Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nutr Neurosci ; 26(11): 1159-1171, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36222323

RESUMEN

Emerging evidence is highlighting the microbiome as a key regulator of the effect of nutrition on gut-brain axis signaling. Nevertheless, it is not yet clear whether the impact of nutrition is moderating the microbiota-gut-brain interaction or if diet has a mediating role on microbiota composition and function to influence central nervous system function, brain phenotypes and behavior. Mechanistic evidence from cell-based in vitro studies, animal models and preclinical intervention studies are linking the gut microbiota to the effects of diet on brain function, but they have had limited translation to human intervention studies. While increasing evidence demonstrates the triangulating relationship between diet, microbiota, and brain function across the lifespan, future mechanistic and translational studies in the field of microbiota and nutritional neuroscience are warranted to inform potential strategies for prevention and management of several neurological, neurodevelopmental, neurodegenerative, and psychiatric disorders. This brief primer provides an overview of the most recent advances in the nutritional neuroscience - microbiome field, highlighting significant opportunities for future research.


Asunto(s)
Microbioma Gastrointestinal , Trastornos Mentales , Microbiota , Animales , Humanos , Encéfalo/fisiología , Eje Cerebro-Intestino , Microbioma Gastrointestinal/fisiología , Microbiota/fisiología
2.
Cell Mol Life Sci ; 79(8): 426, 2022 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-35841486

RESUMEN

Activated ghrelin receptor GHS-R1α triggers cell signalling pathways that modulate energy homeostasis and biosynthetic processes. However, the effects of ghrelin on mRNA translation are unknown. Using various reporter assays, here we demonstrate a rapid elevation of protein synthesis in cells within 15-30 min upon stimulation of GHS-R1α by ghrelin. We further show that ghrelin-induced activation of translation is mediated, at least in part, through the de-phosphorylation (de-suppression) of elongation factor 2 (eEF2). The levels of eEF2 phosphorylation at Thr56 decrease due to the reduced activity of eEF2 kinase, which is inhibited via Ser366 phosphorylation by rpS6 kinases. Being stress-susceptible, the ghrelin-mediated decrease in eEF2 phosphorylation can be abolished by glucose deprivation and mitochondrial uncoupling. We believe that the observed burst of translation benefits rapid restocking of neuropeptides, which are released upon GHS-R1α activation, and represents the most time- and energy-efficient way of prompt recharging the orexigenic neuronal circuitry.


Asunto(s)
Ghrelina , Biosíntesis de Proteínas , Ghrelina/metabolismo , Factor 2 de Elongación Peptídica/metabolismo , Fosforilación , Transducción de Señal/fisiología
3.
Brain Behav Immun ; 106: 115-126, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35995237

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterised by deficits in social behaviour, increased repetitive behaviour, anxiety and gastrointestinal symptoms. The aetiology of ASD is complex and involves an interplay of genetic and environmental factors. Emerging pre-clinical and clinical studies have documented a potential role for the gut microbiome in ASD, and consequently, the microbiota represents a potential target in the development of novel therapeutics for this neurodevelopmental disorder. In this study, we investigate the efficacy of the live biotherapeutic strain, Blautia stercoris MRx0006, in attenuating some of the behavioural deficits in the autism-relevant, genetic mouse model, BTBR T+ Itpr3tf/J (BTBR). We demonstrate that daily oral administration with MRx0006 attenuates social deficits while also decreasing repetitive and anxiety-like behaviour. MRx0006 administration increases the gene expression of oxytocin and its receptor in hypothalamic cells in vitro and increases the expression of hypothalamic arginine vasopressin and oxytocin mRNA in BTBR mice. Additionally at the microbiome level, we observed that MRx0006 administration decreases the abundance of Alistipes putredinis, and modulates the faecal microbial metabolite profile. This alteration in the metabolite profile possibly underlies the observed increase in expression of oxytocin, arginine vasopressin and its receptors, and the consequent improvements in behavioural outcomes. Taken together, these findings suggest that the live biotherapeutic MRx0006 may represent a viable and efficacious treatment option for the management of physiological and behavioural deficits associated with ASD.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Animales , Ansiedad , Arginina Vasopresina , Trastorno del Espectro Autista/metabolismo , Trastorno Autístico/metabolismo , Clostridiales , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos , Oxitocina , ARN Mensajero/metabolismo
4.
J Neurochem ; 157(5): 1495-1524, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33368280

RESUMEN

Our gut harbours trillions of microorganisms essential for the maintenance of homeostasis and host physiology in health and disease. In the last decade, there has been a growing interest in understanding the bidirectional pathway of communication between our microbiota and the central nervous system. With regard to reward processes there is accumulating evidence from both animal and human studies that this axis may be a key factor in gating reward valence. Focusing on the mesocorticolimbic pathway, we will discuss how the intestinal microbiota is involved in regulating brain reward functions, both in natural (i.e. eating, social or sexual behaviours) and non-natural reinforcers (drug addiction behaviours including those relevant to alcohol, psychostimulants, opioids and cannabinoids). We will integrate preclinical and clinical evidence suggesting that the microbiota-gut-brain axis could be implicated in the development of disorders associated with alterations in the reward system and how it may be targeted as a promising therapeutic strategy. Cover Image for this issue: https://doi.org/10.1111/jnc.15065.


Asunto(s)
Encéfalo/fisiología , Microbioma Gastrointestinal/fisiología , Recompensa , Animales , Conducta Adictiva/fisiopatología , Conducta Adictiva/psicología , Humanos , Vías Nerviosas/fisiología
5.
Int J Obes (Lond) ; 45(3): 588-598, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33223517

RESUMEN

BACKGROUND: Early consumption of obesogenic diets, rich in saturated fat and added sugar, is associated with a plethora of biological dysfunctions, at both peripheral and brain levels. Obesity is also linked to decreased vitamin A bioavailability, an essential molecule for brain plasticity and memory function. METHODS: Here we investigated in mice whether dietary vitamin A supplementation (VAS) could prevent some of the metabolic, microbiota, neuronal and cognitive alterations induced by obesogenic, high-fat and high-sugar diet (HFSD) exposure from weaning to adulthood, i.e. covering periadolescent period. RESULTS: As expected, VAS was effective in enhancing peripheral vitamin A levels as well as hippocampal retinoic acid levels, the active metabolite of vitamin A, regardless of the diet. VAS attenuated HFSD-induced excessive weight gain, without affecting metabolic changes, and prevented alterations of gut microbiota α-diversity. In HFSD-fed mice, VAS prevented recognition memory deficits but had no effect on aversive memory enhancement. Interestingly, VAS alleviated both HFSD-induced higher neuronal activation and lower glucocorticoid receptor phosphorylation in the hippocampus after training. CONCLUSION: Dietary VAS was protective against the deleterious effects of early obesogenic diet consumption on hippocampal function, possibly through modulation of the gut-brain axis.


Asunto(s)
Cognición/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Microbioma Gastrointestinal/efectos de los fármacos , Vitamina A , Animales , Eje Cerebro-Intestino/efectos de los fármacos , Hipocampo/química , Hipocampo/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Vitamina A/administración & dosificación , Vitamina A/farmacología
6.
Mol Psychiatry ; 25(10): 2567-2583, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31092898

RESUMEN

Male middle age is a transitional period where many physiological and psychological changes occur leading to cognitive and behavioural alterations, and a deterioration of brain function. However, the mechanisms underpinning such changes are unclear. The gut microbiome has been implicated as a key mediator in the communication between the gut and the brain, and in the regulation of brain homeostasis, including brain immune cell function. Thus, we tested whether targeting the gut microbiome by prebiotic supplementation may alter microglia activation and brain function in ageing. Male young adult (8 weeks) and middle-aged (10 months) C57BL/6 mice received diet enriched with a prebiotic (10% oligofructose-enriched inulin) or control chow for 14 weeks. Prebiotic supplementation differentially altered the gut microbiota profile in young and middle-aged mice with changes correlating with faecal metabolites. Functionally, this translated into a reversal of stress-induced immune priming in middle-aged mice. In addition, a reduction in ageing-induced infiltration of Ly-6Chi monocytes into the brain coupled with a reversal in ageing-related increases in a subset of activated microglia (Ly-6C+) was observed. Taken together, these data highlight a potential pathway by which targeting the gut microbiome with prebiotics can modulate the peripheral immune response and alter neuroinflammation in middle age. Our data highlight a novel strategy for the amelioration of age-related neuroinflammatory pathologies and brain function.


Asunto(s)
Envejecimiento/inmunología , Encéfalo/inmunología , Microbioma Gastrointestinal/fisiología , Prebióticos , Animales , Heces/química , Heces/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología
7.
Int J Mol Sci ; 22(4)2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33672070

RESUMEN

Serotonin communication operates mainly in the extracellular space and cerebrospinal fluid (CSF), using volume transmission with serotonin moving from source to target cells (neurons and astroglia) via energy gradients, leading to the diffusion and convection (flow) of serotonin. One emerging concept in depression is that disturbances in the integrative allosteric receptor-receptor interactions in highly vulnerable 5-HT1A heteroreceptor complexes can contribute to causing major depression and become novel targets for the treatment of major depression (MD) and anxiety. For instance, a disruption and/or dysfunction in the 5-HT1A-FGFR1 heteroreceptor complexes in the raphe-hippocampal serotonin neuron systems can contribute to the development of MD. It leads inter alia to reduced neuroplasticity and potential atrophy in the raphe-cortical and raphe-striatal 5-HT pathways and in all its forebrain networks. Reduced 5-HT1A auto-receptor function, increased plasticity and trophic activity in the midbrain raphe 5-HT neurons can develop via agonist activation of allosteric receptor-receptor interactions in the 5-HT1A-FGFR1 heterocomplex. Additionally, the inhibitory allosteric receptor-receptor interactions in the 5-HT1AR-5-HT2AR isoreceptor complex therefore likely have a significant role in modulating mood, involving a reduction of postjunctional 5-HT1AR protomer signaling in the forebrain upon activation of the 5-HT2AR protomer. In addition, oxytocin receptors (OXTRs) play a significant and impressive role in modulating social and cognitive related behaviors like bonding and attachment, reward and motivation. Pathological blunting of the OXTR protomers in 5-HT2AR and especially in 5-HT2CR heteroreceptor complexes can contribute to the development of depression and other types of psychiatric diseases involving disturbances in social behaviors. The 5-HTR heterocomplexes are novel targets for the treatment of MD.


Asunto(s)
Depresión/metabolismo , Trastorno Depresivo Mayor/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Serotonina/metabolismo , Transducción de Señal , Animales , Humanos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT2A/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Receptores de Oxitocina/metabolismo
8.
FASEB J ; 33(1): 518-531, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30020830

RESUMEN

The ghrelin receptor [growth hormone secretagogue receptor (GHSR)-1a] represents a promising pharmacologic target for the treatment of metabolic disorders, including obesity and cachexia, via central appetite modulation. The GHSR-1a has a complex pharmacology, highlighted by G-protein-dependent and -independent downstream signaling pathways and high basal constitutive activity. The functional selectivity and signaling bias of many GHSR-1a-specific ligands has not been fully characterized. In this study, we investigated the pharmacologic properties of ghrelin, MK-0677, L692,585, and [d-Lys3]-growth hormone-releasing peptide-6 (Dlys), JMV2959, and [d-Arg(1),d-Phe(5),d-Trp(7, 9),Leu(11)]-substance P (SP-analog). We investigated their effect on basal GHSR-1a constitutive signaling, ligand-directed downstream GHSR-1a signaling, functional selectivity, and signaling bias. Dlys behaved as a partial antagonist with a strong bias toward GHSR-1a-ß-arrestin signaling, whereas JMV2959 acted as a full unbiased GHSR-1a antagonist. Moreover, the SP-analog behaved as an inverse agonist increasing G-protein-dependent signaling, but only at high concentrations, whereas, at low concentrations, the SP-analog attenuated ß-arrestin-dependent signaling. Considering the limited success in the clinical development of GHSR-1a-targeted drugs so far, these findings provide a novel insight into the pharmacologic characteristics of GHSR-1a ligands and their signaling bias, which has important implications in the design of novel, more selective GHSR-1a ligands with predictable functional outcome and selectivity for preclinical and clinical drug development.-Ramirez, V. T., van Oeffelen, W. E. P. A., Torres-Fuentes, C., Chruscicka, B., Druelle, C., Golubeva, A. V., van de Wouw, M., Dinan, T. G., Cryan, J. F., Schellekens, H. Differential functional selectivity and downstream signaling bias of ghrelin receptor antagonists and inverse agonists.


Asunto(s)
Ghrelina/farmacología , Fragmentos de Péptidos/farmacología , Receptores de Ghrelina/agonistas , Receptores de Ghrelina/antagonistas & inhibidores , beta-Arrestina 1/metabolismo , Células HEK293 , Humanos , Receptores de Ghrelina/metabolismo , Transducción de Señal
9.
FASEB J ; 33(12): 13546-13559, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31545915

RESUMEN

The gastrointestinal microbiota is emerging as a unique and inexhaustible source for metabolites with potential to modulate G-protein coupled receptors (GPCRs). The ghrelin receptor [growth hormone secretagogue receptor (GHSR)-1a] is a GPCR expressed throughout both the gut and the brain and plays a crucial role in maintaining energy balance, metabolism, and the central modulation of food intake, motivation, reward, and mood. To date, few studies have investigated the potential of the gastrointestinal microbiota and its metabolites to modulate GPCR signaling. Here we investigate the ability of short-chain fatty acids (SCFAs), lactate, and different bacterial strains, including Bifidobacterium and Lactobacillus genera, to modulate GHSR-1a signaling. We identify, for what is to our knowledge the first time, a potent effect of microbiota-derived metabolites on GHSR-1a signaling with potential significant consequences for host metabolism and physiology. We show that SCFAs, lactate, and bacterial supernatants are able to attenuate ghrelin-mediated signaling through the GHSR-1a. We suggest a novel route of communication between the gut microbiota and the host via modulation of GHSR-1a receptor signaling. Together, this highlights the emerging therapeutic potential in the exploration of the microbiota metabolome in the specific targeting of key GPCRs, with pleiotropic actions that span both the CNS and periphery.-Torres-Fuentes, C., Golubeva, A. V., Zhdanov, A. V., Wallace, S., Arboleya, S., Papkovsky, D. B., El Aidy, S., Ross, P., Roy, B. L., Stanton, C., Dinan, T. G., Cryan, J. F., Schellekens, H. Short-chain fatty acids and microbiota metabolites attenuate ghrelin receptor signaling.


Asunto(s)
Bacterias/metabolismo , Ácidos Grasos Volátiles/farmacología , Microbioma Gastrointestinal , Regulación de la Expresión Génica/efectos de los fármacos , Ácido Láctico/farmacología , Receptores de Ghrelina/metabolismo , Ghrelina/farmacología , Células HEK293 , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores de Ghrelina/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
10.
Int J Mol Sci ; 19(6)2018 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-29848961

RESUMEN

Cachexia is a metabolic wasting disorder characterized by progressive weight loss, muscle atrophy, fatigue, weakness, and appetite loss. Cachexia is associated with almost all major chronic illnesses including cancer, heart failure, obstructive pulmonary disease, and kidney disease and significantly impedes treatment outcome and therapy tolerance, reducing physical function and increasing mortality. Current cachexia treatments are limited and new pharmacological strategies are needed. Agonists for the growth hormone secretagogue (GHS-R1a), or ghrelin receptor, prospectively regulate the central regulation of appetite and growth hormone secretion, and therefore have tremendous potential as cachexia therapeutics. Non-peptide GHS-R1a agonists are of particular interest, especially given the high gastrointestinal degradation of peptide-based structures, including that of the endogenous ligand, ghrelin, which has a half-life of only 30 min. However, few compounds have been reported in the literature as non-peptide GHS-R1a agonists. In this paper, we investigate the in vitro potential of quinolone compounds to modulate the GHS-R1a in both transfected human cells and mouse hypothalamic cells. These chemically synthesized compounds demonstrate a promising potential as GHS-R1a agonists, shown by an increased intracellular calcium influx. Further studies are now warranted to substantiate and exploit the potential of these novel quinolone-based compounds as orexigenic therapeutics in conditions of cachexia and other metabolic and eating disorders.


Asunto(s)
Caquexia/tratamiento farmacológico , Caquexia/metabolismo , Quinolonas/farmacología , Quinolonas/uso terapéutico , Receptores de Ghrelina/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Ghrelina/metabolismo , Humanos , Ratones , Transducción de Señal/efectos de los fármacos
11.
Int J Mol Sci ; 19(9)2018 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-30223587

RESUMEN

Recent times have seen an increasing move towards harnessing the health-promoting benefits of food and dietary constituents while providing scientific evidence to substantiate their claims. In particular, the potential for bioactive protein hydrolysates and peptides to enhance health in conjunction with conventional pharmaceutical therapy is being investigated. Dairy-derived proteins have been shown to contain bioactive peptide sequences with various purported health benefits, with effects ranging from the digestive system to cardiovascular circulation, the immune system and the central nervous system. Interestingly, the ability of dairy proteins to modulate metabolism and appetite has recently been reported. The ghrelin receptor (GHSR-1a) is a G-protein coupled receptor which plays a key role in the regulation of food intake. Pharmacological manipulation of the growth hormone secretagogue receptor-type 1a (GHSR-1a) receptor has therefore received a lot of attention as a strategy to combat disorders of appetite and body weight, including age-related malnutrition and the progressive muscle wasting syndrome known as cachexia. In this study, a milk protein-derivative is shown to increase GHSR-1a-mediated intracellular calcium signalling in a concentration-dependent manner in vitro. Significant increases in calcium mobilisation were also observed in a cultured neuronal cell line heterologously expressing the GHS-R1a. In addition, both additive and synergistic effects were observed following co-exposure of GHSR-1a to both the hydrolysate and ghrelin. Subsequent in vivo studies monitored standard chow intake in healthy male and female Sprague-Dawley rats after dosing with the casein hydrolysate (CasHyd). Furthermore, the provision of gastro-protected oral delivery to the bioactive in vivo may aid in the progression of in vitro efficacy to in vivo functionality. In summary, this study reports a ghrelin-stimulating bioactive peptide mixture (CasHyd) with potent effects in vitro. It also provides novel and valuable translational data supporting the potential role of CasHyd as an appetite-enhancing bioactive. Further mechanistic studies are required in order to confirm efficacy as a ghrelinergic bioactive in susceptible population groups.


Asunto(s)
Caseínas/metabolismo , Ingestión de Alimentos , Expresión Génica , Receptores de Ghrelina/genética , Animales , Calcio/metabolismo , Caseínas/química , Línea Celular , Cromatografía Líquida de Alta Presión , Activación Enzimática , Estabilidad de Enzimas , Femenino , Ghrelina/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Masculino , Imagen Molecular/métodos , Ratas , Receptores de Ghrelina/metabolismo
12.
J Nutr ; 147(5): 727-745, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28356427

RESUMEN

The gut harbors an enormous diversity of microbes that are essential for the maintenance of homeostasis in health and disease. A growing body of evidence supports the role of this microbiota in influencing host appetite and food intake. Individual species within the gut microbiota are under selective pressure arising from nutrients available and other bacterial species present. Each bacterial species within the gut aims to increase its own fitness, habitat, and survival via specific fermentation of dietary nutrients and secretion of metabolites, many of which can influence host appetite and eating behavior by directly affecting nutrient sensing and appetite and satiety-regulating systems. These include microbiota-produced neuroactives and short-chain fatty acids. In addition, the gut microbiota is able to manipulate intestinal barrier function, interact with bile acid metabolism, modulate the immune system, and influence host antigen production, thus indirectly affecting eating behavior. A growing body of evidence indicates that there is a crucial role for the microbiota in regulating different aspects of eating-related behavior, as well as behavioral comorbidities of eating and metabolic disorders. The importance of intestinal microbiota composition has now been shown in obesity, anorexia nervosa, and forms of severe acute malnutrition. Understanding the mechanisms in which the gut microbiota can influence host appetite and metabolism will provide a better understanding of conditions wherein appetite is dysregulated, such as obesity and other metabolic or eating disorders, leading to novel biotherapeutic strategies.


Asunto(s)
Apetito , Encéfalo , Ingestión de Alimentos , Trastornos de Alimentación y de la Ingestión de Alimentos , Microbioma Gastrointestinal , Tracto Gastrointestinal/microbiología , Trastornos Nutricionales , Trastornos de Alimentación y de la Ingestión de Alimentos/etiología , Trastornos de Alimentación y de la Ingestión de Alimentos/metabolismo , Tracto Gastrointestinal/metabolismo , Humanos , Trastornos Nutricionales/etiología , Trastornos Nutricionales/metabolismo , Obesidad/etiología , Obesidad/metabolismo
13.
Int J Mol Sci ; 18(2)2017 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-28134808

RESUMEN

Ghrelin is the only known peripherally-derived orexigenic hormone, increasing appetite and subsequent food intake. The ghrelinergic system has therefore received considerable attention as a therapeutic target to reduce appetite in obesity as well as to stimulate food intake in conditions of anorexia, malnutrition and cachexia. As the therapeutic potential of targeting this hormone becomes clearer, it is apparent that its pleiotropic actions span both the central nervous system and peripheral organs. Despite a wealth of research, a therapeutic compound specifically targeting the ghrelin system for appetite modulation remains elusive although some promising effects on metabolic function are emerging. This is due to many factors, ranging from the complexity of the ghrelin receptor (Growth Hormone Secretagogue Receptor, GHSR-1a) internalisation and heterodimerization, to biased ligand interactions and compensatory neuroendocrine outputs. Not least is the ubiquitous expression of the GHSR-1a, which makes it impossible to modulate centrallymediated appetite regulation without encroaching on the various peripheral functions attributable to ghrelin. It is becoming clear that ghrelin's central signalling is critical for its effects on appetite, body weight regulation and incentive salience of food. Improving the ability of ghrelin ligands to penetrate the blood brain barrier would enhance central delivery to GHSR-1a expressing brain regions, particularly within the mesolimbic reward circuitry.


Asunto(s)
Regulación del Apetito , Encéfalo/metabolismo , Ingestión de Alimentos , Mucosa Gástrica/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Ghrelina/farmacocinética , Humanos
14.
Oncologist ; 20(4): 372-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25746344

RESUMEN

BACKGROUND: Knowledge of cancer risk factors is unknown in Ireland. An understanding of risk factors could help inform cancer prevention programs. AIMS AND METHODS: A 48-question online survey was designed to gather data to assess levels of public knowledge about cancer risk factors. RESULTS: There were 748 participants (648 women, 100 men). Mean age was 37 years (range: 18-74 years). For the public, 81% were concerned about developing cancer; however, 20% believed that cancer is unavoidable if a family history exists, 27% believed that >50% of cancers are inherited, and 54% believed that 10%-20% of cancers are inherited; 20% were unaware that risk increases with age. The top five risk factors listed by respondents were smoking (87%), diet (76%), genetics (47%), alcohol (42%), and obesity (33%). Only 32% of the public were aware that obesity is a risk factor, and 33% did not think the location of fat was important. Moreover, 29% and 48% believed that risk could be increased by wearing a tight bra and by a blow to the breast, respectively. In addition, 85% and 86% believed that stress and that mobile phones, respectively, "strongly" increase risk; 12% believed that luck is important in avoiding cancer; 35% thought that "detox" diets could reduce risk; and 61% believed that organic food reduces risk. The majority were aware that physical activity of 30 minutes per day can reduce risk. CONCLUSION: A sizable portion of the population is misinformed about cancer risk. Most participants were aware of classic risk factors (e.g., smoking, diet); however, many overestimated risk attributable to genetics, environment, and stress and underestimated age, obesity, and sunlight. One in seven participants believed that lifetime risk of cancer is not modifiable.


Asunto(s)
Conocimientos, Actitudes y Práctica en Salud , Encuestas Epidemiológicas , Neoplasias/etiología , Neoplasias/prevención & control , Adolescente , Adulto , Anciano , Teléfono Celular , Dieta , Femenino , Alimentos Orgánicos , Predisposición Genética a la Enfermedad , Humanos , Irlanda , Masculino , Persona de Mediana Edad , Actividad Motora , Obesidad/complicaciones , Factores de Riesgo , Fumar/efectos adversos , Adulto Joven
15.
Nutr Neurosci ; 18(2): 49-65, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24621068

RESUMEN

OBJECTIVES: Obesity and obesity-related disorders are reaching epidemic proportions worldwide. In this review, we summarize the accumulating studies that have emerged in the last few decades demonstrating that bioactives from different natural sources could potentially have anti-obesity effects. METHODS: We carried out an extensive search of relevant literature from Pubmed, Web of Knowledge, and other online databases for studies where anti-obesity effects were shown by compounds from natural sources. RESULTS: Appetite suppression, lipid metabolism regulation, and increase of energy expenditure are the main mechanisms by which anti-obesity effects are exerted. Plants represent the most studied natural source of anti-obesity bioactives. Camellia sinensis is the most representative species exerting several anti-obesity effects. Moreover, probiotics (bacteria which bestow health benefit), such as strains of Bifidobacteria and Lactobacillus families, and certain prebiotics (non-viable food components that confers a health benefit on the host associated with modulation of the microbiota effects), such as insulin-type fructans, have also shown capability to combat obesity. Finally, compounds from animal sources, in particular bioactive peptides derived from milk-derived whey and casein protein digestion, high dietary calcium, and omega-3s polyunsaturated fatty acids (n-3 PUFA) present in fish oils, have also shown potential anti-obesity effects. DISCUSSION: Several anti-obesity effects have been observed in different natural bioactives providing an interesting and potentially safer and more desirable treatment strategy for the development of anti-obesity functional or medical foods.


Asunto(s)
Fármacos Antiobesidad/uso terapéutico , Suplementos Dietéticos , Obesidad/prevención & control , Animales , Fármacos Antiobesidad/efectos adversos , Humanos , Obesidad/dietoterapia , Extractos Vegetales/efectos adversos , Extractos Vegetales/uso terapéutico
16.
J Biol Chem ; 288(1): 181-91, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23161547

RESUMEN

G protein-coupled receptors (GPCRs), such as the ghrelin receptor (GHS-R1a), the melanocortin 3 receptor (MC(3)), and the serotonin 2C receptor (5-HT(2C)), are well known for their key role in the homeostatic control of food intake and energy balance. Ghrelin is the only known gut peptide exerting an orexigenic effect and has thus received much attention as an anti-obesity drug target. In addition, recent data have revealed a critical role for ghrelin in dopaminergic mesolimbic circuits involved in food reward signaling. This study investigates the downstream signaling consequences and ligand-mediated co-internalization following heterodimerization of the GHS-R1a receptor with the dopamine 1 receptor, as well as that of the GHS-R1a-MC(3) heterodimer. In addition, a novel heterodimer between the GHS-R1a receptor and the 5-HT(2C) receptor was identified. Interestingly, dimerization of the GHS-R1a receptor with the unedited 5-HT(2C)-INI receptor, but not with the partially edited 5-HT(2C)-VSV isoform, significantly reduced GHS-R1a agonist-mediated calcium influx, which was completely restored following pharmacological blockade of the 5-HT(2C) receptor. These results combined suggest a potential novel mechanism for fine-tuning GHS-R1a receptor-mediated activity via promiscuous dimerization of the GHS-R1a receptor with other G protein-coupled receptors involved in appetite regulation and food reward. These findings may uncover novel mechanisms of significant relevance for the future pharmacological targeting of the GHS-R1a receptor in the homeostatic regulation of energy balance and in hedonic appetite signaling, both of which play a significant role in the development of obesity.


Asunto(s)
Regulación de la Expresión Génica , Ghrelina/metabolismo , Receptores de Ghrelina/metabolismo , Calcio/metabolismo , Dimerización , Ingestión de Alimentos , Conducta Alimentaria , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Homeostasis , Humanos , Ligandos , Microscopía Fluorescente/métodos , Modelos Biológicos , Isoformas de Proteínas , Receptor de Serotonina 5-HT2C/metabolismo , Transducción de Señal
17.
Br J Nutr ; 110(11): 2114-26, 2013 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23731955

RESUMEN

The intake of whey protein isolate (WPI) is known to reduce high-fat diet (HFD)-induced body-weight gain and adiposity. However, the molecular mechanisms are not fully understood. To this end, we fed C57BL/6J mice for 8 weeks with diets containing 10 % energy as fat (low-fat diet, LFD) or 45 % energy as fat (HFD) enriched with either 20 % energy as casein (LFD and HFD) or WPI (high-fat WPI). Metabolic parameters and the hypothalamic and epididymal adipose tissue expression of energy balance-related genes were investigated. The HFD increased fat mass and plasma leptin levels and decreased the dark-phase energy intake, meal number, RER, and metabolic (VO2 and heat) and locomotor activities compared with the LFD. The HFD increased the hypothalamic tissue mRNA expression of the leptin receptor, insulin receptor (INSR) and carnitine palmitoyltransferase 1b (CPT1b). The HFD also reduced the adipose tissue mRNA expression of GLUT4 and INSR. In contrast, WPI reduced fat mass, normalised dark-phase energy intake and increased meal size in HFD-fed mice. The dietary protein did not have an impact on plasma leptin, insulin, glucose or glucagon-like peptide 1 levels, but increased plasma TAG levels in HFD-fed mice. At a cellular level, WPI significantly reduced the HFD-associated increase in the hypothalamic tissue mRNA expression of the leptin receptor, INSR and CPT1b. Also, WPI prevented the HFD-induced reduction in the adipose tissue mRNA expression of INSR and GLUT4. In comparison with casein, the effects of WPI on energy intake and hypothalamic and adipose tissue gene expression may thus represent a state of reduced susceptibility to weight gain on a HFD.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Dieta Alta en Grasa , Ingestión de Energía , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Proteínas de la Leche/uso terapéutico , Sobrepeso/dietoterapia , Adiposidad , Animales , Conducta Animal , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Dieta Alta en Grasa/efectos adversos , Susceptibilidad a Enfermedades , Epidídimo , Conducta Alimentaria , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hipotálamo/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Sobrepeso/etiología , Receptor de Insulina/biosíntesis , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Leptina/biosíntesis , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Proteína de Suero de Leche
18.
J Neuroendocrinol ; 35(9): e13243, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36872624

RESUMEN

Oxytocin is a peptide-hormone extensively studied for its multifaceted biological functions and has recently gained attention for its role in eating behavior, through its action as an anorexigenic neuropeptide. Moreover, the gut microbiota is involved in oxytocinergic signaling through the brain-gut axis, specifically in the regulation of social behavior. The gut microbiota is also implicated in appetite regulation and is postulated to play a role in central regulation of hedonic eating. In this review, we provide an overview on oxytocin and its individual links with the microbiome, the homeostatic and non-homeostatic regulation of eating behavior as well as social behavior and stress.


Asunto(s)
Neuropéptidos , Hormonas Peptídicas , Oxitocina , Conducta Alimentaria/fisiología , Regulación del Apetito , Ingestión de Alimentos/fisiología , Encéfalo/fisiología
19.
Acta Physiol (Oxf) ; 239(3): e14005, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37243893

RESUMEN

AIM: Biological rhythms are endogenously generated natural cycles that act as pacemakers of different physiological mechanisms and homeostasis in the organism, and whose disruption increases metabolic risk. The circadian rhythm is not only reset by light but it is also regulated by behavioral cues such as timing of food intake. This study investigates whether the chronic consumption of a sweet treat before sleeping can disrupt diurnal rhythmicity and metabolism in healthy rats. METHODS: For this, 32 Fischer rats were administered daily a low dose of sugar (160 mg/kg, equivalent to 2.5 g in humans) as a sweet treat at 8:00 a.m. or 8:00 p.m. (ZT0 and ZT12, respectively) for 4 weeks. To elucidate diurnal rhythmicity of clock gene expression and metabolic parameters, animals were sacrificed at different times, including 1, 7, 13, and 19 h after the last sugar dose (ZT1, ZT7, ZT13, and ZT19). RESULTS: Increased body weight gain and higher cardiometabolic risk were observed when sweet treat was administered at the beginning of the resting period. Moreover, central clock and food intake signaling genes varied depending on snack time. Specifically, the hypothalamic expression of Nampt, Bmal1, Rev-erbα, and Cart showed prominent changes in their diurnal expression pattern, highlighting that sweet treat before bedtime disrupts hypothalamic control of energy homeostasis. CONCLUSIONS: These results show that central clock genes and metabolic effects following a low dose of sugar are strongly time-dependent, causing higher circadian metabolic disruption when it is consumed at the beginning of the resting period, that is, with the late-night snack.


Asunto(s)
Ritmo Circadiano , Hipotálamo , Humanos , Ratas , Animales , Ritmo Circadiano/fisiología , Hipotálamo/metabolismo , Sueño , Homeostasis , Azúcares/metabolismo
20.
Mol Metab ; 57: 101427, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34973469

RESUMEN

BACKGROUND: Despite several decades of research, managing body weight remains an unsolved clinical problem. Health problems associated with dysregulated body weight, such as obesity and cachexia, exhibit several gut microbiota alterations. There is an increased interest in utilising the gut microbiota for body weight control, as it responds to intervention and plays an important role in energy extraction from food, as well as biotransformation of nutrients. SCOPE OF THE REVIEW: This review provides an overview of the role of the gut microbiota in the physiological and metabolic alterations observed in two body weight dysregulation-related disorders, namely obesity and cachexia. Second, we assess the available evidence for different strategies, including caloric restriction, intermittent fasting, ketogenic diet, bariatric surgery, probiotics, prebiotics, synbiotics, high-fibre diet, and fermented foods - effects on body weight and gut microbiota composition. This approach was used to give insights into the possible link between body weight control and gut microbiota configuration. MAJOR CONCLUSIONS: Despite extensive associations between body weight and gut microbiota composition, limited success could be achieved in the translation of microbiota-related interventions for body weight control in humans. Manipulation of the gut microbiota alone is insufficient to alter body weight and future research is needed with a combination of strategies to enhance the effects of lifestyle interventions.


Asunto(s)
Microbiota , Probióticos , Simbióticos , Humanos , Obesidad/metabolismo , Prebióticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA