métodos , Pirazóis/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Relação Dose-Resposta a Droga , Impedância Elétrica , Células HEK293 , Humanos , Estrutura Molecular , Pirazóis/síntese química , Pirazóis/química , Receptores de Canabinoides , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
15.
Eur J Med Chem ; 112: 66-80, 2016 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-26890113

RESUMO

Previous studies have investigated the relevance and structure-activity relationships (SARs) of pyrazole derivatives in relation with cannabinoid receptors, and the series of tricyclic 1,4-dihydroindeno[1,2-c]pyrazoles emerged as potent CB2 receptor ligands. In the present study, novel 1,4-dihydroindeno[1,2-c]pyrazole and 1H-benzo[g]indazole carboxamides containing a cyclopropyl or a cyclohexyl substituent were designed and synthesized to evaluate the influence of these structural modifications towards CB1 and CB2 receptor affinities. Among these derivatives, compound 15 (6-cyclopropyl-1-(2,4-dichlorophenyl)-N-(adamantan-1-yl)-1,4-dihydroindeno[1,2-c]pyrazole-3-carboxamide) showed the highest CB2 receptor affinity (Ki = 4 nM) and remarkable selectivity (KiCB1/KiCB2 = 2232), whereas a similar affinity, within the nM range, was seen for the fenchyl derivative (compound 10: Ki = 6 nM), for the bornyl analogue (compound 14: Ki = 38 nM) and, to a lesser extent, for the aminopiperidine derivative (compound 6: Ki = 69 nM). Compounds 10 and 14 were also highly selective for the CB2 receptor (KiCB1/KiCB2 > 1000), whereas compound 6 was relatively selective (KiCB1/KiCB2 = 27). The four compounds were also subjected to GTPγS binding analysis showing antagonist/inverse agonist properties (IC50 for compound 14 = 27 nM, for 15 = 51 nM, for 10 = 80 nM and for 6 = 294 nM), and this activity was confirmed for the three more active compounds in a CB2 receptor-specific in vitro bioassay consisting in the quantification of prostaglandin E2 release by LPS-stimulated BV2 cells, in the presence and absence of WIN55,212-2 and/or the investigated compounds. Modelling studies were also conducted with the four compounds, which conformed with the structural requirements stated for the binding of antagonist compounds to the human CB2 receptor.


Assuntos
Pirazóis/química , Pirazóis/farmacologia , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/antagonistas & inibidores , Humanos , Ligantes , Simulação de Acoplamento Molecular , Pirazóis/síntese química , Receptor CB2 de Canabinoide/metabolismo , Relação Estrutura-Atividade
16.
Sci Rep ; 6: 21703, 2016 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-26887982

RESUMO

Scleroderma is a group of rare diseases associated with early and transient inflammation and vascular injury, followed by fibrosis affecting the skin and multiple internal organs. Fibroblast activation is the hallmark of scleroderma, and disrupting the intracellular TGFß signaling may provide a novel approach to controlling fibrosis. Because of its potential role in modulating inflammatory and fibrotic responses, both PPARγ and CB2 receptors represent attractive targets for the development of cannabinoid-based therapies. We have developed a non-thiophilic and chemically stable derivative of the CBD quinol (VCE-004.8) that behaves as a dual agonist of PPARγ and CB2 receptors, VCE-004.8 inhibited TGFß-induced Col1A2 gene transcription and collagen synthesis. Moreover, VCE-004.8 inhibited TGFß-mediated myofibroblast differentiation and impaired wound-healing activity. The anti-fibrotic efficacy in vivo was investigated in a murine model of dermal fibrosis induced by bleomycin. VCE-004.8 reduced dermal thickness, blood vessels collagen accumulation and prevented mast cell degranulation and macrophage infiltration in the skin. These effects were impaired by the PPARγ antagonist T0070907 and the CB2 antagonist AM630. In addition, VCE-004.8 downregulated the expression of several key genes associated with fibrosis, qualifying this semi-synthetic cannabinoid as a novel compound for the management of scleroderma and, potentially, other fibrotic diseases.


Assuntos
Bleomicina/efeitos adversos , Canabinoides/administração & dosagem , Canabinoides/síntese química , PPAR gama/metabolismo , Receptor CB2 de Canabinoide/metabolismo , Esclerodermia Localizada/tratamento farmacológico , Animais , Canabinoides/química , Canabinoides/farmacologia , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Hidroquinonas/administração & dosagem , Hidroquinonas/síntese química , Hidroquinonas/química , Hidroquinonas/farmacologia , Camundongos , Células NIH 3T3 , PPAR gama/agonistas , Receptor CB2 de Canabinoide/agonistas , Esclerodermia Localizada/induzido quimicamente , Esclerodermia Localizada/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Neuropharmacology ; 71: 282-91, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23587650

RESUMO

The mechanisms underlying the neuroprotective effects of cannabidiol (CBD) were studied in vivo using a hypoxic-ischemic (HI) brain injury model in newborn pigs. One- to two-day-old piglets were exposed to HI for 30 min by interrupting carotid blood flow and reducing the fraction of inspired oxygen to 10%. Thirty minutes after HI, the piglets were treated with vehicle (HV) or 1 mg/kg CBD, alone (HC) or in combination with 1 mg/kg of a CB2 receptor antagonist (AM630) or a serotonin 5HT(1A) receptor antagonist (WAY100635). HI decreased the number of viable neurons and affected the amplitude-integrated EEG background activity as well as different prognostic proton-magnetic-resonance-spectroscopy (H(±)-MRS)-detectable biomarkers (lactate/N-acetylaspartate and N-acetylaspartate/choline ratios). HI brain damage was also associated with increases in excitotoxicity (increased glutamate/N-acetylaspartate ratio), oxidative stress (decreased glutathione/creatine ratio and increased protein carbonylation) and inflammation (increased brain IL-1 levels). CBD administration after HI prevented all these alterations, although this CBD-mediated neuroprotection was reversed by co-administration of either WAY100635 or AM630, suggesting the involvement of CB2 and 5HT(1A) receptors. The involvement of CB2 receptors was not dependent on a CBD-mediated increase in endocannabinoids. Finally, bioluminescence resonance energy transfer studies indicated that CB2 and 5HT(1A) receptors may form heteromers in living HEK-293T cells. In conclusion, our findings demonstrate that CBD exerts robust neuroprotective effects in vivo in HI piglets, modulating excitotoxicity, oxidative stress and inflammation, and that both CB2 and 5HT(1A) receptors are implicated in these effects.


Assuntos
Canabidiol/uso terapêutico , Modelos Animais de Doenças , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Receptor CB2 de Canabinoide/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/fisiopatologia , Canabidiol/antagonistas & inibidores , Canabidiol/farmacocinética , Canabidiol/farmacologia , Antagonistas de Receptores de Canabinoides/farmacologia , Canabinoides/metabolismo , Canabinoides/farmacocinética , Canabinoides/uso terapêutico , Células HEK293 , Humanos , Hipóxia-Isquemia Encefálica/etiologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Masculino , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/antagonistas & inibidores , Fármacos Neuroprotetores/farmacocinética , Fármacos Neuroprotetores/farmacologia , Multimerização Proteica , Distribuição Aleatória , Receptor CB2 de Canabinoide/antagonistas & inibidores , Receptor CB2 de Canabinoide/genética , Receptor 5-HT1A de Serotonina/química , Receptor 5-HT1A de Serotonina/genética , Proteínas Recombinantes de Fusão/metabolismo , Antagonistas do Receptor 5-HT1 de Serotonina/farmacologia , Sus scrofa
18.
Recent Pat CNS Drug Discov ; 7(1): 41-8, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-22280340

RESUMO

Cannabinoid pharmacology has experienced a notable increase in the last 3 decades which is allowing the development of novel cannabinoid-based medicines for the treatment of different human pathologies, for example, Cesamet® (nabilone) or Marinol® (synthetic Δ9-tetrahydrocannabinol for oral administration) that were approved in 80s for the treatment of nausea and vomiting associated with chemotherapy treatment in cancer patients and in 90s for anorexiacachexia associated with AIDS therapy. Recently, the british company GW Pharmaceuticals plc has developed an oromucosal spray called Sativex®, which is constituted by an equimolecular combination of Δ9-tetrahydrocannabinol- and cannabidiol- enriched botanical extracts. Sativex® has been approved for the treatment of specific symptoms (i.e. spasticity and pain) of multiple sclerosis patients in various countries (i.e. Canada, UK, Spain, New Zealand). However, this cannabis- based medicine has been also proposed to be useful in other neurological disorders given the analgesic, antitumoral, anti-inflammatory, and neuroprotective properties of their components demonstrated in preclinical models. Numerous clinical trials are presently being conducted to confirm this potential in patients. We are particularly interested in the case of Huntington's disease (HD), an autosomal-dominant inherited disorder caused by an excess of CAG repeats in the genomic allele resulting in a polyQ expansion in the encoded protein called huntingtin, and that affects primarily striatal and cortical neurons thus producing motor abnormalities (i.e. chorea) and dementia. Cannabinoids have been studied for alleviation of hyperkinetic symptoms, given their inhibitory effects on movement, and, in particular, as disease-modifying agents due to their anti-inflammatory, neuroprotective and neuroregenerative properties. This potential has been corroborated in different experimental models of HD and using different types of cannabinoid agonists, including the phytocannabinoids present in Sativex®, and we are close to initiate a clinical trial with this cannabis-based medicine to evaluate its capability as a disease-modifying agent in a population of HD patients. The present review will address all preclinical evidence supporting the potential of Sativex® for the treatment of disease progression in HD patients. The article presents some promising patents on the cannabinoids.


Assuntos
Canabinoides/metabolismo , Canabinoides/uso terapêutico , Doença de Huntington/tratamento farmacológico , Doença de Huntington/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Animais , Descoberta de Drogas/métodos , Descoberta de Drogas/tendências , Humanos , Doença de Huntington/genética , Modelos Biológicos
19.
Pediatr Res ; 62(3): 255-60, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17622949

RESUMO

Neonatal hypoxic-ischemic encephalopathy (NHIE) is a devastating condition for which effective therapeutic treatments are still unavailable. Cannabinoids emerge as neuroprotective substances in adult animal studies; therefore, we aimed herein to test whether cannabinoids might reduce brain damage induced by hypoxiaischemia (HI) in newborn rats. Thus, 7-d-old Wistar rats (P7) were exposed to 8% O2 for 120 min after left carotid artery ligature, then received s.c. vehicle (VEH) (HI+VEH), the cannabinoid agonist WIN55212 (WIN) (0.1 mg/kg), or WIN with the CB1 or CB2 receptor antagonist SR141617 (SR1) (3 mg/kg) or SR141588 (SR2) (2 mg/kg). Brain damage was assessed by magnetic resonance imaging (MRI) at 1, 3, and 7 d after the insult. At the end of the experiment, MRI findings were corroborated by histology (Nissl staining). HI+VEH showed an area of cytotoxic and vasogenic edema at 24 h after the insult, then evolving to necrosis. HI+WIN showed a similar damaged area at 24 h after the insult, but the final necrotic area was reduced by 66%. Coadministration of either SR1 or SR2 reversed the effects of WIN. In conclusion, likely by activating CB1 and CB2 receptors, WIN afforded robust neuroprotection in newborn rats after HI.


Assuntos
Analgésicos/farmacologia , Benzoxazinas/farmacologia , Encéfalo , Canabinoides/agonistas , Hipóxia-Isquemia Encefálica/patologia , Morfolinas/farmacologia , Naftalenos/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Canabinoides/metabolismo , Imageamento por Ressonância Magnética , Distribuição Aleatória , Ratos , Ratos Wistar
20.
Synapse ; 53(4): 208-13, 2004 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-15266552

RESUMO

Two types of cannabinoid receptors have been characterized so far, CB1 and CB2. While CB1 receptors are present both in the CNS and in the periphery, CB2 receptors showed an almost exclusive distribution within the immune system. We now report that CB2 receptors are present in a specific microglial cell type of the human cerebellum. Thus, we have performed immunohistochemical analysis of tissue sections of white matter areas of the human cerebellum and detected the presence of CB2 receptors in perivascular microglial cells. These findings match with the well-known immunomodulatory role of CB2 receptors and open new perspectives on the possible role that these receptors may play in pathophysiological events.


Assuntos
Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Cerebelo/citologia , Cerebelo/metabolismo , Microglia/citologia , Microglia/metabolismo , Receptor CB2 de Canabinoide/biossíntese , Adulto , Biomarcadores , Western Blotting , Moduladores de Receptores de Canabinoides/metabolismo , Cerebelo/química , Feminino , Imunofluorescência , Antígenos HLA-DP/biossíntese , Antígenos HLA-DQ/biossíntese , Antígenos HLA-DR/biossíntese , Humanos , Imuno-Histoquímica , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA