Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Front Endocrinol (Lausanne) ; 14: 1158287, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37234803

RESUMEN

Introduction: Oleoylethanolamide (OEA), an endogenous N-acylethanolamine acting as a gut-to-brain signal to control food intake and metabolism, has been attracting attention as a target for novel therapies against obesity and eating disorders. Numerous observations suggested that the OEA effects might be peripherally mediated, although they involve central pathways including noradrenergic, histaminergic and oxytocinergic systems of the brainstem and the hypothalamus. Whether these pathways are activated directly by OEA or whether they are downstream of afferent nerves is still highly debated. Some early studies suggested vagal afferent fibers as the main route, but our previous observations have contradicted this idea and led us to consider the blood circulation as an alternative way for OEA's central actions. Methods: To test this hypothesis, we first investigated the impact of subdiaphragmatic vagal deafferentation (SDA) on the OEA-induced activation of selected brain nuclei. Then, we analyzed the pattern of OEA distribution in plasma and brain at different time points after intraperitoneal administration in addition to measuring food intake. Results: Confirming and extending our previous findings that subdiaphragmatic vagal afferents are not necessary for the eating-inhibitory effect of exogenous OEA, our present results demonstrate that vagal sensory fibers are also not necessary for the neurochemical effects of OEA. Rather, within a few minutes after intraperitoneal administration, we found an increased concentration of intact OEA in different brain areas, associated with the inhibition of food intake. Conclusion: Our results support that systemic OEA rapidly reaches the brain via the circulation and inhibits eating by acting directly on selected brain nuclei.


Asunto(s)
Encéfalo , Ingestión de Alimentos , Ingestión de Alimentos/fisiología , Encéfalo/metabolismo , Endocannabinoides/farmacología , Endocannabinoides/metabolismo , Ácidos Oléicos/farmacología , Ácidos Oléicos/metabolismo
2.
Physiol Rev ; 103(2): 1423-1485, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36422994

RESUMEN

The cephalic phase insulin response (CPIR) is classically defined as a head receptor-induced early release of insulin during eating that precedes a postabsorptive rise in blood glucose. Here we discuss, first, the various stimuli that elicit the CPIR and the sensory signaling pathways (sensory limb) involved; second, the efferent pathways that control the various endocrine events associated with eating (motor limb); and third, what is known about the central integrative processes linking the sensory and motor limbs. Fourth, in doing so, we identify open questions and problems with respect to the CPIR in general. Specifically, we consider test conditions that allow, or may not allow, the stimulus to reach the potentially relevant taste receptors and to trigger a CPIR. The possible significance of sweetness and palatability as crucial stimulus features and whether conditioning plays a role in the CPIR are also discussed. Moreover, we ponder the utility of the strict classical CPIR definition based on what is known about the effects of vagal motor neuron activation and thereby acetylcholine on the ß-cells, together with the difficulties of the accurate assessment of insulin release. Finally, we weigh the evidence of the physiological and clinical relevance of the cephalic contribution to the release of insulin that occurs during and after a meal. These points are critical for the interpretation of the existing data, and they support a sharper focus on the role of head receptors in the overall insulin response to eating rather than relying solely on the classical CPIR definition.


Asunto(s)
Insulina , Papilas Gustativas , Humanos , Insulina/metabolismo , Gusto/fisiología , Glucemia/metabolismo , Transducción de Señal
3.
Food Funct ; 13(17): 9010-9020, 2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-35942900

RESUMEN

Lipid emulsions (LEs) with tailored digestibility have the potential to modulate satiation or act as delivery systems for lipophilic nutrients and drugs. The digestion of LEs is governed by their interfacial emulsifier layer which determines their gastric structuring and accessibility for lipases. A plethora of LEs that potentially modulate digestion have been proposed in recent years, however, in vivo validations of altered LE digestion remain scarce. Here, we report on the in vivo digestion and satiation of three novel LEs stabilized by whey protein isolate (WPI), thermo-gelling methylcellulose (MC), or cellulose nanocrystals (CNCs) in comparison to an extensively studied surfactant-stabilized LE. LE digestion and satiation were determined in terms of gastric emptying, postprandial plasma hormone and metabolite levels characteristic for lipid digestion, perceived hunger/fullness sensations, and postprandial food intake. No major variations in gastric fat emptying were observed despite distinct gastric structuring of the LEs. The plasma satiation hormone and metabolite response was fastest and highest for WPI-stabilized LEs, indicating a limited capability of proteins to prevent lipolysis due to fast hydrolysis under gastric conditions and displacement by lipases. MC-stabilized LEs show a similar gastric structuring as surfactant-stabilized LEs but slightly reduced hormone and metabolite responses, suggesting that thermo-gelling MC prevents lipase adsorption more effectively. Ultimately, CNC-stabilized LEs showed a drastic reduction (>70%) in plasma hormone and metabolite responses. This confirms the efficiency of particle (Pickering) stabilized LEs to prevent lipolysis proposed in literature based on in vitro experiments. Subjects reported more hunger and less fullness after consumption of LEs stabilized with MC and CNCs which were able to limit satiation responses. We do not find evidence for the widely postulated ileal brake, i.e. that delivery of undigested nutrients to the ileum triggers increased satiation. On the contrary, we find decreased satiation for LEs that are able to delay lipolysis. No differences in food intake were observed 5 h after LE consumption. In conclusion, LE interfacial design modulates in vivo digestion and satiation response in humans. In particular, Pickering LEs show extraordinary capability to prevent lipolysis and qualify as oral delivery systems for lipophilic nutrients and drugs.


Asunto(s)
Digestión , Lípidos , Celulosa/química , Emulsiones/química , Hormonas , Humanos , Lipasa/metabolismo , Lípidos/química , Saciedad , Tensoactivos/farmacología
4.
Diabetes Obes Metab ; 24(2): 268-280, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34658116

RESUMEN

Peripheral glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK) are secreted from enteroendocrine cells, and their plasma concentrations increase in response to eating. While the satiating effect of gut-derived CCK on food-intake control is well documented, the effect of peripheral GLP-1 is less clear. There is evidence that native GLP-1 can inhibit food intake only in the fed state but not in the fasting state. We therefore hypothesized that other gut peptides released during a meal might influence the subsequent effect of endogenous GLP-1 and investigated whether CCK could do so. We found that intraperitoneal injection of CCK in food-restricted mice inhibited food intake during the first 30-minute segment of a 1-hour session of ad libitum chow intake and that mice compensated by increasing their intake during the second half of the session. Importantly, this compensatory behaviour was abolished by an intraperitoneal injection of GLP-1 administered following an intraperitoneal injection of CCK and prior to the 1-hour session. In vivo activation of the free fatty acid 1 (FFA1) receptor with orally administered TAK875 increased plasma CCK concentration and, consistent with the effect of exogenous CCK, we found that prior oral administration of TAK875 increased the eating inhibitory effect of peripherally administered GLP-1. To examine the role of the vagus nerve in this effect, we utilized a saporin-based lesioning procedure to selectively ablate the CCK receptor-expressing gastrointestinal vagal afferent neurones (VANs). We found that the combined anorectic effect of TAK875 and GLP-1 was significantly attenuated in the absence of CCK receptor expressing VANs. Taken together, our results indicate that endogenous CCK interacts with GLP-1 to promote satiation and that activation of the FFA1 receptor can initiate this interaction by stimulating the release of CCK.


Asunto(s)
Colecistoquinina , Péptido 1 Similar al Glucagón , Animales , Ingestión de Alimentos , Humanos , Ratones , Receptores de Colecistoquinina , Saciedad/fisiología , Nervio Vago/fisiología
5.
Physiol Rev ; 102(2): 689-813, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486393

RESUMEN

During the past 30 yr, investigating the physiology of eating behaviors has generated a truly vast literature. This is fueled in part by a dramatic increase in obesity and its comorbidities that has coincided with an ever increasing sophistication of genetically based manipulations. These techniques have produced results with a remarkable degree of cell specificity, particularly at the cell signaling level, and have played a lead role in advancing the field. However, putting these findings into a brain-wide context that connects physiological signals and neurons to behavior and somatic physiology requires a thorough consideration of neuronal connections: a field that has also seen an extraordinary technological revolution. Our goal is to present a comprehensive and balanced assessment of how physiological signals associated with energy homeostasis interact at many brain levels to control eating behaviors. A major theme is that these signals engage sets of interacting neural networks throughout the brain that are defined by specific neural connections. We begin by discussing some fundamental concepts, including ones that still engender vigorous debate, that provide the necessary frameworks for understanding how the brain controls meal initiation and termination. These include key word definitions, ATP availability as the pivotal regulated variable in energy homeostasis, neuropeptide signaling, homeostatic and hedonic eating, and meal structure. Within this context, we discuss network models of how key regions in the endbrain (or telencephalon), hypothalamus, hindbrain, medulla, vagus nerve, and spinal cord work together with the gastrointestinal tract to enable the complex motor events that permit animals to eat in diverse situations.


Asunto(s)
Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Animales , Homeostasis/fisiología , Humanos , Transducción de Señal/fisiología
6.
Mol Metab ; 51: 101237, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33878401

RESUMEN

BACKGROUND: A chronic imbalance of energy intake and energy expenditure results in excess fat storage. The obesity often caused by this overweight is detrimental to the health of millions of people. Understanding both sides of the energy balance equation and their counter-regulatory mechanisms is critical to the development of effective therapies to treat this epidemic. SCOPE OF REVIEW: Behaviors surrounding ingestion have been reviewed extensively. This review focuses more specifically on energy expenditure regarding bodyweight control, with a particular emphasis on the organs and attractive metabolic processes known to reduce bodyweight. Moreover, previous and current attempts at anti-obesity strategies focusing on energy expenditure are highlighted. Precise measurements of energy expenditure, which consist of cellular, animal, and human models, as well as measurements of their translatability, are required to provide the most effective therapies. MAJOR CONCLUSIONS: A precise understanding of the components surrounding energy expenditure, including tailored approaches based on genetic, biomarker, or physical characteristics, must be integrated into future anti-obesity treatments. Further comprehensive investigations are required to define suitable treatments, especially because the complex nature of the human perspective remains poorly understood.


Asunto(s)
Ingestión de Energía , Metabolismo Energético/fisiología , Obesidad/terapia , Animales , Modelos Animales de Enfermedad , Humanos , Obesidad/metabolismo , Obesidad/fisiopatología
7.
Int J Behav Med ; 28(5): 641-646, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33721233

RESUMEN

BACKGROUND: Weight gain is common as women approach mid-life. Reduced levels of leptin, an anorexigenic hormone, may facilitate this. Studies in middle-aged women with obesity have shown that dysfunctional eating behaviour, such as restrained eating, is linked to lower leptin. Furthermore, states of low oestradiol signalling, as are found in post-menopause or anorexia nervosa, have been found to impact leptin levels. The aim of this study was to investigate, for the first time, how different aspects of dysfunctional eating, menopausal status, and a history of anorexia nervosa relate to leptin levels in normal-weight middle-aged women. METHODS: A total of N = 57 women were recruited. Thirty-one were post-menopausal, and 27 had a history of anorexia nervosa. Dysfunctional eating behaviour was measured by the Three-Factor Eating Questionnaire, which contains three subscales: susceptibility/responsiveness to hunger, restraint, and disinhibition. Body composition was assessed by bioelectrical impedance analysis. A fasting blood sample was obtained to determine leptin. RESULTS: Controlling for age, body mass index, and fat mass, susceptibility/responsiveness to hunger was positively associated with leptin (ß = 0.267, p = 0.031), whereas restrained eating (ß = - 0.183, p = 0.079) and a history of anorexia nervosa (ß = - 0.221, p = 0.059) were, by trend, negatively associated with leptin. Neither disinhibited eating nor menopausal status was related to leptin. CONCLUSIONS: Leptin may decline as a response to repeated states of a negative energy balance. A possible implication is that mid-life weight management should avoid extreme changes in eating behaviour and instead focus on the macronutrient composition of diet and physical activity. Further, longitudinal enquiries are warranted to investigate these relationships.

8.
Nat Metab ; 3(2): 258-273, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33589843

RESUMEN

The anorexigenic peptide glucagon-like peptide-1 (GLP-1) is secreted from gut enteroendocrine cells and brain preproglucagon (PPG) neurons, which, respectively, define the peripheral and central GLP-1 systems. PPG neurons in the nucleus tractus solitarii (NTS) are widely assumed to link the peripheral and central GLP-1 systems in a unified gut-brain satiation circuit. However, direct evidence for this hypothesis is lacking, and the necessary circuitry remains to be demonstrated. Here we show that PPGNTS neurons encode satiation in mice, consistent with vagal signalling of gastrointestinal distension. However, PPGNTS neurons predominantly receive vagal input from oxytocin-receptor-expressing vagal neurons, rather than those expressing GLP-1 receptors. PPGNTS neurons are not necessary for eating suppression by GLP-1 receptor agonists, and concurrent PPGNTS neuron activation suppresses eating more potently than semaglutide alone. We conclude that central and peripheral GLP-1 systems suppress eating via independent gut-brain circuits, providing a rationale for pharmacological activation of PPGNTS neurons in combination with GLP-1 receptor agonists as an obesity treatment strategy.


Asunto(s)
Sistema Nervioso Central/fisiología , Péptido 1 Similar al Glucagón/fisiología , Sistema Nervioso Periférico/fisiología , Respuesta de Saciedad/fisiología , Animales , Ingestión de Alimentos , Femenino , Tracto Gastrointestinal/inervación , Tracto Gastrointestinal/fisiología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Péptidos Similares al Glucagón/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Proglucagón/metabolismo , Receptores de Oxitocina/metabolismo , Nervio Vago/fisiología
9.
eNeuro ; 8(1)2021.
Artículo en Inglés | MEDLINE | ID: mdl-33495245

RESUMEN

Vagal and spinal sensory endings in the wall of the hepatic portal and superior mesenteric veins (PMV) provide the brain with chemosensory information important for energy balance and other functions. To determine their medullary neuronal targets, we injected the transsynaptic anterograde viral tracer HSV-1 H129-772 (H129) into the PMV wall or left nodose ganglion (LNG) of male rats, followed by immunohistochemistry (IHC) and high-resolution imaging. We also determined the chemical phenotype of H129-infected neurons, and potential vagal and spinal axon terminal appositions in the dorsal motor nucleus of the vagus (DMX) and the nucleus of the solitary tract (NTS). PMV wall injections generated H129-infected neurons in both nodose ganglia and in thoracic dorsal root ganglia (DRGs). In the medulla, cholinergic preganglionic parasympathetic neurons in the DMX were virtually the only targets of chemosensory information from the PMV wall. H129-infected terminal appositions were identified on H129-infected somata and dendrites in the DMX, and on H129-infected DMX dendrites that extend into the NTS. Sensory transmission via vagal and possibly spinal routes from the PMV wall therefore reaches DMX neurons via axo-somatic appositions in the DMX and axo-dendritic appositions in the NTS. However, the dearth of H129-infected NTS neurons indicates that sensory information from the PMV wall terminates on DMX neurons without engaging NTS neurons. These previously underappreciated direct sensory routes into the DMX enable a vago-vagal and possibly spino-vagal reflexes that can directly influence visceral function.


Asunto(s)
Venas Mesentéricas , Ganglio Nudoso , Animales , Masculino , Neuronas , Ratas , Núcleo Solitario , Nervio Vago
10.
11.
Nat Commun ; 11(1): 2080, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32350271

RESUMEN

Excessive insulin signaling through the insulin receptor (IR) may play a role in the pathogenesis of diet-induced metabolic disease, including obesity and type 2 diabetes. Here we investigate whether heterozygous impairment of insulin receptor (IR) expression limited to peripheral, i.e. non-CNS, tissues of adult mice impacts the development of high-fat diet-induced metabolic deterioration. While exhibiting some features of insulin resistance, PerIRKO+/- mice display a hepatic energy deficit accompanied by induction of energy-sensing AMPK, mitochondrial biogenesis, PPARα, unexpectedly leading to protection from, and reversal of hepatic lipid accumulation (steatosis hepatis, NAFLD). Consistently, and unlike in control mice, the PPARα activator fenofibrate fails to further affect hepatic lipid accumulation in PerIRKO+/- mice. Taken together, and opposing previously established diabetogenic features of insulin resistance, incomplete impairment of insulin signaling may mimic central aspects of calorie restriction to limit hepatic lipid accumulation during conditions of metabolic stress.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Ayuno/metabolismo , Hígado Graso/etiología , Hígado Graso/prevención & control , Receptor de Insulina/metabolismo , Animales , Composición Corporal , Metabolismo Energético , Conducta Alimentaria , Glucosa/metabolismo , Homeostasis , Resistencia a la Insulina , Hígado/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Cell Rep ; 30(6): 2028-2039.e4, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32049029

RESUMEN

The vagus nerve conveys gastrointestinal cues to the brain to control eating behavior. In obesity, vagally mediated gut-brain signaling is disrupted. Here, we show that the cocaine- and amphetamine-regulated transcript (CART) is a neuropeptide synthesized proportional to the food consumed in vagal afferent neurons (VANs) of chow-fed rats. CART injection into the nucleus tractus solitarii (NTS), the site of vagal afferent central termination, reduces food intake. Conversely, blocking endogenous CART action in the NTS increases food intake in chow-fed rats, and this requires intact VANs. Viral-mediated Cartpt knockdown in VANs increases weight gain and daily food intake via larger meals and faster ingestion rate. In obese rats fed a high-fat, high-sugar diet, meal-induced CART synthesis in VANs is blunted and CART antibody fails to increase food intake. However, CART injection into the NTS retains its anorexigenic effect in obese rats. Restoring disrupted VAN CART signaling in obesity could be a promising therapeutic approach.


Asunto(s)
Hiperfagia/genética , Proteínas del Tejido Nervioso/metabolismo , Nervio Vago/efectos de los fármacos , Aumento de Peso/genética , Animales , Humanos , Masculino , Ratas
13.
Front Nutr ; 6: 170, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31781572

RESUMEN

A better understanding of how dietary lipids are processed by the human body is necessary to allow for the control of satiation and energy intake by tailored lipid systems. To examine whether rats are a valid model of human dietary lipid processing and therefore useful for further mechanistic studies in this context, we tested in rats three lipid emulsions of different stability, which alter satiety responses in humans. Different sets of 15 adult male Sprague Dawley rats, equipped with gastric catheters alone or combined with hepatic portal vein (HPV) and vena cava (VC) catheters were maintained on a medium-fat diet and adapted to an 8 h deprivation/16 h feeding schedule. Experiments were performed in a randomized cross-over study design. After gastric infusion of the lipid emulsions, we assessed gastric emptying by the paracetamol absorption test and recorded in separate experiments food intake and plasma levels of gastrointestinal hormones and metabolites in the HPV. For an acid stable emulsion, slower gastric emptying and an enhanced release of satiating gastrointestinal (GI) hormones were observed and were associated with lower short-term energy intake in rats and less hunger in humans, respectively. The magnitude of hormonal responses was related to the acid stability and redispersibility of the emulsions and thus seems to depend on the availability of lipids for digestion. Plasma metabolite levels were unaffected by the emulsion induced changes in lipolysis. The results support that structured lipid systems are digested similarly in rats and humans. Thus unstable emulsions undergo the same intragastric destabilization in both species, i.e., increased droplet size and creaming. This work establishes the rat as a viable animal model for in vivo studies on the control of satiation and energy intake by tailored lipid systems.

15.
J Affect Disord ; 252: 404-412, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31003109

RESUMEN

BACKGROUND: There is growing evidence for a role of abnormal gut-brain signaling in disorders involving altered mood and affect, including depression. Studies using vagus nerve stimulation (VNS) suggest that the disruption of vagal afferent signaling may contribute to these abnormalities. To test this hypothesis, we used a rat model of subdiaphragmatic vagal deafferentation (SDA), the most complete and selective vagal deafferentation method existing to date, to study the consequences of complete disconnection of abdominal vagal afferents on affective behaviors. METHODS: SDA- and Sham-operated male rats were subjected to several tests that are commonly used in preclinical rodent models to assess the presence of anhedonic behavior, namely the novel object-induced exploration test, the novelty-suppressed eating test, and the sucrose preference test. In addition, we compared SDA and Sham rats in a social interaction test and the forced swim test to assess sociability and behavioral despair, respectively. RESULTS: Compared to Sham controls, SDA rats consistently displayed signs of anhedonic behavior in all test settings used. SDA rats also showed increased immobility and reduced swimming in the forced swim test, whereas they did not differ from Sham controls with regards to social approach behavior. LIMITATIONS: This study was conducted in male rats only. Hence, possible sex-specific effects of SDA on affective behaviors remained unexamined. CONCLUSIONS: Our findings demonstrate that hedonic behavior and behavioral despair are subject to visceral modulation through abdominal vagal afferents. These data are compatible with preclinical models and clinical trials showing beneficial effects of VNS on depression-like and affective behaviors.


Asunto(s)
Afecto , Vías Aferentes , Trastornos del Humor/terapia , Estimulación del Nervio Vago , Nervio Vago/fisiología , Abdomen/inervación , Animales , Modelos Animales de Enfermedad , Masculino , Trastornos del Humor/fisiopatología , Ratas , Ratas Sprague-Dawley , Natación
16.
Mol Metab ; 23: 51-59, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30905616

RESUMEN

OBJECTIVE: Roux-en-Y gastric bypass (RYGB) surgery produces rapid and persistent reductions in plasma triglyceride (TG) levels associated with fewer cardiovascular events. The mechanisms of the reduction in systemic TG levels remain unclear. We hypothesized that RYGB reduces intestinal TG secretion via altered enterocyte lipid handling. METHODS: RYGB or Sham surgery was performed in diet-induced obese, insulin-resistant male Sprague-Dawley rats. First, we tested whether RYGB reduced test meal-induced TG levels in the intestinal lymph, a direct readout of enterocyte lipid secretion. Second, we examined whether RYGB modified TG enterocyte secretion at the single lipid level and in comparison to other lipid subclasses, applying mass spectrometry lipidomics to the intestinal lymph of RYGB and Sham rats (0-21 days after surgery). Third, we explored whether RYGB modulated the metabolic characteristics of primary enterocytes using transcriptional and functional assays relevant to TG absorption, reesterification, storage in lipid droplets, and oxidation. RESULTS: RYGB reduced overall postprandial TG concentrations compared to Sham surgery in plasma and intestinal lymph similarly. RYGB reduced lymphatic TG concentrations more than other lipid subclasses, and shifted the remaining TG pool towards long-chain, unsaturated species. In enterocytes of fasted RYGB rats, lipid uptake was transcriptionally (Fatp4, Fabp2, Cd36) and functionally reduced compared to Sham, whereas TG reesterification genes were upregulated. CONCLUSION: Our results show that RYGB substantially reduces intestinal TG secretion and modifies enterocyte lipid absorption and handling in rats. These changes likely contribute to the improvements in the plasma TG profile observed after RYGB in humans.


Asunto(s)
Enterocitos/metabolismo , Derivación Gástrica/métodos , Periodo Posprandial/fisiología , Triglicéridos/sangre , Animales , Glucemia , Diglicéridos/metabolismo , Modelos Animales de Enfermedad , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Mucosa Intestinal/metabolismo , Linfa/metabolismo , Masculino , Obesidad/cirugía , Ratas , Ratas Sprague-Dawley
17.
Physiol Behav ; 204: 76-85, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30753847

RESUMEN

I was flattered and felt tremendously honored to receive the 2018 Distinguished Career Award (DCA) from SSIB, the society that I always considered my scientific home, my family. Preparing the award lecture, I reflected about defining features of my career. This paper summarizes this very personal retrospective. As you will read, serendipity and more or less spontaneous decisions; i.e., some luck to be in the right place at the right time, and spontaneity to grab an opportunity when it presented itself, played a major role, and not necessarily a thorough analysis of my life situation at various junctions of my career path. Luck also often had the name of a fantastic tutor or mentor, or came in the form of enlightening discussions with a friend. Science is teamwork, which emphasizes how important collaborators, post-docs, students and technicians are. Although deep thinking was not necessarily crucial for my career path, a thorough examination is of course necessary when analyzing data, which were often most important when they did not confirm my hypothesis. Science is also hard work considering how much time one spends, but it never seemed like work to me because I had always this desire to find out how things in the organism work, and I always felt privileged to be able to pursue my "hobby" and even get a decent pay for it. In short, being a scientist is probably one of the most rewarding professional activities that life can offer.


Asunto(s)
Neurofisiología/historia , Animales , Ingestión de Alimentos/fisiología , Ingestión de Alimentos/psicología , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Medicina Veterinaria/historia
18.
J Neuroendocrinol ; 31(7): e12661, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30447166

RESUMEN

Middle age has been linked with various dysfunctional eating patterns in women. The hormone ghrelin is related to food intake, with plasma levels rising before eating and decreasing immediately afterwards. Animal research has shown that oestradiol is an antagonist of ghrelin. Given that both menopause and anorexia nervosa (AN) are states characterised by reduced oestradiol, the present study aimed to investigate for the first time whether menopausal status and a history of AN are linked with altered ghrelin levels in middle-aged women. Based on previous research, we hypothesised that (i) post-menopausal women would demonstrate comparably increased ghrelin after food intake and (ii) women with a history of AN would exhibit increased total ghrelin levels. Healthy, middle-aged women (n = 57) were recruited. Of the women, 31 were post-menopausal and 27 had a history of AN. Plasma ghrelin was repeatedly collected before and after a meal standardised in terms of caloric content. Areas under the curves were calculated to indicate total (AUCg) and postprandial ghrelin (AUCi). Menopausal status was linked with postprandial ghrelin (AUCi -1.6 ± 2.2 vs -2.9 ± 2.6; P = 0.058), whereas a history of AN was linked with total ghrelin (AUCg 36.2 ± 5.6 vs 39.0 ± 3.7; P = 0.050). There were no interaction effects (both P > 0.466). A closer examination of the effects revealed that post-menopausal women showed marginally greater decreases in ghrelin immediately after food intake (P = 0.064) and marginally greater re-increases after 60 minutes (P = 0.084) compared to pre-menopausal women. Women with a history of AN had significantly higher total ghrelin compared to women without a history of AN (P = 0.042). Post-menopause was linked with higher sensitivity of ghrelin to food intake (trend), whereas a history of AN was related to greater total ghrelin. Future research should investigate to what extent the observed alterations in ghrelin may affect dysfunctional eating behaviour during middle age.


Asunto(s)
Anorexia Nerviosa/sangre , Ghrelina/sangre , Menopausia/sangre , Ingestión de Alimentos/fisiología , Femenino , Humanos , Persona de Mediana Edad , Periodo Posprandial
19.
JCI Insight ; 3(14)2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30046010

RESUMEN

Aldosterone synthase inhibitors (ASIs) should alleviate obesity-related cardiovascular and renal problems resulting partly from aldosterone excess, but their clinical use may have limitations. To improve knowledge for the use of ASIs, we investigated physiology in aldosterone synthase-knockout (ASKO) mice. On regular chow diet (CD), ASKO mice ate more and weighed less than WT mice, largely because they hyperventilated to eliminate acid as CO2. Replacing CD with high-fat diet (HFD) lessened the respiratory burden in ASKO mice, as did 12- to 15-hour fasting. The latter eliminated the genotype differences in respiratory workload and energy expenditure (EE). Thus, aldosterone deficiency burdened the organism more when the animals ate carbohydrate-rich chow than when they ate a HFD. Chronic HFD exposure further promoted hyperinsulinemia in ASKO mice that contributed to visceral fat accumulation accompanied by reduced lipolysis, thermogenic reprogramming, and the absence of weight-gain-related EE increases. Intracerebroventricular aldosterone supplementation in ASKO mice attenuated the HFD-induced hyperinsulinemia, but did not affect EE, suggesting that the presence of aldosterone increased the body's energetic efficiency, thus counteracting the EE-increasing effect of low insulin. ASIs may therefore cause acid-overload-induced respiratory burden and promote obesity. Their use in patients with preexisting renal and cardiopulmonary diseases might be contraindicated.


Asunto(s)
Aldosterona/metabolismo , Citocromo P-450 CYP11B2/genética , Hiperinsulinismo/genética , Obesidad/genética , Adipocitos/citología , Animales , Citocromo P-450 CYP11B2/efectos de los fármacos , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Metabolismo Energético , Heces , Femenino , Hipoaldosteronismo/genética , Insulina/sangre , Resistencia a la Insulina/genética , Lipólisis , Locomoción , Masculino , Ratones , Ratones Noqueados , Pletismografía , Termogénesis
20.
Sci Rep ; 8(1): 10818, 2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018405

RESUMEN

Studies indicate that modulating enterocyte metabolism might affect whole body glucose homeostasis and the development of diet-induced obesity (DIO). We tested whether enhancing enterocyte fatty acid oxidation (FAO) could protect mice from DIO and impaired glycemic control. To this end, we used mice expressing a mutant form of carnitine palmitoyltransferase-1a (CPT1mt), insensitive to inhibition by malonyl-CoA, in their enterocytes (iCPT1mt) and fed them low-fat control diet (CD) or high-fat diet (HFD) chronically. CPT1mt expression led to an upregulation of FAO in the enterocytes. On CD, iCPT1mt mice had impaired glycemic control and showed concomitant activation of lipogenesis, glycolysis and gluconeogenesis in their enterocytes. On HFD, both iCPT1mt and control mice developed DIO, but iCPT1mt mice showed improved glycemic control and reduced visceral fat mass. Together these data indicate that modulating enterocyte metabolism in iCPT1mt mice affects glycemic control in a body weight-independent, but dietary fat-dependent manner.


Asunto(s)
Dieta Alta en Grasa , Ácidos Grasos/química , Peroxidación de Lípido , Animales , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Células Cultivadas , Duodeno/patología , Enterocitos/citología , Enterocitos/metabolismo , Prueba de Tolerancia a la Glucosa , Glucólisis , Lipogénesis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Obesidad/patología , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...