Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cereb Cortex ; 34(5)2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38802684

RESUMEN

The ε4 allele of the APOE gene heightens the risk of late onset Alzheimer's disease. ε4 carriers, may exhibit cognitive and neural changes early on. Given the known memory-enhancing effects of physical exercise, particularly through hippocampal plasticity via endocannabinoid signaling, here we aimed to test whether a single session of physical exercise may benefit memory and underlying neurophysiological processes in young ε3 carriers (ε3/ε4 heterozygotes, risk group) compared with a matched control group (homozygotes for ε3). Participants underwent fMRI while learning picture sequences, followed by cycling or rest before a memory test. Blood samples measured endocannabinoid levels. At the behavioral level, the risk group exhibited poorer associative memory performance, regardless of the exercising condition. At the brain level, the risk group showed increased medial temporal lobe activity during memory retrieval irrespective of exercise (suggesting neural compensatory effects even at baseline), whereas, in the control group, such increase was only detectable after physical exercise. Critically, an exercise-related endocannabinoid increase correlated with task-related hippocampal activation in the control group only. In conclusion, healthy young individuals carrying the ε4 allele may present suboptimal associative memory performance (when compared with homozygote ε3 carriers), together with reduced plasticity (and functional over-compensation) within medial temporal structures.


Asunto(s)
Enfermedad de Alzheimer , Ejercicio Físico , Imagen por Resonancia Magnética , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/diagnóstico por imagen , Masculino , Femenino , Ejercicio Físico/fisiología , Adulto , Adulto Joven , Memoria/fisiología , Endocannabinoides/genética , Predisposición Genética a la Enfermedad , Aprendizaje por Asociación/fisiología , Apolipoproteína E4/genética , Hipocampo/diagnóstico por imagen , Hipocampo/fisiología , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Heterocigoto
2.
Proc Natl Acad Sci U S A ; 119(37): e2122700119, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36067295

RESUMEN

Columnar structure is one of the most fundamental morphological features of the cerebral cortex and is thought to be the basis of information processing in higher animals. Yet, how such a topographically precise structure is formed is largely unknown. Formation of columnar projection of layer 4 (L4) axons is preceded by thalamocortical formation, in which type 1 cannabinoid receptors (CB1R) play an important role in shaping barrel-specific targeted projection by operating spike timing-dependent plasticity during development (Itami et al., J. Neurosci. 36, 7039-7054 [2016]; Kimura & Itami, J. Neurosci. 39, 3784-3791 [2019]). Right after the formation of thalamocortical projections, CB1Rs start to function at L4 axon terminals (Itami & Kimura, J. Neurosci. 32, 15000-15011 [2012]), which coincides with the timing of columnar shaping of L4 axons. Here, we show that the endocannabinoid 2-arachidonoylglycerol (2-AG) plays a crucial role in columnar shaping. We found that L4 axon projections were less organized until P12 and then became columnar after CB1Rs became functional. By contrast, the columnar organization of L4 axons was collapsed in mice genetically lacking diacylglycerol lipase α, the major enzyme for 2-AG synthesis. Intraperitoneally administered CB1R agonists shortened axon length, whereas knockout of CB1R in L4 neurons impaired columnar projection of their axons. Our results suggest that endocannabinoid signaling is crucial for shaping columnar axonal projection in the cerebral cortex.


Asunto(s)
Axones , Corteza Cerebral , Endocannabinoides , Animales , Axones/fisiología , Corteza Cerebral/crecimiento & desarrollo , Endocannabinoides/genética , Endocannabinoides/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Mutantes , Neuronas/fisiología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Corteza Somatosensorial/crecimiento & desarrollo
3.
Int J Obes (Lond) ; 48(2): 188-201, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38114812

RESUMEN

BACKGROUND: Overweight and obesity are the consequence of a sustained positive energy balance. Twin studies show high heritability rates pointing to genetics as one of the principal risk factors. By 2022, genomic studies led to the identification of almost 300 obesity-associated variants that could help to fill the gap of the high heritability rates. The endocannabinoid system is a critical regulator of metabolism for its effects on the central nervous system and peripheral tissues. Fatty acid amide hydrolase (FAAH) is a key enzyme in the inactivation of one of the two endocannabinoids, anandamide, and of its congeners. The rs324420 variant within the FAAH gene is a nucleotide missense change at position 385 from cytosine to adenine, resulting in a non-synonymous amino acid substitution from proline to threonine in the FAAH enzyme. This change increases sensitivity to proteolytic degradation, leading to reduced FAAH levels and increased levels of anandamide, associated with obesity-related traits. However, association studies of this variant with metabolic parameters have found conflicting results. This work aims to perform a systematic review of the existing literature on the association of the rs324420 variant in the FAAH gene with obesity and its related traits. METHODS: A literature search was conducted in PubMed, Web of Science, and Scopus. A total of 645 eligible studies were identified for the review. RESULTS/CONCLUSIONS: After the identification, duplicate elimination, title and abstract screening, and full-text evaluation, 28 studies were included, involving 28 183 individuals. We show some evidence of associations between the presence of the variant allele and higher body mass index, waist circumference, fat mass, and waist-to-hip ratio levels and alterations in glucose and lipid homeostasis. However, this evidence should be taken with caution, as many included studies did not report a significant difference between genotypes. These discordant results could be explained mainly by the pleiotropy of the endocannabinoid system, the increase of other anandamide-like mediators metabolized by FAAH, and the influence of gene-environment interactions. More research is necessary to study the endocannabinoidomic profiles and their association with metabolic diseases.


Asunto(s)
Amidohidrolasas , Ácidos Araquidónicos , Endocannabinoides , Obesidad , Alcamidas Poliinsaturadas , Humanos , Endocannabinoides/genética , Endocannabinoides/metabolismo , Obesidad/genética , Fenotipo
4.
Development ; 147(24)2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33168583

RESUMEN

The endocannabinoid (eCB) system, via the cannabinoid CB1 receptor, regulates neurodevelopment by controlling neural progenitor proliferation and neurogenesis. CB1 receptor signalling in vivo drives corticofugal deep layer projection neuron development through the regulation of BCL11B and SATB2 transcription factors. Here, we investigated the role of eCB signalling in mouse pluripotent embryonic stem cell-derived neuronal differentiation. Characterization of the eCB system revealed increased expression of eCB-metabolizing enzymes, eCB ligands and CB1 receptors during neuronal differentiation. CB1 receptor knockdown inhibited neuronal differentiation of deep layer neurons and increased upper layer neuron generation, and this phenotype was rescued by CB1 re-expression. Pharmacological regulation with CB1 receptor agonists or elevation of eCB tone with a monoacylglycerol lipase inhibitor promoted neuronal differentiation of deep layer neurons at the expense of upper layer neurons. Patch-clamp analyses revealed that enhancing cannabinoid signalling facilitated neuronal differentiation and functionality. Noteworthy, incubation with CB1 receptor agonists during human iPSC-derived cerebral organoid formation also promoted the expansion of BCL11B+ neurons. These findings unveil a cell-autonomous role of eCB signalling that, via the CB1 receptor, promotes mouse and human deep layer cortical neuron development.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Neuronas/metabolismo , Receptor Cannabinoide CB1/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Animales , Proliferación Celular/efectos de los fármacos , Cerebelo/crecimiento & desarrollo , Desarrollo Embrionario/genética , Endocannabinoides/agonistas , Endocannabinoides/genética , Endocannabinoides/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Organoides/crecimiento & desarrollo , Transducción de Señal/genética
5.
Eur J Neurol ; 30(10): 3212-3220, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37337838

RESUMEN

BACKGROUND AND PURPOSE: The endocannabinoid system (ECS) has been found altered in patients with multiple sclerosis (MS). However, whether the ECS alteration is present in the early stage of MS remains unknown. First, we aimed to compare the ECS profile between newly diagnosed MS patients and healthy controls (HCs). Next, we explored the association of the ECS, biomarkers of inflammation, and clinical parameters in newly diagnosed MS patients. METHODS: Whole blood gene expression of ECS components and levels of endocannabinoids in plasma were measured by real-time quantitative polymerase chain reaction and ultra-high-pressure liquid chromatography-mass spectrometry, respectively, in 66 untreated MS patients and 46 HCs. RESULTS: No differences were found in the gene expression or plasma levels of the selected ECS components between newly diagnosed MS patients and HCs. Interferon-γ, encoded by the gene IFNG, correlated positively (ρ = 0.60) with the expression of G protein-coupled receptor 55 (GPR55), and interleukin1ß (IL1B) correlated negatively (ρ = -0.50) with cannabinoid receptor 2 (CNR2) in HCs. CONCLUSIONS: We found no alteration in the peripheral ECS between untreated patients with MS and HC. Furthermore, our results indicate that the ECS has a minor overall involvement in the early stage of MS on inflammatory markers and clinical parameters when compared with HCs.


Asunto(s)
Endocannabinoides , Esclerosis Múltiple , Humanos , Endocannabinoides/genética , Endocannabinoides/metabolismo , Endocannabinoides/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Inflamación , Espectrometría de Masas , Biomarcadores
6.
J Neurosci Res ; 100(3): 731-743, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34496065

RESUMEN

The endocannabinoid system is an important regulator of emotional responses such as fear, and a number of studies have implicated endocannabinoid signaling in anxiety. The fatty acid amide hydrolase (FAAH) C385A polymorphism, which is associated with enhanced endocannabinoid signaling in the brain, has been identified across species as a potential protective factor from anxiety. In particular, adults with the variant FAAH 385A allele have greater fronto-amygdala connectivity and lower anxiety symptoms. Whether broader network-level differences in connectivity exist, and when during development this neural phenotype emerges, remains unknown and represents an important next step in understanding how the FAAH C385A polymorphism impacts neurodevelopment and risk for anxiety disorders. Here, we leveraged data from 3,109 participants in the nationwide Adolescent Brain Cognitive Development Study℠ (10.04 ± 0.62 years old; 44.23% female, 55.77% male) and a cross-validated, data-driven approach to examine associations between genetic variation and large-scale resting-state brain networks. Our findings revealed a distributed brain network, comprising functional connections that were both significantly greater (95% CI for p values = [<0.001, <0.001]) and lesser (95% CI for p values = [0.006, <0.001]) in A-allele carriers relative to non-carriers. Furthermore, there was a significant interaction between genotype and the summarized connectivity of functional connections that were greater in A-allele carriers, such that non-carriers with connectivity more similar to A-allele carriers (i.e., greater connectivity) had lower anxiety symptoms (ß = -0.041, p = 0.030). These findings provide novel evidence of network-level changes in neural connectivity associated with genetic variation in endocannabinoid signaling and suggest that genotype-associated neural differences may emerge at a younger age than genotype-associated differences in anxiety.


Asunto(s)
Amígdala del Cerebelo , Endocannabinoides , Adolescente , Amígdala del Cerebelo/fisiología , Ansiedad/genética , Trastornos de Ansiedad , Endocannabinoides/genética , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Polimorfismo de Nucleótido Simple/genética
7.
Nat Chem Biol ; 16(2): 197-205, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31844304

RESUMEN

Phospholipids, the most abundant membrane lipid components, are crucial in maintaining membrane structures and homeostasis for biofunctions. As a structurally diverse and tightly regulated system involved in multiple organelles, phospholipid metabolism is complicated to manipulate. Thus, repurposing phospholipids for lipid-derived chemical production remains unexplored. Herein, we develop a Saccharomyces cerevisiae platform for de novo production of oleoylethanolamide, a phospholipid derivative with promising pharmacological applications in ameliorating lipid dysfunction and neurobehavioral symptoms. Through deregulation of phospholipid metabolism, screening of biosynthetic enzymes, engineering of subcellular trafficking and process optimization, we could produce oleoylethanolamide at a titer of 8,115.7 µg l-1 and a yield on glucose of 405.8 µg g-1. Our work provides a proof-of-concept study for systemically repurposing phospholipid metabolism for conversion towards value-added biological chemicals, and this multi-faceted framework may shed light on tailoring phospholipid metabolism in other microbial hosts.


Asunto(s)
Endocannabinoides/biosíntesis , Ingeniería Metabólica/métodos , Ácidos Oléicos/biosíntesis , Fosfolípidos/metabolismo , Saccharomyces cerevisiae/metabolismo , Acilcoenzima A/genética , CDPdiacilglicerol-Serina O-Fosfatidiltransferasa/genética , CDPdiacilglicerol-Serina O-Fosfatidiltransferasa/metabolismo , Coenzima A Ligasas/genética , Endocannabinoides/genética , Enzimas/genética , Enzimas/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Fúngica de la Expresión Génica , Glucosa/metabolismo , Lisofosfolipasa/genética , Lisofosfolipasa/metabolismo , Microorganismos Modificados Genéticamente , Monoacilglicerol Lipasas/genética , Monoacilglicerol Lipasas/metabolismo , Ácidos Oléicos/genética , Proteínas Periplasmáticas/genética , Proteínas Periplasmáticas/metabolismo , Fosfolípidos/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
8.
J Cell Physiol ; 236(2): 1445-1453, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32696508

RESUMEN

Endocannabinoids are well-known regulators of neurotransmission by activating the cannabinoid (CB) receptors. Endocannabinoids are being used extensively for the treatment of various neurological disorders such as Alzheimer's and Parkinson's diseases. Although endocannabinoids are well studied in cell survival, proliferation, and differentiation in various neurological disorders and several cancers, the functional role in the regulation of blood cell development is less examined. In the present study, virodhamine, which is an agonist of CB receptor-2, was used to examine its effect on megakaryocytic development from a megakaryoblastic cell. We observed that virodhamine increases cell adherence, cell size, and cytoplasmic protrusions. Interestingly, we have also observed large nucleus and increased expression of megakaryocytic marker (CD61), which are the typical hallmarks of megakaryocytic differentiation. Furthermore, the increased expression of CB2 receptor was noticed in virodhamine-induced megakaryocytic cells. The effect of virodhamine on megakaryocytic differentiation could be mediated through CB2 receptor. Therefore, we have studied virodhamine induced molecular regulation of megakaryocytic differentiation; mitogen-activated protein kinase (MAPK) activity, mitochondrial function, and reactive oxygen species (ROS) production were majorly affected. The altered mitochondrial functions and ROS production is the crucial event associated with megakaryocytic differentiation and maturation. In the present study, we report that virodhamine induces megakaryocytic differentiation by triggering MAPK signaling and ROS production either through MAPK effects on ROS-generating enzymes or by the target vanilloid receptor 1-mediated regulation of mitochondrial function.


Asunto(s)
Endocannabinoides/metabolismo , Hematopoyesis/genética , Receptor Cannabinoide CB2/genética , Canales Catiónicos TRPV/genética , Ácidos Araquidónicos/metabolismo , Cannabinoides/farmacología , Adhesión Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Endocannabinoides/genética , Regulación del Desarrollo de la Expresión Génica/genética , Hematopoyesis/efectos de los fármacos , Humanos , Megacariocitos/efectos de los fármacos , Megacariocitos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor Cannabinoide CB1
9.
Neuroimage ; 224: 117426, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33035668

RESUMEN

Evidence from neuroimaging and genetic studies supports the concept that brain aging mirrors development. However, it is unclear whether mechanisms linking brain development and aging provide new insights to delay aging and potentially reverse it. This study determined biological mechanisms and phenotypic traits underpinning brain alterations across the lifespan and in aging by examining spatio-temporal correlations between gene expression and cortical volumes using datasets d with the age range from 2 to 82 years. We revealed that a large proportion of genes whose expression was associated with cortical volumes across the lifespan were in astrocytes. These genes, which showed up-regulation during development and down-regulation during aging, contributed to fundamental homeostatic functions of astrocytes. Included among these genes were those encoding components of cAMP, Ras, and retrograde endocannabinoid signaling pathways. Genes associated with cortical volumes in the same data aged above 55 years were also enriched for the sphingolipid, renin-angiotensin system (RAS), proteasome, and TGF-ß signaling pathway, which is linked to senescence-associated secretory phenotypes. Neuroticism, drinking, and smoking were the common phenotypic traits in the lifespan and aging, while memory was the unique phenotype associated with aging. These findings provide biological mechanisms mirroring development and aging as well as unique to aging.


Asunto(s)
Envejecimiento/genética , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Expresión Génica/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Astrocitos/metabolismo , Grosor de la Corteza Cerebral , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/patología , Niño , Preescolar , AMP Cíclico/genética , AMP Cíclico/metabolismo , Regulación hacia Abajo , Endocannabinoides/genética , Endocannabinoides/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Complejo de la Endopetidasa Proteasomal/genética , Sistema Renina-Angiotensina/genética , Análisis Espacio-Temporal , Esfingolípidos/genética , Esfingolípidos/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , Adulto Joven , Proteínas ras/genética , Proteínas ras/metabolismo
10.
Int J Mol Sci ; 22(6)2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33809047

RESUMEN

The administration of a ketogenic diet (KD) has been considered therapeutic in subjects with irritable bowel syndrome (IBS). This study aimed to investigate the molecular mechanisms by which a low-carbohydrate diet, such as KD, can improve gastrointestinal symptoms and functions in an animal model of IBS by evaluating possible changes in intestinal tissue expression of endocannabinoid receptors. In rats fed a KD, we detected a significant restoration of cell damage to the intestinal crypt base, a histological feature of IBS condition, and upregulation of CB1 and CB2 receptors. The diet also affected glucose metabolism and intestinal membrane permeability, with an overexpression of the glucose transporter GLUT1 and tight junction proteins in treated rats. The present data suggest that CB receptors represent one of the molecular pathways through which the KD works and support possible cannabinoid-mediated protection at the intestinal level in the IBS rats after dietary treatment.


Asunto(s)
Síndrome del Colon Irritable/dietoterapia , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB2/genética , Receptores de Cannabinoides/genética , Animales , Cannabinoides/metabolismo , Dieta Cetogénica/efectos adversos , Modelos Animales de Enfermedad , Endocannabinoides/genética , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Síndrome del Colon Irritable/genética , Síndrome del Colon Irritable/patología , Ratas
11.
Molecules ; 27(1)2021 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-35011234

RESUMEN

The endocannabinoidome (expanded endocannabinoid system, eCBome)-gut microbiome (mBIome) axis plays a fundamental role in the control of energy intake and processing. The liver-expressed antimicrobial peptide 2 (LEAP2) is a recently identified molecule acting as an antagonist of the ghrelin receptor and hence a potential effector of energy metabolism, also at the level of the gastrointestinal system. Here we investigated the role of the eCBome-gut mBIome axis in the control of the expression of LEAP2 in the liver and, particularly, the intestine. We confirm that the small intestine is a strong contributor to the circulating levels of LEAP2 in mice, and show that: (1) intestinal Leap2 expression is profoundly altered in the liver and small intestine of 13 week-old germ-free (GF) male mice, which also exhibit strong alterations in eCBome signaling; fecal microbiota transfer (FMT) from conventionally raised to GF mice completely restored normal Leap2 expression after 7 days from this procedure; in 13 week-old female GF mice no significant change was observed; (2) Leap2 expression in organoids prepared from the mouse duodenum is elevated by the endocannabinoid noladin ether, whereas in human Caco-2/15 epithelial intestinal cells it is elevated by PPARγ activation by rosiglitazone; (3) Leap2 expression is elevated in the ileum of mice with either high-fat diet-or genetic leptin signaling deficiency-(i.e., ob/ob and db/db mice) induced obesity. Based on these results, we propose that LEAP2 originating from the small intestine may represent a player in eCBome- and/or gut mBIome-mediated effects on food intake and energy metabolism.


Asunto(s)
Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Endocannabinoides/genética , Microbioma Gastrointestinal/genética , Receptores de Ghrelina/antagonistas & inhibidores , Animales , Células CACO-2 , Dieta Alta en Grasa , Femenino , Glicéridos/metabolismo , Humanos , Intestinos , Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Obesidad , ARN Mensajero/genética , Rosiglitazona/metabolismo , Transducción de Señal , Espectrometría de Masas en Tándem
12.
J Cell Physiol ; 235(12): 9795-9805, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32488870

RESUMEN

Electrical stimulation (ES) has been shown to improve some of impairments after spinal cord injury (SCI), but the underlying mechanisms remain unclear. The Wnt signaling pathways and the endocannabinoid system appear to be modulated in response to SCI. This study aimed to investigate the effect of ES therapy on the activity of canonical/noncanonical Wnt signaling pathways, brain-derived neurotrophic factor (BDNF), and fatty-acid amide hydrolase (FAAH), which regulate endocannabinoids levels. Forty male Wistar rats were randomly divided into four groups: (a) Sham, (b) laminectomy + epidural subthreshold ES, (c) SCI, and (d) SCI + epidural subthreshold ES. A moderate contusion SCI was performed at the thoracic level (T10). Epidural subthreshold ES was delivered to upper the level of T10 segment every day (1 hr/rat) for 2 weeks. Then, animals were killed and immunoblotting was used to assess spinal cord parameters. Results revealed that ES intervention for 14 days could significantly increase wingless-type3 (Wnt3), Wnt7, ß-catenin, Nestin, and cyclin D1 levels, as well as phosphorylation of glycogen synthase kinase 3ß and Jun N-terminal kinase. Additionally, SCI reduced BDNF and FAAH levels, and ES increased BDNF and FAAH levels in the injury site. We propose that ES therapy may improve some of impairments after SCI through Wnt signaling pathways. Outcomes also suggest that BDNF and FAAH are important players in the beneficial impacts of ES therapy. However, the precise mechanism of BDNF, FAAH, and Wnt signaling pathways on SCI requires further investigation.


Asunto(s)
Amidohidrolasas/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Endocannabinoides/genética , Traumatismos de la Médula Espinal/terapia , Animales , Modelos Animales de Enfermedad , Estimulación Eléctrica/métodos , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Masculino , Ratas , Recuperación de la Función/efectos de los fármacos , Médula Espinal/patología , Médula Espinal/efectos de la radiación , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/patología , Tórax/patología , Tórax/efectos de la radiación , Vía de Señalización Wnt/efectos de la radiación , beta Catenina/genética
13.
Hum Reprod ; 35(2): 265-274, 2020 02 29.
Artículo en Inglés | MEDLINE | ID: mdl-31990346

RESUMEN

STUDY QUESTION: What are the effects of endocannabinoid anandamide (AEA) in uterine natural killer (unK) cells from miscarriage decidua, regarding their cytokine profile and endometrial stromal cell (ESC) crosstalk? SUMMARY ANSWER: uNK-conditioned media from miscarriage samples present high TNF-α levels which inhibit ESC decidualisation. WHAT IS KNOWN ALREADY: AEA plasma levels are higher in women who have suffered a miscarriage. Moreover, AEA inhibits ESC proliferation and differentiation, although the levels and impact on the uNK cell cytokine profile at the feto-maternal interface remain elusive. STUDY DESIGN, SIZE, DURATION: This laboratory-based study used human primary uNK cells which were isolated from first-trimester decidua (gestational age, 5-12 weeks) derived from 8 women with elective pregnancy termination and 18 women who suffered a miscarriage. PARTICIPANTS/MATERIALS, SETTING, METHODS: The first-trimester placental tissues were assayed for AEA levels by UPLC-MS/MS and respective enzymatic profile by western blot. The uNK cells were isolated and maintained in culture. The expression of angiogenic markers in uNK cells was examined by quantitative PCR (qPCR). The uNK-conditioned medium was analysed for IFN-γ, TNF-α and IL-10 production by enzyme-linked immunosorbent assay, and the impact on ESC differentiation was assessed by measuring decidual markers Prl, Igfbp-1 and Fox01 mRNA expression using qPCR. MAIN RESULTS AND THE ROLE OF CHANCE: AEA levels were higher in miscarriage decidua compared with decidua from elective terminations. The uNK cell-conditioned medium from the miscarriage samples exhibited high TNF-α levels and interfered with the decidualisation of ESCs. Exacerbated inflammation and elevated TNF-α levels at the feto-maternal interface may trigger AEA signalling pathways that, in turn, may impact decidualisation and the angiogenic ability of uNK cells. LARGE-SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Primary uNK cell responses are based on a simple in vitro model. Thus, in complex microenvironments, such as the feto-maternal interface, the mechanisms may not be exactly the same. Also, the inflammatory events of miscarriage that, in this study, have happened prior to processing of the samples may cause different responses to that observed. In addition, the magnitude of the inflammatory response, required to trigger the AEA pathways that impact decidualisation and the uNK angiogenic ability in vivo, is still unclear. WIDER IMPLICATIONS OF THE FINDINGS: The endocannabinoid AEA is a modulator of reproductive competence. AEA not only may contribute to neuroendocrine homeostasis but also can take part in uterine changes occurring during early pregnancy. STUDY FUNDING/COMPETING INTEREST(S): The work was supported by UID/MULTI/04378/2019 with funding from Fundação para a Ciência e a Tecnologia (FCT)/MCTES through national funds and PORTUGAL 2020 Partnership Agreement, NORTE-01-0145-FEDER-000024. S.C. Cunha acknowledges FCT for the IF/01616/2015 contract. There are no conflicts of interest.


Asunto(s)
Aborto Espontáneo/metabolismo , Cannabinoides/metabolismo , Endocannabinoides/fisiología , Células Asesinas Naturales/metabolismo , Placenta/metabolismo , Receptores de Cannabinoides/fisiología , Células del Estroma/metabolismo , Ácidos Araquidónicos , Agonistas de Receptores de Cannabinoides , Endocannabinoides/genética , Endocannabinoides/metabolismo , Endometrio/metabolismo , Femenino , Humanos , Alcamidas Poliinsaturadas , Portugal , Embarazo , Receptores de Cannabinoides/genética , Receptores de Cannabinoides/metabolismo
14.
Nat Rev Neurosci ; 16(10): 579-94, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26373473

RESUMEN

Brain endocannabinoid (eCB) signalling influences the motivation for natural rewards (such as palatable food, sexual activity and social interaction) and modulates the rewarding effects of addictive drugs. Pathological forms of natural and drug-induced reward are associated with dysregulated eCB signalling that may derive from pre-existing genetic factors or from prolonged drug exposure. Impaired eCB signalling contributes to dysregulated synaptic plasticity, increased stress responsivity, negative emotional states and cravings that propel addiction. Understanding the contributions of eCB disruptions to behavioural and physiological traits provides insight into the eCB influence on addiction vulnerability.


Asunto(s)
Endocannabinoides/fisiología , Abuso de Marihuana/fisiopatología , Recompensa , Transducción de Señal/fisiología , Animales , Conducta Adictiva/psicología , Encéfalo/fisiología , Encéfalo/fisiopatología , Endocannabinoides/genética , Humanos , Vías Nerviosas/fisiopatología , Transducción de Señal/genética
15.
Arch Toxicol ; 94(12): 4131-4141, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32833042

RESUMEN

In animals, research in the past two decades has demonstrated the strong involvement of the endocannabinoid system (ECS) in numerous steps of the reproductive process, including ovarian physiology. Reproductive lifespan is closely related to the number of nongrowing ovarian follicles, called ovarian reserve (OR), which is definitively established during foetal life. Thus, OR damage may lead to poor reproductive outcomes and a shortened reproductive lifespan. We investigated whether prenatal ECS modulation had an effect on the OR at different ages in the rat offspring. Four groups of gestating female rats (F0) were exposed to the CB1-/CB2-receptor agonist WIN55212 (0.5 mg/kg), the CB1R inverse agonist SR141716 (3 mg/kg) or Δ9THC (5 mg/kg) and were compared to negative control groups. OR was histologically assessed at different postnatal timepoints (F1 individuals): postnatal day (PND) 6, PND40 and PND90. At PND6, prenatal exposure had no effect on OR. In the young adult group (PND90) exposed during gestation to WIN55212, we observed a CB1R-mediated delayed OR decrease, which was reversed by prenatal CB1R blockade by SR141716. Conversely, after prenatal SR141716 exposure, we observed higher OR counts at PND90. RT-PCR experiments also showed that prenatal ECS modulation perturbed the mRNA levels of ECS enzymes and OR regulation genes. Our findings support the role of the ECS in OR regulation during the foetal life of rats and highlight the need for further studies to elucidate its precise role in OR physiology.


Asunto(s)
Agonistas de Receptores de Cannabinoides/toxicidad , Dronabinol/toxicidad , Reserva Ovárica/efectos de los fármacos , Ovario/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal , Receptor Cannabinoide CB1/agonistas , Animales , Benzoxazinas/toxicidad , Antagonistas de Receptores de Cannabinoides/farmacología , Agonismo Inverso de Drogas , Endocannabinoides/genética , Endocannabinoides/metabolismo , Femenino , Regulación de la Expresión Génica , Edad Gestacional , Morfolinas/toxicidad , Naftalenos/toxicidad , Reserva Ovárica/genética , Ovario/metabolismo , Ovario/fisiopatología , Embarazo , Ratas Wistar , Receptor Cannabinoide CB1/metabolismo , Rimonabant/farmacología
16.
Int J Mol Sci ; 21(19)2020 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-32992491

RESUMEN

Different Follicle Stimulating Hormone (FSH) formulation and Luteinizing Hormone (LH) are used in Assisted Reproductive Technology (ART) to induce follicles development and oocytes maturation, but it is still under debate which protocol is to be preferred. In the present study, the different effects on cumulus cells (CCs) of three controlled ovarian stimulation (COS) protocols, based on urinary FSH, recombinant FSH, or human Menopausal Gonadotropin (hMG) administration, were assessed. CCs were obtained from 42 normal-responders women undergoing COS, randomly divided into three groups according to the used gonadotropin formulation. Differences were found in the expression of genes belonging to the endocannabinoid system (the receptors CNR1, CNR2 and TRPV1, and the enzymes involved in the metabolisms of anandamide, NAPE-PLD and FAAH, and 2-acylglycerol, DAGL and MAGL); consistently, changes in lipid (PPARα, and FASN) and carbohydrate (GLUT1 and GLUT9) metabolisms, in CCs' macromolecules composition (highlighted by Fourier Transform Infrared Microspectroscopy, FTIRM), and in the number of retrieved oocytes were found. For the first time, statistically significant evidence on the differences related to each COS protocol on the endocannabinoid system, metabolism and macromolecular composition of CCs was found, representing a proof of concept to be further confirmed in a larger cohort of patients.


Asunto(s)
Células del Cúmulo/efectos de los fármacos , Células del Cúmulo/metabolismo , Endocannabinoides/metabolismo , Hormona Folículo Estimulante Humana/farmacología , Menotropinas/farmacología , Inducción de la Ovulación/métodos , Transducción de Señal/efectos de los fármacos , Urofolitropina/farmacología , Adulto , Ácidos Araquidónicos/genética , Ácidos Araquidónicos/metabolismo , Células Cultivadas , Estudios de Cohortes , Endocannabinoides/genética , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Recuperación del Oocito , Alcamidas Poliinsaturadas/metabolismo , Proteínas Recombinantes/farmacología , Espectroscopía Infrarroja por Transformada de Fourier
17.
Int J Mol Sci ; 21(19)2020 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-33023013

RESUMEN

BACKGROUND: Cannabinoids induce biphasic effects on memory depending on stress levels. We previously demonstrated that different stress intensities, experienced soon after encoding, impaired rat short-term recognition memory in a time-of-day-dependent manner, and that boosting endocannabinoid anandamide (AEA) levels restored memory performance. Here, we examined if two different stress intensities and time-of-day alter hippocampal endocannabinoid tone, and whether these changes modulate short-term memory. METHODS: Male Sprague-Dawley rats were subjected to an object recognition task and exposed, at two different times of the day (i.e., morning or afternoon), to low or high stress conditions, immediately after encoding. Memory retention was assessed 1 hr later. Hippocampal AEA and 2-arachidonoyl glycerol (2-AG) content and the activity of their primary degrading enzymes, fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), were measured soon after testing. RESULTS: Consistent with our previous findings, low stress impaired 1-hr memory performance only in the morning, whereas exposure to high stress impaired memory independently of testing time. Stress exposure decreased AEA levels independently of memory alterations. Interestingly, exposure to high stress decreased 2-AG content and, accordingly, increased MAGL activity, selectively in the afternoon. Thus, to further evaluate 2-AG's role in the modulation of short-term recognition memory, rats were given bilateral intra-hippocampal injections of the 2-AG hydrolysis inhibitor KML29 immediately after training, then subjected to low or high stress conditions and tested 1 hr later. CONCLUSIONS: KML29 abolished the time-of-day-dependent impairing effects of stress on short-term memory, ameliorating short-term recognition memory performance.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Endocannabinoides/metabolismo , Glicéridos/metabolismo , Hipocampo/metabolismo , Memoria a Corto Plazo/fisiología , Amidohidrolasas/genética , Animales , Ácidos Araquidónicos/antagonistas & inhibidores , Ácidos Araquidónicos/genética , Benzodioxoles/farmacología , Emociones/fisiología , Endocannabinoides/antagonistas & inhibidores , Endocannabinoides/genética , Glicéridos/antagonistas & inhibidores , Glicéridos/genética , Hipocampo/efectos de los fármacos , Hipocampo/fisiología , Humanos , Masculino , Monoacilglicerol Lipasas/genética , Piperidinas/farmacología , Alcamidas Poliinsaturadas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética
18.
Int J Mol Sci ; 21(19)2020 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-33036283

RESUMEN

Osteoarthritis (OA) is a degenerative joint disease manifested by movement limitations and chronic pain. Endocannabinoid system (ECS) may modulate nociception via cannabinoid and TRPV1 receptors. The purpose of our study was to examine alterations in the spinal and joint endocannabinoid system during pain development in an animal model of OA. Wistar rats received intra-articular injection of 3mg of sodium monoiodoacetate (MIA) into the knee joint. Animals were sacrificed on day 2, 7, 14, 21, 28 after injection and lumbar spinal cord, cartilage and synovium were collected. Changes in the transcription levels of the ECS elements were measured. At the spinal level, gene expression levels of the cannabinoid and TRPV1 receptors as well as enzymes involved in anandamide synthesis and degradation were elevated in the advanced OA phase. In the joint, an important role of the synovium was demonstrated, since cartilage degeneration resulted in attenuation of the changes in the gene expression. Enzymes responsible for anandamide synthesis and degradation were upregulated particularly in the early stages of OA, presumably in response to early local joint inflammation. The presented study provides missing information about the MIA-induced OA model and encourages the development of a therapy focused on the molecular role of ECS.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Endocannabinoides/metabolismo , Osteoartritis/metabolismo , Dolor/metabolismo , Alcamidas Poliinsaturadas/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Ácidos Araquidónicos/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endocannabinoides/genética , Regulación de la Expresión Génica , Inyecciones Intraarticulares , Ácido Yodoacético/efectos adversos , Ácido Yodoacético/toxicidad , Articulación de la Rodilla/metabolismo , Osteoartritis/complicaciones , Osteoartritis/genética , Dolor/etiología , Dolor/genética , Ratas , Ratas Wistar , Canales Catiónicos TRPV/genética
19.
J Lipid Res ; 60(8): 1396-1409, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31167809

RESUMEN

Mammalian ω3- and ω6-PUFAs are synthesized from essential fatty acids (EFAs) or supplied by the diet. PUFAs are constitutive elements of membrane architecture and precursors of lipid signaling molecules. EFAs and long-chain (LC)-PUFAs are precursors in the synthesis of endocannabinoid ligands of Gi/o protein-coupled cannabinoid receptor (CB)1 and CB2 in the endocannabinoid system, which critically regulate energy homeostasis as the metabolic signaling system in hypothalamic neuronal circuits and behavioral parameters. We utilized the auxotrophic fatty acid desaturase 2-deficient (fads2-/-) mouse, deficient in LC-PUFA synthesis, to follow the age-dependent dynamics of the PUFA pattern in the CNS-phospholipidome in unbiased dietary studies of three cohorts on sustained LC-PUFA-free ω6-arachidonic acid- and DHA-supplemented diets and their impact on the precursor pool of CB1 ligands. We discovered the transformation of eicosa-all cis-5,11,14-trienoic acid, uncommon in mammalian lipidomes, into two novel endocannabinoids, 20:35,11,14-ethanolamide and 2-20:35,11,14-glycerol. Their function as ligands of CB1 has been characterized in HEK293 cells. Labeling experiments excluded Δ8-desaturase activity and proved the position specificity of FADS2. The fads2-/- mutant might serve as an unbiased model in vivo in the development of novel CB1 agonists and antagonists.


Asunto(s)
Endocannabinoides/metabolismo , Ácidos Grasos Omega-3/deficiencia , Ácidos Grasos Omega-6/deficiencia , Receptor Cannabinoide CB1/agonistas , Animales , Endocannabinoides/genética , Ácido Graso Desaturasas/deficiencia , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-6/farmacología , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo
20.
J Cell Physiol ; 234(6): 7893-7902, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30537148

RESUMEN

Obesity as a multifactorial disorder has been shown a dramatically growing trend recently. Besides genetic and environmental factors, dysregulation of the endocannabinoid system tone is involved in the pathogenesis of obesity. This study reviewed the potential efficacy of Oleoylethanolamide (OEA) as an endocannabinoid-like compound in the energy homeostasis and appetite control in people with obesity. OEA as a lipid mediator and bioactive endogenous ethanolamide fatty acid is structurally similar to the endocannabinoid system compounds; nevertheless, it is unable to induce to the cannabinoid receptors. Unlike endocannabinoids, OEA negatively acts on the food intake and suppress appetite via various mechanisms. Indeed, OEA as a ligand of PPAR-α, GPR-119, and TRPV1 receptors participates in the regulation of energy intake and energy expenditure, feeding behavior, and weight gain control. OEA delays meal initiation, reduces meal size, and increases intervals between meals. Considering side effects of some approaches used for the management of obesity such as antiobesity drugs and surgery as well as based on sufficient evidence about the protective effects of OEA in the improvement of common abnormalities in people with obese, its supplementation as a novel efficient and FDA approved pharmaceutical agent can be recommended.


Asunto(s)
Regulación del Apetito/efectos de los fármacos , Endocannabinoides/uso terapéutico , Metabolismo Energético/efectos de los fármacos , Obesidad/tratamiento farmacológico , Ácidos Oléicos/uso terapéutico , Endocannabinoides/genética , Endocannabinoides/metabolismo , Ácidos Grasos/metabolismo , Humanos , Lípidos/genética , Obesidad/genética , Obesidad/patología , Ácidos Oléicos/genética , PPAR alfa/metabolismo , Receptores Acoplados a Proteínas G/genética , Canales Catiónicos TRPV/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA