Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Neurosci ; 44(23)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38719448

ABSTRACT

Physical exercise is a robust lifestyle intervention known for its enhancement of cognitive abilities. Nevertheless, the extent to which these benefits can be transmitted across generations (intergenerational inheritance to F1, and transgenerational to F2 and beyond) remains a topic of limited comprehension. We have already shown that cognitive improvements resulting from physical exercise can be inherited from parents to their offspring, proving intergenerational effects. So, we set out to explore whether these enhancements might extend transgenerationally, impacting the F2 generation. In this study, we initially examined the behavioral traits of second generation (F2) male mice, whose grandfathers (F0) had an exercise intervention. Our findings revealed that F2 mice with physically active grandpaternal F0 progenitors displayed significantly improved memory recall, encompassing both spatial and non-spatial information when compared to their counterparts from sedentary F0 progenitors, and proving for the first time the transgenerational inheritance of physical exercise induced cognitive enhancement. Surprisingly, while F2 memory improved (as was the case with F1), adult hippocampal neurogenesis remained unchanged between experimental and control groups (unlike in F1). Additionally, our analysis of small RNA sequences in the hippocampus identified 35 differentially expressed miRNAs linked to important brain function categories. Notably, two of these miRNAs, miRNA-144 and miRNA-298, displayed a robust negative correlation with cognitive performance. These findings highlight the enduring transgenerational transmission of cognitive benefits associated with exercise, even after two generations, suggesting that moderate exercise training can have lasting positive effects, possibly orchestrated by a specific set of miRNAs that exert their influence across multiple generations.


Subject(s)
Cognition , Hippocampus , Physical Conditioning, Animal , Animals , Male , Mice , Cognition/physiology , Physical Conditioning, Animal/physiology , Hippocampus/physiology , Hippocampus/metabolism , Female , Neurogenesis/physiology , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics
2.
J Cereb Blood Flow Metab ; : 271678X231217669, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-38017004

ABSTRACT

Increased neurotrophic support, including insulin-like growth factor I (IGF-I), is an important aspect of the adaptive response to ischemic insult. However, recent findings indicate that the IGF-I receptor (IGF-IR) in neurons plays a detrimental role in the response to stroke. Thus, we investigated the role of astrocytic IGF-IR on ischemic insults using tamoxifen-regulated Cre deletion of IGF-IR in glial fibrillary acidic protein (GFAP) astrocytes, a major cellular component in the response to injury. Ablation of IGF-IR in astrocytes (GFAP-IGF-IR KO mice) resulted in larger ischemic lesions, greater blood-brain-barrier disruption and more deteriorated sensorimotor coordination. RNAseq detected increases in inflammatory, cell adhesion and angiogenic pathways, while the expression of various classical biomarkers of response to ischemic lesion were significantly increased at the lesion site compared to control littermates. While serum IGF-I levels after injury were decreased in both control and GFAP-IR KO mice, brain IGF-I mRNA expression show larger increases in the latter. Further, greater damage was also accompanied by altered glial reactivity as reflected by changes in the morphology of GFAP astrocytes, and relative abundance of ionized calcium binding adaptor molecule 1 (Iba 1) microglia. These results suggest a protective role for astrocytic IGF-IR in the response to ischemic injury.

3.
Front Neuroanat ; 16: 910477, 2022.
Article in English | MEDLINE | ID: mdl-35958733

ABSTRACT

The propensity to develop neurodegenerative diseases is influenced by diverse factors including genetic background, sex, lifestyle, including dietary habits and being overweight, and age. Indeed, with aging, there is an increased incidence of obesity and neurodegenerative processes, both of which are associated with inflammatory responses, in a sex-specific manner. High fat diet (HFD) commonly leads to obesity and markedly affects metabolism, both peripherally and centrally. Here we analyzed the metabolic and inflammatory responses of middle-aged (11-12 months old) transgenic amyloid precursor protein (TgAPP) mice of both sexes to HFD for 18 weeks (starting at 7-8 months of age). We found clear sex differences with females gaining significantly more weight and fat mass than males, with a larger increase in circulating leptin levels and expression of inflammatory markers in visceral adipose tissue. Glycemia and insulin levels increased in HFD fed mice of both sexes, with TgAPP mice being more affected than wild type (WT) mice. In the hypothalamus, murine amyloid ß (Aß) levels were increased by HFD intake exclusively in males, reaching statistical significance in TgAPP males. On a low fat diet (LFD), TgAPP males had significantly lower mRNA levels of the anorexigenic neuropeptide proopiomelanocortin (POMC) than WT males, with HFD intake decreasing the expression of the orexigenic neuropeptides Agouti-related peptide (AgRP) and neuropeptide Y (NPY), especially in TgAPP mice. In females, HFD increased POMC mRNA levels but had no effect on AgRP or NPY mRNA levels, and with no effect on genotype. There was no effect of diet or genotype on the hypothalamic inflammatory markers analyzed or the astrogliosis marker glial acidic protein (GFAP); however, levels of the microglial marker Iba-1 increased selectively in male TgAPP mice. In summary, the response to HFD intake was significantly affected by sex, with fewer effects due to genotype. Hypothalamic inflammatory cytokine expression and astrogliosis were little affected by HFD in middle-aged mice, although in TgAPP males, which showed increased Aß, there was microglial activation. Thus, excess intake of diets high in fat should be avoided because of its possible detrimental consequences.

4.
Proc Natl Acad Sci U S A ; 119(29): e2204527119, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35858325

ABSTRACT

Mice with insulin receptor (IR)-deficient astrocytes (GFAP-IR knockout [KO] mice) show blunted responses to insulin and reduced brain glucose uptake, whereas IR-deficient astrocytes show disturbed mitochondrial responses to glucose. While exploring the functional impact of disturbed mitochondrial function in astrocytes, we observed that GFAP-IR KO mice show uncoupling of brain blood flow with glucose uptake. Since IR-deficient astrocytes show higher levels of reactive oxidant species (ROS), this leads to stimulation of hypoxia-inducible factor-1α and, consequently, of the vascular endothelial growth factor angiogenic pathway. Indeed, GFAP-IR KO mice show disturbed brain vascularity and blood flow that is normalized by treatment with the antioxidant N-acetylcysteine (NAC). NAC ameliorated high ROS levels, normalized angiogenic signaling and mitochondrial function in IR-deficient astrocytes, and normalized neurovascular coupling in GFAP-IR KO mice. Our results indicate that by modulating glucose uptake and angiogenesis, insulin receptors in astrocytes participate in neurovascular coupling.


Subject(s)
Astrocytes , Brain , Insulin , Neovascularization, Physiologic , Neurovascular Coupling , Animals , Astrocytes/metabolism , Brain/blood supply , Glial Fibrillary Acidic Protein/genetics , Glucose/metabolism , Insulin/metabolism , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , Receptor, Insulin/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
5.
Neurobiol Dis ; 159: 105495, 2021 11.
Article in English | MEDLINE | ID: mdl-34478848

ABSTRACT

Long-term high-fat diet (HFD) consumption commonly leads to obesity, a major health concern of western societies and a risk factor for Alzheimer's disease (AD). Both conditions present glial activation and inflammation and show sex differences in their incidence, clinical manifestation, and disease course. HFD intake has an important impact on gut microbiota, the bacteria present in the gut, and microbiota dysbiosis is associated with inflammation and certain mental disorders such as anxiety. In this study, we have analyzed the effects of a prolonged (18 weeks, starting at 7 months of age) HFD on male and female mice, both wild type (WT) and TgAPP mice, a model for AD, investigating the behavioral profile, gut microbiota composition and inflammatory/phagocytosis-related gene expression in hippocampus. In the open-field test, no overt differences in motor activity were observed between male and female or WT and TgAPP mice on a low-fat diet (LFD). However, HFD induced anxiety, as judged by decreased motor activity and increased time in the margins in the open-field, and a trend towards increased immobility time in the tail suspension test, with increased defecation. Intriguingly, female TgAPP mice on HFD showed less immobility and defecation compared to female WT mice on HFD. HFD induced dysbiosis of gut microbiota, resulting in reduced microbiota diversity and abundance compared with LFD fed mice, with some significant differences due to sex and little effect of genotype. Gene expression of pro-inflammatory/phagocytic markers in the hippocampus were not different between male and female WT mice, and in TgAPP mice of both sexes, some cytokines (IL-6 and IFNγ) were higher than in WT mice on LFD, more so in female TgAPP (IL-6). HFD induced few alterations in mRNA expression of inflammatory/phagocytosis-related genes in male mice, whether WT (IL-1ß, MHCII), or TgAPP (IL-6). However, in female TgAPP, altered gene expression returned towards control levels following prolonged HFD (IL-6, IL-12ß, TNFα, CD36, IRAK4, PYRY6). In summary, we demonstrate that HFD induces anxiogenic symptoms, marked alterations in gut microbiota, and increased expression of inflammatory genes, except for female TgAPP that appear to be resistant to the diet effects. Lifestyle interventions should be introduced to prevent AD onset or exacerbation by reducing inflammation and its associated symptoms; however, our results suggest that the eventual goal of developing prevention and treatment strategies should take sex into consideration.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Behavior, Animal/physiology , Diet, High-Fat , Dysbiosis/genetics , Gastrointestinal Microbiome/physiology , Inflammation/genetics , Stress, Psychological/genetics , Alzheimer Disease/physiopathology , Animals , Disease Models, Animal , Dysbiosis/physiopathology , Female , Histocompatibility Antigens Class II/genetics , Humans , Interleukin-1beta/genetics , Interleukin-6/genetics , Male , Mice , Mice, Transgenic , Phagocytosis/genetics , RNA, Messenger/metabolism , Sex Characteristics , Stress, Psychological/physiopathology
6.
Mech Ageing Dev ; 195: 111462, 2021 04.
Article in English | MEDLINE | ID: mdl-33609535

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia and has a higher incidence in women. The main component of the senile plaques characteristic of AD is amyloid-beta (Aß), with surrounding astrocytes contributing to the degenerative process. We hypothesized that the sex difference in the incidence of AD could be partially due to differential astrocytic responses to Aß. Thus, the effect of Aß1-40 on cell viability, the inflammatory response, and oxidative status was studied in cultures of hippocampal astrocytes from male and female rats. Aß1-40 increased astrocyte viability in both female and male cultures by activating proliferation and survival pathways. Pro-inflammatory and anti-inflammatory responses were induced in astrocytes from both sexes. Aß1-40 did not affect endoplasmic reticulum stress although it induced oxidative stress in male and female astrocytes. Interestingly, male astrocytes had an increase in cell number and significantly lower cell death in response to Aß1-40. Conversely, astrocytes from females displayed a greater inflammatory response after the Aß1-40 challenge. These results suggest that the inflammatory and oxidative environment induced by Aß1-40 in female astrocytes may contribute to enhance the vulnerability to AD and warrants further studies to unveil the mechanisms underlying sex differences in astrocytic responses.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes , Neuroimmunomodulation/physiology , Peptide Fragments/metabolism , Animals , Astrocytes/immunology , Astrocytes/metabolism , Cell Proliferation , Cell Survival/immunology , Cells, Cultured , Female , Hippocampus/immunology , Hippocampus/metabolism , Male , Oxidative Stress , Rats , Sex Characteristics , Sex Factors
7.
Eur J Med Chem ; 166: 90-107, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30685536

ABSTRACT

Multitarget cannabinoids could be a promising therapeutic strategic to fight against Alzheimer's disease. In this sense, our group has developed a new family of indazolylketones with multitarget profile including cannabinoids, cholinesterase and BACE-1 activity. A medicinal chemistry program that includes computational design, synthesis and in vitro and cellular evaluation has allowed to us to achieve lead compounds. In this work, the synthesis and evaluation of a new class of indazolylketones have been performed. Pharmacological evaluation includes functional activity for cannabinoid receptors on isolated tissue. In addition, in vitro inhibitory assays in AChE/BuChE enzymes and BACE-1 have been carried out. Furthermore, studies of neuroprotective effects in human neuroblastoma SH-SY5Y cells and studies of the mechanisms of survival/death in lymphoblasts of patients with Alzheimer's disease have been achieved. The results of pharmacological tests have revealed that some of these derivatives (5, 6) behave as CB2 cannabinoid agonists and simultaneously show BuChE and/or BACE-1 inhibition.


Subject(s)
Cannabinoids/chemistry , Cannabinoids/pharmacology , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/pharmacology , Indazoles/chemistry , Ketones/chemistry , Ketones/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Butyrylcholinesterase/metabolism , Cannabinoids/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Cholinesterase Inhibitors/chemical synthesis , Drug Design , Humans , Ketones/chemical synthesis , Neurons/cytology , Neurons/drug effects , Receptor, Cannabinoid, CB2/antagonists & inhibitors
8.
Glia ; 66(3): 522-537, 2018 03.
Article in English | MEDLINE | ID: mdl-29139169

ABSTRACT

Sex differences in the incidence, clinical manifestation, disease course, and prognosis of neurological diseases, such as autism spectrum disorders or Alzheimer's disease, have been reported. Obesity has been postulated as a risk factor for cognitive decline and Alzheimer's disease and, during pregnancy, increases the risk of autism spectrum disorders in the offspring. Obesity is associated with increased serum and brain levels of free fatty acids, such as palmitic acid, which activate microglial cells triggering a potent inflammatory cascade. In this study, we have determined the effect of palmitic acid in the inflammatory profile, motility, and phagocytosis of primary male and female microglia, both in basal conditions and in the presence of a pro-inflammatory stimulus (interferon-γ). Male microglia in vitro showed higher migration than female microglia under basal and stimulated conditions. In contrast, female microglia had higher basal and stimulated phagocytic activity than male microglia. Palmitic acid did not affect basal migration or phagocytosis, but abolished the migration and phagocytic activity of male and female microglia in response to interferon-γ. These findings extend previous observations of sex differences in microglia and suggest that palmitic acid impairs the protective responses of these cells.


Subject(s)
Cell Movement/physiology , Microglia/metabolism , Palmitic Acid/toxicity , Phagocytosis/physiology , Sex Characteristics , Animals , Cell Movement/drug effects , Cells, Cultured , Female , Inflammation/metabolism , Inflammation/pathology , Interferon-gamma/administration & dosage , Interferon-gamma/metabolism , Male , Microglia/drug effects , Microglia/pathology , Phagocytosis/drug effects , Prosencephalon/drug effects , Prosencephalon/metabolism , Prosencephalon/pathology , RNA, Messenger/metabolism , Rats, Wistar
10.
JMIR Res Protoc ; 6(1): e8, 2017 Jan 24.
Article in English | MEDLINE | ID: mdl-28119279

ABSTRACT

BACKGROUND: Neuroplasticity-based approaches seem to offer promising ways of maintaining cognitive health in older adults and postponing the onset of cognitive decline symptoms. Although previous research suggests that training can produce transfer effects, this study was designed to overcome some limitations of previous studies by incorporating an active control group and the assessment of training expectations. OBJECTIVE: The main objectives of this study are (1) to evaluate the effects of a randomized computer-based intervention consisting of training older adults with nonaction video games on brain and cognitive functions that decline with age, including attention and spatial working memory, using behavioral measures and electrophysiological recordings (event-related potentials [ERPs]) just after training and after a 6-month no-contact period; (2) to explore whether motivation, engagement, or expectations might account for possible training-related improvements; and (3) to examine whether inflammatory mechanisms assessed with noninvasive measurement of C-reactive protein in saliva impair cognitive training-induced effects. A better understanding of these mechanisms could elucidate pathways that could be targeted in the future by either behavioral or neuropsychological interventions. METHODS: A single-blinded randomized controlled trial with an experimental group and an active control group, pretest, posttest, and 6-month follow-up repeated measures design is used in this study. A total of 75 cognitively healthy older adults were randomly distributed into experimental and active control groups. Participants in the experimental group received 16 1-hour training sessions with cognitive nonaction video games selected from Lumosity, a commercial brain training package. The active control group received the same number of training sessions with The Sims and SimCity, a simulation strategy game. RESULTS: We have recruited participants, have conducted the training protocol and pretest assessments, and are currently conducting posttest evaluations. The study will conclude in the first semester of 2017. Data analysis will take place during 2017. The primary outcome is transfer of benefit from training to attention and working memory functions and the neural mechanisms underlying possible cognitive improvements. CONCLUSIONS: We expect that mental stimulation with video games will improve attention and memory both at the behavioral level and in ERP components promoting brain and mental health and extending independence among elderly people by avoiding the negative personal and economic consequences of long-term care. TRIAL REGISTRATION: Clinicaltrials.gov NCT02796508; https://clinicaltrials.gov/ct2/show/NCT02796508 (archived by WebCite at http://www.webcitation.org/6nFeKeFNB).

11.
Neuropharmacology ; 116: 174-187, 2017 04.
Article in English | MEDLINE | ID: mdl-28012947

ABSTRACT

Glycogen synthase kinase 3 (GSK-3) is a constitutively active kinase that has been implicated in the mechanism of action of mood stabilizers. According to the neurogenic hypothesis of depression, newborn neurons in the adult dentate gyrus are required for the antidepressant effects of certain agents. We demonstrate that administration of the GSK-3 inhibitor VP2.51 (2.5 mg/kg ip, for 3.5 weeks) increases cell proliferation (pH3+ cells), as well as the short- and long-term survival of newborn neurons (assessed by the 24 h survival of BrdU+ and DCX+ neurons), while significantly increasing the commitment of cells to the granule neuron lineage (Prox1 immunoreactivity). In parallel, VP2.51 induces a net antidepressant effect, as judged by the decrease in the immobility time in the forced swim test of naïve mice (non-stressed mice), as well as a therapeutic effect on previously stressed mice (Porsolt-induced stress). Interestingly, the morphological changes were found prominently in the ventral region of the hippocampus. We found that these effects are neurogenesis dependent by combining the antimitotic temozolomide (50 mg/kg ip) with the drug. Importantly VP2.51 did not provoke changes in weight or in a battery of behavioral tests (learning/memory and activity tests). As the effects of VP2.51 were concomitant with the increase in ß-catenin expression and a shift towards the inactive form of GSK-3, we suggest that VP2.51 has therapeutic benefits following stress, and it may be a preventive treatment in situations where a potential depressive state and/or loss of memory is associated with diminished neurogenesis, through selective GSK3-beta inhibition.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hippocampus/drug effects , Neurogenesis/drug effects , Animals , Antimitotic Agents/pharmacology , Avoidance Learning/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Depressive Disorder/enzymology , Depressive Disorder/pathology , Disease Models, Animal , Doublecortin Protein , Glycogen Synthase Kinase 3/metabolism , Hippocampus/enzymology , Hippocampus/pathology , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Mitosis/drug effects , Mitosis/physiology , Motor Activity/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Neurogenesis/physiology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Random Allocation , Stress, Psychological/drug therapy , Stress, Psychological/enzymology , Stress, Psychological/pathology , Temozolomide , beta Catenin/metabolism
12.
Front Neurosci ; 10: 422, 2016.
Article in English | MEDLINE | ID: mdl-27695396

ABSTRACT

There is evidence of altered vascular function, including cerebrovascular, in Alzheimer's disease (AD) and transgenic models of the disease. Indeed vasoconstrictor responses are increased, while vasodilation is reduced in both conditions. ß-Amyloid (Aß) appears to be responsible, at least in part, of alterations in vascular function. Cannabinoids, neuroprotective and anti-inflammatory agents, induce vasodilation both in vivo and in vitro. We have demonstrated a beneficial effect of cannabinoids in models of AD by preventing glial activation. In this work we have studied the effects of these compounds on vessel density in amyloid precursor protein (APP) transgenic mice, line 2576, and on altered vascular responses in aortae isolated ring. First we showed increased collagen IV positive vessels in AD brain compared to control subjects, with a similar increase in TgAPP mice, which was normalized by prolonged oral treatment with the CB1/CB2 mixed agonist WIN 55,212-2 (WIN) and the CB2 selective agonist JWH-133 (JWH). In Tg APP mice the vasoconstriction induced by phenylephrine and the thromboxane agonist U46619 was significantly increased, and no change in the vasodilation to acetylcholine (ACh) was observed. Tg APP displayed decreased vasodilation to both cannabinoid agonists, which were able to prevent decreased ACh relaxation in the presence of Aß. In summary, we have confirmed and extended the existence of altered vascular responses in Tg APP mice. Moreover, our results suggest that treatment with cannabinoids may ameliorate the vascular responses in AD-type pathology.

13.
Autophagy ; 12(10): 1902-1916, 2016 10 02.
Article in English | MEDLINE | ID: mdl-27427974

ABSTRACT

Autophagy is a highly coordinated process that is controlled at several levels including transcriptional regulation. Here, we identify the transcription factor NFE2L2/NRF2 (nuclear factor, erythroid 2 like 2) as a regulator of autophagy gene expression and its relevance in a mouse model of Alzheimer disease (AD) that reproduces impaired APP (amyloid ß precursor protein) and human (Hs)MAPT/TAU processing, clearance and aggregation. We screened the chromatin immunoprecipitation database ENCODE for 2 proteins, MAFK and BACH1, that bind the NFE2L2-regulated enhancer antioxidant response element (ARE). Using a script generated from the JASPAR's consensus ARE sequence, we identified 27 putative AREs in 16 autophagy-related genes. Twelve of these sequences were validated as NFE2L2 regulated AREs in 9 autophagy genes by additional ChIP assays and quantitative RT-PCR on human and mouse cells after NFE2L2 activation with sulforaphane. Mouse embryo fibroblasts of nfe2l2-knockout mice exhibited reduced expression of autophagy genes, which was rescued by an NFE2L2 expressing lentivirus, and impaired autophagy flux when exposed to hydrogen peroxide. NFE2L2-deficient mice co-expressing HsAPPV717I and HsMAPTP301L, exhibited more intracellular aggregates of these proteins and reduced neuronal levels of SQSTM1/p62, CALCOCO2/NDP52, ULK1, ATG5 and GABARAPL1. Also, colocalization of HsAPPV717I and HsMAPTP301L with the NFE2L2-regulated autophagy marker SQSTM1/p62 was reduced in the absence of NFE2L2. In AD patients, neurons expressing high levels of APP or MAPT also expressed SQSTM1/p62 and nuclear NFE2L2, suggesting their attempt to degrade intraneuronal aggregates through autophagy. This study shows that NFE2L2 modulates autophagy gene expression and suggests a new strategy to combat proteinopathies.


Subject(s)
Autophagy/genetics , Gene Expression Regulation , NF-E2-Related Factor 2/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Antioxidant Response Elements/genetics , Disease Models, Animal , HEK293 Cells , Humans , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Middle Aged , NF-E2-Related Factor 2/deficiency , Neurons/metabolism , Neurons/pathology , Promoter Regions, Genetic/genetics
14.
Neuropharmacology ; 110(Pt A): 519-529, 2016 11.
Article in English | MEDLINE | ID: mdl-26976670

ABSTRACT

Cannabinoid CB2 receptors (CB2Rs) are emerging as important therapeutic targets in brain disorders that typically involve neurometabolic alterations. We here addressed the possible role of CB2Rs in the regulation of glucose uptake in the mouse brain. To that aim, we have undertaken 1) measurement of (3)H-deoxyglucose uptake in cultured cortical astrocytes and neurons and in acute hippocampal slices; 2) real-time visualization of fluorescently labeled deoxyglucose uptake in superfused hippocampal slices; and 3) in vivo PET imaging of cerebral (18)F-fluorodeoxyglucose uptake. We now show that both selective (JWH133 and GP1a) as well as non-selective (WIN55212-2) CB2R agonists, but not the CB1R-selective agonist, ACEA, stimulate glucose uptake, in a manner that is sensitive to the CB2R-selective antagonist, AM630. Glucose uptake is stimulated in astrocytes and neurons in culture, in acute hippocampal slices, in different brain areas of young adult male C57Bl/6j and CD-1 mice, as well as in middle-aged C57Bl/6j mice. Among the endocannabinoid metabolizing enzymes, the selective inhibition of COX-2, rather than that of FAAH, MAGL or α,ßDH6/12, also stimulates the uptake of glucose in hippocampal slices of middle-aged mice, an effect that was again prevented by AM630. However, we found the levels of the endocannabinoid, anandamide reduced in the hippocampus of TgAPP-2576 mice (a model of ß-amyloidosis), and likely as a consequence, COX-2 inhibition failed to stimulate glucose uptake in these mice. Together, these results reveal a novel general glucoregulatory role for CB2Rs in the brain, raising therapeutic interest in CB2R agonists as nootropic agents.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Glucose/metabolism , Receptor, Cannabinoid, CB2/metabolism , Aging/drug effects , Aging/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor , Amyloidosis/diagnostic imaging , Amyloidosis/drug therapy , Amyloidosis/metabolism , Animals , Arachidonic Acids/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Brain/diagnostic imaging , Brain/drug effects , Cannabinoid Receptor Modulators/pharmacology , Cells, Cultured , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Endocannabinoids/metabolism , Hydroxyethylrutoside , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Nootropic Agents/pharmacology , Polyunsaturated Alkamides/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Tissue Culture Techniques
15.
Eur J Med Chem ; 101: 604-15, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26204508

ABSTRACT

The synthesis of the new radiotracer precursor 4-Br-NITTP and the radiolabeling of the new tracer 1-(4-bromo-2-nitroimidazol-1-yl)-3-[(18)F]fluoropropan-2-ol (4-Br-[(18)F]FMISO) is reported. The cyclic voltammetry behaviour, neuronal cell toxicity, transport through the brain endothelial cell monolayer, in vivo PET imaging and preliminary calculations of the tracer uptake for a rodent model of stroke were studied for the new compound and the results were compared to those obtained with [(18)F]FMISO, the current gold standard PET hypoxia tracer. The new PET brain hypoxia tracer is more easily reduced, has higher CLogP than [(18)F]FMISO and it diffuses more rapidly through brain endothelial cells. The new compound is non-toxic to neuronal cells and it allows the in vivo mapping of stroke in mice with higher sensitivity. 4-Br-[(18)F]FMISO is a good candidate for further development in ischemic stroke.


Subject(s)
Disease Models, Animal , Hypoxia, Brain/diagnostic imaging , Nitroimidazoles/pharmacokinetics , Positron-Emission Tomography/methods , Propanols/pharmacokinetics , Stroke/diagnostic imaging , Animals , Cell Line , Male , Mice , Molecular Structure , Nitroimidazoles/chemical synthesis , Nitroimidazoles/chemistry , Propanols/chemical synthesis , Propanols/chemistry , Rats , Rats, Inbred F344
16.
J Clin Invest ; 122(11): 3900-13, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064363

ABSTRACT

Glial cells perform critical functions that alter the metabolism and activity of neurons, and there is increasing interest in their role in appetite and energy balance. Leptin, a key regulator of appetite and metabolism, has previously been reported to influence glial structural proteins and morphology. Here, we demonstrate that metabolic status and leptin also modify astrocyte-specific glutamate and glucose transporters, indicating that metabolic signals influence synaptic efficacy and glucose uptake and, ultimately, neuronal function. We found that basal and glucose-stimulated electrical activity of hypothalamic proopiomelanocortin (POMC) neurons in mice were altered in the offspring of mothers fed a high-fat diet. In adulthood, increased body weight and fasting also altered the expression of glucose and glutamate transporters. These results demonstrate that whole-organism metabolism alters hypothalamic glial cell activity and suggest that these cells play an important role in the pathology of obesity.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Astrocytes/metabolism , Dietary Fats/adverse effects , Glucose Transport Proteins, Facilitative/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Obesity/metabolism , Animals , Astrocytes/pathology , Dietary Fats/pharmacology , Hypothalamus/pathology , Mice , Neurons/metabolism , Neurons/pathology , Obesity/chemically induced , Obesity/pathology , Pro-Opiomelanocortin/metabolism , Rats , Rats, Wistar
17.
J Neuroinflammation ; 9: 8, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22248049

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) brain shows an ongoing inflammatory condition and non-steroidal anti-inflammatories diminish the risk of suffering the neurologic disease. Cannabinoids are neuroprotective and anti-inflammatory agents with therapeutic potential. METHODS: We have studied the effects of prolonged oral administration of transgenic amyloid precursor protein (APP) mice with two pharmacologically different cannabinoids (WIN 55,212-2 and JWH-133, 0.2 mg/kg/day in the drinking water during 4 months) on inflammatory and cognitive parameters, and on ¹8F-fluoro-deoxyglucose (¹8FDG) uptake by positron emission tomography (PET). RESULTS: Novel object recognition was significantly reduced in 11 month old Tg APP mice and 4 month administration of JWH was able to normalize this cognitive deficit, although WIN was ineffective. Wild type mice cognitive performance was unaltered by cannabinoid administration. Tg APP mice showed decreased ¹8FDG uptake in hippocampus and cortical regions, which was counteracted by oral JWH treatment. Hippocampal GFAP immunoreactivity and cortical protein expression was unaffected by genotype or treatment. In contrast, the density of Iba1 positive microglia was increased in Tg APP mice, and normalized following JWH chronic treatment. Both cannabinoids were effective at reducing the enhancement of COX-2 protein levels and TNF-α mRNA expression found in the AD model. Increased cortical ß-amyloid (Aß) levels were significantly reduced in the mouse model by both cannabinoids. Noteworthy both cannabinoids enhanced Aß transport across choroid plexus cells in vitro. CONCLUSIONS: In summary we have shown that chronically administered cannabinoid showed marked beneficial effects concomitant with inflammation reduction and increased Aß clearance.


Subject(s)
Amyloid beta-Peptides/metabolism , Cannabinoids/administration & dosage , Cognition Disorders/prevention & control , Encephalitis/prevention & control , Administration, Oral , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Benzoxazines/administration & dosage , Choroid Plexus/metabolism , Choroid Plexus/pathology , Cognition Disorders/diagnostic imaging , Cognition Disorders/etiology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Encephalitis/diagnostic imaging , Encephalitis/etiology , Enzyme-Linked Immunosorbent Assay , Fluorodeoxyglucose F18/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Morpholines/administration & dosage , Naphthalenes/administration & dosage , Positron-Emission Tomography , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB2/metabolism , Time Factors
18.
J Neurosci Methods ; 204(2): 366-73, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22155442

ABSTRACT

Several techniques exist to study the rate of glucose uptake and metabolism in the brain but most of them are not sufficiently robust to permit extensive pharmacological analysis. Here we optimized an in vitro measurement of the simultaneous accumulation of the metabolizable and non-metabolizable (3)H and (14)C d-glucose analogues; permitting convenient large-scale studies on glucose uptake and metabolism in brain slices. Next, we performed an extensive pharmacological characterization on the putative glucoregulator role of the endocannabinoid system in the hippocampal slices of the rat, and the wild-type and the CB(1) cannabinoid receptor (CB(1)R) knockout mice. We observed that (3)H-3-O-methylglucose is a poor substrate to measure glucose uptake in the hippocampus. (3)H-2-deoxyglucose is a better substrate but its uptake is still lower than that of (14)C-U-d-glucose, from which the slices constantly metabolize and dissipate (14)C atoms. Thus, uptake and the metabolism values are not to be used as standalones but as differences between a control and a treatment. The CB(1)R knockout mice exhibited ∼10% less glucose uptake and glucose carbon atom dissipation in comparison with the wild-type mice. This may represent congenital defects as acute treatments of the rat and mouse slices with cannabinoid agonists, antagonists and inhibitors of endocannabinoid uptake/metabolism failed to induce robust changes in either the uptake or the metabolism of glucose. In summary, we report here an optimized technique ideal to complement other metabolic approaches of high spatiotemporal resolution. This technique allowed us concluding that CB(1)Rs are at least indirectly involved in hippocampal glucoregulation.


Subject(s)
Glucose/metabolism , Hippocampus/metabolism , Receptor, Cannabinoid, CB1/deficiency , 3-O-Methylglucose/metabolism , Analysis of Variance , Animals , Cannabinoids/agonists , Cannabinoids/antagonists & inhibitors , Cannabinoids/pharmacology , Deoxyglucose/metabolism , Dose-Response Relationship, Drug , Glucose/analogs & derivatives , Hippocampus/drug effects , In Vitro Techniques , Isotopes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neocortex/drug effects , Neocortex/metabolism , Potassium/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/genetics
19.
Brain ; 134(Pt 4): 1041-60, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21459826

ABSTRACT

Retrograde messengers adjust the precise timing of neurotransmitter release from the presynapse, thus modulating synaptic efficacy and neuronal activity. 2-Arachidonoyl glycerol, an endocannabinoid, is one such messenger produced in the postsynapse that inhibits neurotransmitter release upon activating presynaptic CB(1) cannabinoid receptors. Cognitive decline in Alzheimer's disease is due to synaptic failure in hippocampal neuronal networks. We hypothesized that errant retrograde 2-arachidonoyl glycerol signalling impairs synaptic neurotransmission in Alzheimer's disease. Comparative protein profiling and quantitative morphometry showed that overall CB(1) cannabinoid receptor protein levels in the hippocampi of patients with Alzheimer's disease remain unchanged relative to age-matched controls, and CB(1) cannabinoid receptor-positive presynapses engulf amyloid-ß-containing senile plaques. Hippocampal protein concentrations for the sn-1-diacylglycerol lipase α and ß isoforms, synthesizing 2-arachidonoyl glycerol, significantly increased in definite Alzheimer's (Braak stage VI), with ectopic sn-1-diacylglycerol lipase ß expression found in microglia accumulating near senile plaques and apposing CB(1) cannabinoid receptor-positive presynapses. We found that microglia, expressing two 2-arachidonoyl glycerol-degrading enzymes, serine hydrolase α/ß-hydrolase domain-containing 6 and monoacylglycerol lipase, begin to surround senile plaques in probable Alzheimer's disease (Braak stage III). However, Alzheimer's pathology differentially impacts serine hydrolase α/ß-hydrolase domain-containing 6 and monoacylglycerol lipase in hippocampal neurons: serine hydrolase α/ß-hydrolase domain-containing 6 expression ceases in neurofibrillary tangle-bearing pyramidal cells. In contrast, pyramidal cells containing hyperphosphorylated tau retain monoacylglycerol lipase expression, although at levels significantly lower than in neurons lacking neurofibrillary pathology. Here, monoacylglycerol lipase accumulates in CB(1) cannabinoid receptor-positive presynapses. Subcellular fractionation revealed impaired monoacylglycerol lipase recruitment to biological membranes in post-mortem Alzheimer's tissues, suggesting that disease progression slows the termination of 2-arachidonoyl glycerol signalling. We have experimentally confirmed that altered 2-arachidonoyl glycerol signalling could contribute to synapse silencing in Alzheimer's disease by demonstrating significantly prolonged depolarization-induced suppression of inhibition when superfusing mouse hippocampi with amyloid-ß. We propose that the temporal dynamics and cellular specificity of molecular rearrangements impairing 2-arachidonoyl glycerol availability and actions may differ from those of anandamide. Thus, enhanced endocannabinoid signalling, particularly around senile plaques, can exacerbate synaptic failure in Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Arachidonic Acids/metabolism , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Glycerides/metabolism , Hippocampus/metabolism , Neurons/metabolism , Alzheimer Disease/pathology , Animals , Blotting, Western , Electrophysiology , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Mice , Neurons/pathology , Receptors, Cannabinoid/metabolism , Signal Transduction/physiology , Synaptic Transmission/physiology , Wnt Proteins/metabolism , Wnt3 Protein
20.
Mol Pharmacol ; 79(6): 964-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21350020

ABSTRACT

Microglial activation is an invariant feature of Alzheimer's disease (AD). It is noteworthy that cannabinoids are neuroprotective by preventing ß-amyloid (Aß)-induced microglial activation both in vitro and in vivo. On the other hand, the phytocannabinoid cannabidiol (CBD) has shown anti-inflammatory properties in different paradigms. In the present study, we compared the effects of CBD with those of other cannabinoids on microglial cell functions in vitro and on learning behavior and cytokine expression after Aß intraventricular administration to mice. CBD, (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl) pyrrolo-[1,2,3-d,e]-1,4-benzoxazin-6-yl]-1-naphthalenyl-methanone [WIN 55,212-2 (WIN)], a mixed CB(1)/CB(2) agonist, and 1,1-dimethylbutyl-1-deoxy-Δ(9)-tetrahydrocannabinol [JWH-133 (JWH)], a CB(2)-selective agonist, concentration-dependently decreased ATP-induced (400 µM) increase in intracellular calcium ([Ca(2+)](i)) in cultured N13 microglial cells and in rat primary microglia. In contrast, 4-[4-(1,1-dimethylheptyl)-2,6-dimethoxyphenyl]-6,6-dimethyl-bicyclo[3.1.1]hept-2-ene-2-methanol [HU-308 (HU)], another CB(2) agonist, was without effect. Cannabinoid and adenosine A(2A) receptors may be involved in the CBD action. CBD- and WIN-promoted primary microglia migration was blocked by CB(1) and/or CB(2) antagonists. JWH and HU-induced migration was blocked by a CB(2) antagonist only. All of the cannabinoids decreased lipopolysaccharide-induced nitrite generation, which was insensitive to cannabinoid antagonism. Finally, both CBD and WIN, after subchronic administration for 3 weeks, were able to prevent learning of a spatial navigation task and cytokine gene expression in ß-amyloid-injected mice. In summary, CBD is able to modulate microglial cell function in vitro and induce beneficial effects in an in vivo model of AD. Given that CBD lacks psychoactivity, it may represent a novel therapeutic approach for this neurological disease.


Subject(s)
Alzheimer Disease/pathology , Cannabidiol/pharmacology , Cannabinoids/pharmacology , Microglia/drug effects , Animals , Base Sequence , Cells, Cultured , DNA Primers , Humans , Immunohistochemistry , In Vitro Techniques , Mice , Nitric Oxide/biosynthesis , Polymerase Chain Reaction , RNA, Messenger/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...