Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Biochimie ; 176: 110-116, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32623049

ABSTRACT

Obesity is a major health problem worldwide. Overweight and obesity directly affect health-related quality of life and also have an important economic impact on healthcare systems. In experimental models, obesity leads to hypothalamic inflammation and loss of metabolic homeostasis. It is known that macroautophagy is decreased in the hypothalamus of obese mice but the role of chaperone-mediated autophagy is still unknown. In this study, we aimed to investigate the role of hypothalamic chaperone-mediated autophagy in response to high-fat diet and also the direct effect of palmitate on hypothalamic neurons. Mice received chow or high-fat diet for 3 days or 1 week. At the end of the experimental protocol, chaperone-mediated autophagy in hypothalamus was investigated, as well as cytokines expression. In other set of experiments, neuronal cell lines were treated with palmitic acid, a saturated fatty acid. We show that chaperone-mediated autophagy is differently regulated in response to high-fat diet intake for 3 days or 1 week. Also, when hypothalamic neurons are directly exposed to palmitate there is activation of chaperone-mediated autophagy. High-fat diet causes hypothalamic inflammation concomitantly to changes in the content of chaperone-mediated autophagy machinery. It remains to be studied the direct role of inflammation and lipids itself on the activation of chaperone-mediated autophagy in the hypothalamus in vivo and also the neuronal implications of chaperone-mediated autophagy inhibition in response to obesity.


Subject(s)
Chaperone-Mediated Autophagy/drug effects , Diet, High-Fat/adverse effects , Hypothalamus/metabolism , Neurons/metabolism , Obesity/metabolism , Palmitic Acid/pharmacology , Animals , Cell Line , Hypothalamus/pathology , Mice , Neurons/pathology , Obesity/chemically induced , Obesity/pathology , Palmitic Acid/metabolism
2.
Diabetes Metab ; 46(2): 144-149, 2020 04.
Article in English | MEDLINE | ID: mdl-31212089

ABSTRACT

AIM: In the D.E.S.I.R. cohort, higher consumption of dairy products was associated with lower incidence of hyperglycaemia, and dihydroceramide concentrations were higher in those who progressed to diabetes. Our aim here was to study the relationships between dairy consumption and concentrations of dihydroceramides and ceramides. METHODS: In the D.E.S.I.R. cohort, men and women aged 30-65 years, volunteers from West-Central France, were included in a 9-year follow-up with examinations every 3 years, including food-frequency questionnaires. Two items concerned dairy products (cheese, other dairy products except cheese). At each examination, dihydroceramides and ceramides were determined by mass spectrometry in a cohort subset; in the present study, the 105 people who did not progress to type 2 diabetes were analyzed, as the disorder per se might be a confounding factor. RESULTS: Higher consumption of dairy products (except cheese) was associated with total plasma dihydroceramides during the follow-up, but only in women (P=0.01 for gender interaction). In fact, dihydroceramide levels were lower in women with high vs low consumption (P=0.03), and were significantly increased during follow-up (P=0.01) in low consumers only. There was also a trend for lower ceramides in women with high dairy (except cheese) intakes (P=0.08). Cheese was associated with dihydroceramide and ceramide changes during follow-up (P=0.04 for both), but no clear trend was evident in either low or high consumers. CONCLUSION: These results show that, in women, there is an inverse association between fresh dairy product consumption and predictive markers (dihydroceramides) of type 2 diabetes.


Subject(s)
Ceramides/blood , Dairy Products , Diabetes Mellitus, Type 2/epidemiology , Diet , Adult , Aged , Diabetes Mellitus, Type 2/blood , Female , Humans , Incidence , Longitudinal Studies , Middle Aged
3.
Mol Neurobiol ; 56(6): 3958-3971, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30238388

ABSTRACT

Cystathionine beta synthase (CBS) is one of the 225 genes on chromosome 21 (HSA 21) that are triplicated in persons with trisomy 21 (Down syndrome). Although most triplicate HSA21 genes have their orthologous genes on murine chromosome 16, the murine ortholog of hCBS is on murine chromosome 17 and thus is not present in the well-studied Ts65Dn mouse model of trisomy 21. Persons with trisomy 21 (T21) present deficits in neurotransmission and exhibit early brain aging that can partially be explained by monoamine neurotransmitter alterations. We used transgenic mice for the hCBS gene, which overexpress the CBS protein in various brain regions, to study if CBS overexpression induces modifications in the monoamine neurotransmitters in the hypothalamus, thalamus, hippocampus, and striatum from transgenic and control female and male mice aged 3-4 months and 11-12 months. Sex, age, and brain area each influenced neurotransmitter levels. Briefly, the serotonin pathway was modified by CBS overexpression in various brain areas in female mice but not in male mice. The dopamine pathway was modified in brain regions according to sex and age. These results may allow us to better understand the role of the transsulfuration pathway and especially CBS overexpression in the metabolism of biogenic amines and the catecholamine catabolism in persons with trisomy 21.


Subject(s)
Brain/metabolism , Cystathionine beta-Synthase/metabolism , Dopamine/metabolism , Serotonin/metabolism , Animals , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Statistics, Nonparametric
5.
Free Radic Biol Med ; 114: 122-130, 2018 01.
Article in English | MEDLINE | ID: mdl-28958596

ABSTRACT

Down syndrome (DS) is caused by the trisomy of human chromosome 21 and is the most common genetic cause of intellectual disability. In addition to the intellectual deficiencies and physical anomalies, DS individuals present a higher prevalence of obesity and subsequent metabolic disorders than healthy adults. There is increasing evidence from both clinical and experimental studies indicating the association of visceral obesity with a pro-inflammatory status and recent studies have reported that obese people with DS suffer from low-grade systemic inflammation. However, the link between adiposity and inflammation has not been explored in DS. Here we used Ts65Dn mice, a validated DS mouse model, for the study of obesity-related inflammatory markers. Ts65Dn mice presented increased energy intake, and a positive energy balance leading to increased adiposity (fat mass per body weight), but did not show overweight, which only was apparent upon high fat diet induced obesity. Trisomic mice also had fasting hyperglycemia and hypoinsulinemia, and normal incretin levels. Those trisomy-associated changes were accompanied by reduced ghrelin plasma levels and slightly but not significantly increased leptin levels. Upon a glucose load, Ts65Dn mice showed normal increase of incretins accompanied by over-responses of leptin and resistin, while maintaining the hyperglycemic and hypoinsulinemic phenotype. These changes in the adipoinsular axis were accompanied by increased plasma levels of inflammatory biomarkers previously correlated with obesity galectin-3 and HSP72, and reduced IL-6. Taken together, these results suggest that increased adiposity, and pro-inflammatory adipokines leading to low-grade inflammation are important players in the propensity to obesity in DS. We conclude that DS would be a case of impaired metabolic-inflammatory axis.


Subject(s)
Disease Models, Animal , Down Syndrome/complications , Inflammation Mediators/blood , Obesity/etiology , Animals , Down Syndrome/blood , Down Syndrome/pathology , Mice , Obesity/blood , Obesity/pathology , Risk Factors
6.
Obes Surg ; 27(3): 630-640, 2017 03.
Article in English | MEDLINE | ID: mdl-27448233

ABSTRACT

BACKGROUND: Laparoscopic adjustable gastric banding (LAGB) remains one of the most performed bariatric procedures worldwide, but a few long-term studies have been reported often with limited data at time of longest follow-up. We review our 18-year LAGB experience with special regard to weight loss failure and long-term complications leading to band removal. METHODS: We performed 897 LAGB procedures from April 1996 to December 2007: 376 using the perigastric dissection and 521 using the pars flaccida dissection. We performed a retrospective analysis of the data of this consecutive series. Failure was defined as band removal with or without conversion to another procedure or excess weight loss (EWL%) <25 %. RESULTS: There were 120 men and 770 women. Mean age was 39.5 years, and mean BMI was 45.6 kg/m2. Mean follow-up was 14.6 years (range 101-228 months) with 90 % follow-up beyond 10 years. Ten (1.1 %) had early complications and 504 (56 %) late complications. Overall, 374 (41.6 %) bands were explanted for complications, weight regain, or intolerance. Mean 15-year EWL% in patients with band in place was 41.73 %. Over time, band failure rate increases from 18.4 % at 2 years to 43 % at 10 years and more than 70 % beyond 15 years. CONCLUSIONS: Despite good initial results, late complications, weight regain, and intolerance lead to band removal in nearly half of the patients over time. However, given that there is no good information on alternative procedures in the long term and considering its reversibility and safety still has a place in the treatment of morbid obesity for informed and motivated patients.


Subject(s)
Gastroplasty , Obesity, Morbid/surgery , Adult , Female , Follow-Up Studies , Gastroplasty/methods , Gastroplasty/rehabilitation , Hospitals, University , Humans , Laparoscopy/methods , Male , Middle Aged , Obesity, Morbid/rehabilitation , Reoperation , Retrospective Studies , Time Factors , Treatment Outcome , Weight Loss
7.
Diabetes Obes Metab ; 16 Suppl 1: 49-55, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25200296

ABSTRACT

Fatty acid (FA)-sensitive neurons are present in the brain, especially the hypothalamus, and play a key role in the neural control of energy homeostasis. Through neuronal output, FA may modulate feeding behaviour as well as insulin secretion and action. Subpopulations of neurons in the ventromedial and arcuate hypothalamic nuclei are selectively either inhibited or activated by FA. Molecular effectors of these FA effects probably include chloride or potassium ion channels. While intracellular metabolism and activation of the ATP-sensitive K⁺ channel appear to be necessary for some of the signalling effects of FA, at least half of the FA responses in ventromedial hypothalamic neurons are mediated by interaction with FAT/CD36, an FA transporter/receptor that does not require intracellular metabolism to activate downstream signalling. Thus, FA or their metabolites can modulate neuronal activity as a means of directly monitoring ongoing fuel availability by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. Recently, the role of lipoprotein lipase in FA sensing has also been shown in animal models not only in hypothalamus, but also in hippocampus and striatum. Finally, FA overload might impair neural control of energy homeostasis through enhanced ceramide synthesis and may contribute to obesity and/or type 2 diabetes pathogenesis in predisposed subjects.


Subject(s)
CD36 Antigens/metabolism , Fatty Acids, Nonesterified/metabolism , Feedback, Physiological , Lipid Metabolism , Models, Neurological , Neurons/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Appetite Regulation , Corpus Striatum/cytology , Corpus Striatum/metabolism , Fatty Acids, Nonesterified/blood , Hippocampus/cytology , Hippocampus/metabolism , Humans , Lipoprotein Lipase/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Organ Specificity , Ventromedial Hypothalamic Nucleus/cytology
8.
Mol Psychiatry ; 19(10): 1095-105, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24732670

ABSTRACT

Circulating triglycerides (TGs) normally increase after a meal but are altered in pathophysiological conditions, such as obesity. Although TG metabolism in the brain remains poorly understood, several brain structures express enzymes that process TG-enriched particles, including mesolimbic structures. For this reason, and because consumption of high-fat diet alters dopamine signaling, we tested the hypothesis that TG might directly target mesolimbic reward circuits to control reward-seeking behaviors. We found that the delivery of small amounts of TG to the brain through the carotid artery rapidly reduced both spontaneous and amphetamine-induced locomotion, abolished preference for palatable food and reduced the motivation to engage in food-seeking behavior. Conversely, targeted disruption of the TG-hydrolyzing enzyme lipoprotein lipase specifically in the nucleus accumbens increased palatable food preference and food-seeking behavior. Finally, prolonged TG perfusion resulted in a return to normal palatable food preference despite continued locomotor suppression, suggesting that adaptive mechanisms occur. These findings reveal new mechanisms by which dietary fat may alter mesolimbic circuit function and reward seeking.


Subject(s)
Brain/metabolism , Feeding Behavior/physiology , Motivation/physiology , Reward , Triglycerides/blood , Amphetamine/pharmacology , Animals , Carotid Arteries/metabolism , Central Nervous System Stimulants/pharmacology , Lipoprotein Lipase/metabolism , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology
9.
Diabetes Metab ; 40(1): 29-33, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24210646

ABSTRACT

Nutrient-sensitive neurons [to glucose and fatty acids (FAs)] are present at many sites throughout the brain, including the hypothalamus and brain stem, and play a key role in the neural control of energy and glucose homoeostasis. Through their neuronal output, FAs can modulate feeding behaviour as well as insulin secretion and activity. Central administration of oleate, for example, inhibits food intake and glucose production in rats. This suggests that daily variations in plasma FA concentrations could be detected by the central nervous system as a signal that contributes to regulation of energy balance. At the cellular level, subpopulations of neurons in the ventromedial and arcuate hypothalamic nuclei are selectively either inhibited or activated by FAs. Possible molecular effectors of these FA effects most likely include the chloride and potassium ion channels. While intracellular metabolism and activation of the ATP-sensitive K(+) channels appear to be necessary for some signalling effects of FAs, at least half the FA responses in ventromedial hypothalamic neurons are mediated by interaction with fatty acid translocase (FAT)/CD36, an FA transporter/receptor that does not require intracellular metabolism to activate downstream signalling. Thus, FAs and their metabolites can modulate neuronal activity by directly monitoring the ongoing fuel availability for brain nutrient-sensing neurons involved in the regulation of energy and glucose homoeostasis. Besides these physiological effects, FA overload or metabolic dysfunction may also impair neural control of energy homoeostasis and contribute to obesity and/or type 2 diabetes in predisposed subjects.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Fatty Acids/metabolism , Feeding Behavior , Hypothalamus/metabolism , Neurons/metabolism , Obesity/metabolism , Eating , Energy Metabolism , Female , Genetic Predisposition to Disease , Homeostasis , Humans , Hypothalamus/physiology , KATP Channels/metabolism , Lipid Metabolism , Male , Potassium Channels/metabolism , Signal Transduction
10.
Diabetes Metab ; 37(2): 83-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21185213

ABSTRACT

Nutrient sensitive neurons (glucose and fatty acids (FA)) are present in many sites throughout the brain, including the hypothalamus and brainstem, and play a key role in the neural control of energy and glucose homeostasis. Through neuronal output, FA may modulate feeding behaviour as well as both insulin secretion and action. For example, central administration of oleate inhibits food intake and glucose production in rats. This suggests that daily variations in plasma FA concentrations might be detected by the central nervous system as a signal which contributes to the regulation of energy balance. At the cellular level, subpopulations of neurons in the ventromedial and arcuate hypothalamic nuclei are selectively either inhibited or activated by FA. Possible molecular effectors of these FA effects likely include chloride or potassium ion channels. While intracellular metabolism and activation of the ATP-sensitive K(+) channel appear to be necessary for some of the signaling effects of FA, at least half of the FA responses in ventromedial hypothalamic neurons are mediated by interaction with FAT/CD36, a FA transporter/receptor that does not require intracellular metabolism to activate downstream signaling. Thus, FA or their metabolites can modulate neuronal activity as a means of directly monitoring ongoing fuel availability by brain nutrient-sensing neurons involved in the regulation of energy and glucose homeostasis. Besides these physiological effects, FA overload or metabolic dysfunction might impair neural control of energy homeostasis and contribute to obesity and/or type 2 diabetes in predisposed subjects.


Subject(s)
Brain/physiology , Energy Metabolism/physiology , Fatty Acids/blood , Fatty Acids/metabolism , Homeostasis/physiology , Animals , Biological Transport , Humans , Hypothalamus/cytology , Neurons/physiology , Neuropeptides/physiology , Neurotransmitter Agents/physiology
11.
Diabetes Metab ; 36(4): 257-62, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20561808

ABSTRACT

Glycolysis is an essential metabolic function that lies at the core of any cellular life. Glucose homoeostasis is, thus, a crucial physiological function of living organisms. A system of plasma glucose-sensing in the portal vein plays a key role in this homoeostasis. Connected to the hypothalamus via the peripheral nervous system, the system allows the body to adapt its response to any variation of portal glycaemia. The hypothalamus controls food intake (exogenous glucose supply) and hepatic glycogenolysis (endogenous glucose supply). Intestinal gluconeogenesis, via the release of glucose into the portal vein, plays a key role in the control of hunger and satiety, and of endogenous glucose production through the modulation of liver insulin sensitivity. The induction of intestinal gluconeogenesis provides a physiological explanation for the satiety effects induced by protein-enriched diets. In particular, the influence of protein-enriched diets on the hypothalamus is comparable to the activation observed after glucose infusion into the portal vein. The induction of intestinal gluconeogenesis also offers an explanation for the early improvement in glycaemia control observed in obese diabetic patients treated by gastric-bypass surgery. In addition to intestinal gluconeogenesis, a number of gastrointestinal hormones involved in the control of food intake exert their effects, at least in part, via the peripheral afferent nervous system. These data emphasize the importance of the gut-brain axis in the understanding and treatment of obesity and type 2 diabetes.


Subject(s)
Appetite Regulation , Blood Glucose/metabolism , Eating , Hypothalamus/metabolism , Insulin/metabolism , Intestinal Mucosa/metabolism , Portal Vein/metabolism , Blood-Brain Barrier , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Gluconeogenesis , Glycolysis , Humans , Insulin/blood , Insulin Resistance , Obesity/metabolism , Obesity/therapy
12.
Diabetologia ; 53(9): 1935-46, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20499047

ABSTRACT

AIMS/HYPOTHESIS: Suppressor of cytokine signalling (SOCS) proteins are powerful inhibitors of pathways involved in survival and function of pancreatic beta cells. Whereas SOCS1 and SOCS3 have been involved in immune and inflammatory processes, respectively, in beta cells, nothing is known about SOCS2 implication in the pancreas. METHODS: Transgenic (tg) mice were generated that constitutively produced SOCS2 in beta cells (betaSOCS2) to define whether this protein is implicated in beta cell functioning and/or survival. RESULTS: Constitutive production of SOCS2 in beta cells leads to hyperglycaemia and glucose intolerance. This phenotype is not a consequence of decreased beta cell mass or inhibition of insulin synthesis. However, insulin secretion to various secretagogues is profoundly altered in intact animals and isolated islets. Interestingly, constitutive SOCS2 production dampens the rise in cytosolic free calcium concentration induced by glucose, while glucose metabolism is unchanged. Moreover, tg islets have a depletion in endoplasmic reticulum Ca(2+) stores, suggesting that SOCS2 interferes with calcium fluxes. Finally, in betaSOCS2 mice proinsulin maturation is impaired, leading to an altered structure of insulin secretory granules and augmented levels of proinsulin. The latter is likely to be due to decreased production of prohormone convertase 1 (PC1/3), which plays a key role in proinsulin cleavage. CONCLUSIONS/INTERPRETATIONS: SOCS2 was shown to be a potent regulator of proinsulin processing and insulin secretion in beta cells. While its constitutive production is insufficient to induce overt diabetes in this mouse model, it causes glucose intolerance. Thus, increased SOCS2 production could be an important event predisposing to beta cell failure.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Blotting, Western , Body Weight/genetics , Body Weight/physiology , Electrophoresis, Polyacrylamide Gel , Endoplasmic Reticulum/metabolism , Insulin Secretion , Mice , Mice, Inbred C57BL , Phenotype , Rats , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling Proteins/genetics
13.
Diabetes Metab ; 35(3): 178-84, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19264524

ABSTRACT

Citrullus colocynthis (colocynth) seeds are traditionally used as antidiabetic medication in Mediterranean countries. The present study evaluated the differential effects of diets enriched with C. colocynthis, sunflower or olive oils on the pancreatic beta-cell mass in streptozotocin (STZ)-induced diabetes in rats. STZ injection induced rapid hyperglycaemia in all animals. However, 2 months later, hyperglycaemia was significantly less pronounced in the rats fed a C. colocynthis oil-enriched diet compared with other rat groups (7.9mM versus 12mM and 16mM with colocynth versus olive and sunflower oils, respectively). Assessment of insulin sensitivity using the homoeostasis model assessment (HOMA) method also indicated less insulin resistance in the rats fed a C. colocynthis oil-enriched diet versus the other rats. Finally, 2 months after STZ injection, the pancreatic beta-cell mass was similar in both the STZ-treated rats fed the colocynth oil-enriched diet and their controls fed the same diet. In contrast, the pancreatic beta-cell mass remained lower in the STZ-induced diabetic rats fed with olive oil- and sunflower oil-enriched diets compared with the C. colocynthis group. We conclude that C. colocynthis oil supplementation may have a beneficial effect by partly preserving or restoring pancreatic beta-cell mass in the STZ-induced diabetes rat model.


Subject(s)
Citrullus , Diabetes Mellitus, Experimental/blood , Helianthus , Plant Oils/therapeutic use , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Insulin/blood , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Male , Olive Oil , Rats , Rats, Wistar
14.
Diabetes Metab ; 35(2): 77-84, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19251449

ABSTRACT

In the adult, the pancreatic beta-cell mass adapts insulin secretion to meet long-term changes in insulin demand and, in particular, in the presence of insulin resistance that is either physiological, such as pregnancy, or pathophysiological, such as obesity. The failure of beta cells to compensate for insulin resistance is a major component of impaired glucose homeostasis and overt diabetes. This defect is clearly the consequence of a decline of insulin response to glucose due to functional beta-cell deficiency. It is also the consequence of an inability of the endocrine pancreas to adapt the beta-cell mass to insulin demand (pancreas plasticity), which eventually leads to a decrease in functional beta-cell mass. This idea has resulted in considerable attention being paid to the development of new therapeutic strategies aiming to preserve and/or regenerate functional beta-cell mass. The latter is governed by a constant balance between beta-cell growth (replication from pre-existing beta cells and neogenesis from precursor cells) and beta-cell death (mainly apoptosis). Disruption of this balance may lead to rapid and marked changes in beta-cell mass. Glucagon-like peptide-1 (GLP-1), an incretin, enhances beta-cell survival (by activating beta-cell proliferation and differentiation, and inhibiting beta-cell apoptosis), thus contributing to the long-term regulation of insulin secretion by maintaining a functional beta-cell mass. The development of drugs regulating this parameter will be the major challenge of the next few years in the management of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Glucagon-Like Peptide 1/physiology , Incretins/physiology , Insulin-Secreting Cells/physiology , Adaptation, Physiological , Dipeptidyl Peptidase 4 , Dipeptidyl-Peptidase IV Inhibitors , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/therapeutic use , Humans , Incretins/therapeutic use , Insulin Resistance , Insulin-Secreting Cells/cytology
15.
Diabetes Metab ; 35(6 Pt 2): 499-507, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20152734

ABSTRACT

Bariatric surgery is indicated in cases of severe obesity. However, malabsorption-based techniques (gastric bypass and biliopancreatic diversion, both of which exclude the duodenum and jejunum from the alimentary circuit), but not restrictive techniques, can abolish type 2 diabetes within days of surgery, even before any significant weight loss has occurred. This means that calorie restriction alone cannot entirely account for this effect. In Goto-Kakizaki rats, a type 2 diabetes model, glycaemic equilibrium is improved by surgical exclusion of the proximal intestine, but deteriorates again when the proximal intestine is reconnected to the circuit in the same animals. This effect is independent of weight, suggesting that the intestine is itself involved in the immediate regulation of carbohydrate homoeostasis. In humans, the rapid improvement in carbohydrate homoeostasis observed after bypass surgery is secondary to an increase in insulin sensitivity rather than an increase in insulin secretion, which occurs later. Several mechanisms are involved--disappearance of hypertriglyceridaemia and decrease in levels of circulating fatty acids, disappearance of the mechanisms of lipotoxicity in the liver and skeletal muscle, and increases in secretion of GLP-1 and PYY--and may be intricately linked. In the medium term and in parallel with weight loss, a decrease in fatty tissue inflammation (which is also seen with restrictive techniques) may also be involved in metabolic improvement. Other mechanisms specific to malabsorption-based techniques (due to the required exclusion of part of the intestine), such as changes in the activity of digestive vagal afferents, changes in intestinal flora and stimulation of intestinal neoglucogenesis, also need to be studied in greater detail. The intestine is, thus, a key organ in the regulation of glycaemic equilibrium and may even be involved in the pathophysiology of type 2 diabetes.


Subject(s)
Bariatric Surgery , Diabetes Complications/prevention & control , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/surgery , Dietary Carbohydrates/metabolism , Insulin Resistance , Obesity, Morbid/physiopathology , Obesity, Morbid/surgery , Abdominal Fat , Animals , Blood Glucose/metabolism , Caloric Restriction , Confounding Factors, Epidemiologic , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Gastric Bypass , Gastroplasty , Gluconeogenesis , Humans , Inflammation , Jejunoileal Bypass , Lipid Metabolism , Obesity, Morbid/complications , Obesity, Morbid/metabolism , Weight Loss
16.
Diabetes Metab ; 33(3): 177-82, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17475532

ABSTRACT

Nutrient sensitive neurons (glucose and fatty acids, FA) are present in both the hypothalamus and the brainstem and play a key role in nervous control of energy homeostasis. Through neuronal output, especially the autonomic nervous system, it is now evidenced that FA may modulate food behaviour and both insulin secretion and action. For example, central administration of oleate inhibits both food intake and hepatic glucose production in rats. This suggests that a slight increase in plasma FA concentrations in the postprandial state might be detected by the central nervous system as a satiety signal. At cellular levels, subpopulations of FA-sensitive neurons (either excited or inhibited by FA) are now identified within the hypothalamus. However molecular effectors of FA effects remain unclear. They probably include ionic channels such as chloride or potassium. FA metabolism seems also required to induce neuronal response. Thus, FA per se or their metabolites modulate neuronal activity, as a mean of directly monitoring ongoing fuel availability by CNS nutrient-sensing neurons involved in the regulation of insulin secretion. Beside these physiological effects, FA overload or dysfunction of their metabolism could impair nervous control of energy homeostasis and contribute to development of obesity and/or type 2 diabetes in predisposed subjects.


Subject(s)
Energy Metabolism , Fatty Acids/physiology , Nervous System Physiological Phenomena , Animals , Biological Transport , Brain/metabolism , Energy Metabolism/drug effects , Fatty Acids/blood , Fatty Acids/pharmacology , Homeostasis , Humans , Hypothalamus/metabolism , Models, Animal , Nervous System/drug effects
18.
J Neurophysiol ; 95(3): 1491-8, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16306178

ABSTRACT

Pharmacological manipulation of fatty acid metabolism in the hypothalamic arcuate nucleus (ARC) alters energy balance and glucose homeostasis. Thus, we tested the hypotheses that distinctive populations of ARC neurons are oleic acid (OA) sensors that exhibit a glucose dependency, independent of whether some of these OA sensors are also glucose-sensing neurons. We used patch-clamp recordings to investigate the effects of OA on ARC neurons in brain slices from 14- to 21-day-old Sprague-Dawley (SD) rats. Additionally, we recorded spontaneous discharge rate in ARC neurons in 8-wk-old fed and fasted SD rats in vivo. Patch-clamp studies showed that in 2.5 mM glucose 12 of 94 (13%) ARC neurons were excited by 2 microM OA (OA-excited or OAE neurons), whereas six of 94 (6%) were inhibited (OA-inhibited2.5 or OAI2.5 neurons). In contrast, in 0.1 mM glucose, OA inhibited six of 20 (30%) ARC neurons (OAI0.1 neurons); none was excited. None of the OAI0.1 neurons responded to OA in 2.5 mM glucose. Thus OAI2.5 and OAI0.1 neurons are distinct. Similarly, in seven of 20 fed rats (35%) the overall response was OAE-like, whereas in three of 20 (15%) it was OAI-like. In contrast, in fasted rats only OAI-like response were observed (three of 15; 20%). There was minimal overlap between OA-sensing neurons and glucose-sensing neurons. In conclusion, OA regulated three distinct subpopulations of ARC neurons in a glucose-dependent fashion. These data suggest that an interaction between glucose and fatty acids regulates OA sensing in ARC neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Fatty Acids/metabolism , Glucose/metabolism , Nerve Net/physiology , Neurons/classification , Neurons/physiology , Oleic Acid/administration & dosage , Action Potentials/drug effects , Action Potentials/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Extracellular Fluid/metabolism , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Metabolic Clearance Rate/drug effects , Nerve Net/drug effects , Neurons/drug effects , Rats , Rats, Sprague-Dawley
19.
Diabetologia ; 48(6): 1189-97, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15889232

ABSTRACT

AIMS/HYPOTHESIS: IGFs, IGF receptors and IGF binding proteins (IGFBPs) are widely expressed in the central nervous system. To investigate the physiological significance of IGFBP-6 in the brain we established two transgenic mouse lines overexpressing human (h)-IGFBP-6 under the control of glial fibrillary acidic protein promoter. Increasing evidence suggests that insulin/IGF signalling pathways could be implicated in the neuroendocrine regulation of energy homeostasis. We explored the impact of brain IGFBP-6 overexpression on the regulation of food intake and energy balance. METHODS: Transgenic mice were fed either a control diet or a high-fat diet for up to 3 months. Glucose and insulin tolerance tests were carried out before and after the diet period. Plasma parameters (insulin, leptin, glucose, NEFAs and triglycerides) were measured, and uncoupling protein 1 (UCP-1) expression was quantified in brown adipose tissue. Oxygen consumption was also measured in both groups. RESULTS: The transgenic mice fed a high-fat diet for 3 months developed obesity, showing increases in plasma leptin, glucose and insulin levels and mild insulin resistance. As compared with wild-type mice, no significant differences were found in the quantity of food intake. However, UCP-1 expression was down-regulated in the brown adipose tissue of the transgenic mice. CONCLUSIONS/INTERPRETATION: Our results show that brain IGFBP-6 has an impact on the regulation of energy homeostasis. These transgenic h-IGFBP-6 mice may be considered a new tool for studies of the involvement of the brain IGF system in metabolism control and obesity.


Subject(s)
Brain/physiology , Energy Metabolism , Insulin-Like Growth Factor Binding Protein 6/genetics , Adipose Tissue, Brown/metabolism , Animals , Blood Glucose/metabolism , Brain/drug effects , Carrier Proteins/metabolism , Energy Intake , Fatty Acids, Nonesterified/blood , Glucose/pharmacology , Homeostasis , Humans , Insulin/blood , Ion Channels , Leptin/blood , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitochondrial Proteins , Oxygen Consumption/drug effects , Triglycerides/blood , Uncoupling Protein 1
20.
Am J Physiol Endocrinol Metab ; 288(1): E148-54, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15353406

ABSTRACT

To evaluate the relationship between the development of obesity, nervous system activity, and insulin secretion and action, we tested the effect of a 2-mo high-fat diet in rats (HF rats) on glucose tolerance, glucose-induced insulin secretion (GIIS), and glucose turnover rate compared with chow-fed rats (C rats). Moreover, we measured pancreatic and hepatic norepinephrine (NE) turnover, as assessment of sympathetic tone, and performed hypothalamic microdialysis to quantify extracellular NE turnover. Baseline plasma triglyceride, free fatty acid, insulin, and glucose concentrations were similar in both groups. After 2 days of diet, GIIS was elevated more in HF than in C rats, whereas plasma glucose time course was similar. There was a significant increase in basal pancreatic NE level of HF rats, and a twofold decrease in the fractional turnover constant was observed, indicating a change in sympathetic tone. In ventromedian hypothalamus of HF rats, the decrease in NE extracellular concentration after a glucose challenge was lower compared with C rats, suggesting changes in overall activity. After 7 days, insulin hypersecretion persisted, and glucose intolerance appeared. Later (2 mo), there was no longer insulin hypersecretion, whereas glucose intolerance worsened. At all times, HF rats also displayed hepatic insulin resistance. On day 2 of HF diet, GIIS returned to normal after treatment with oxymetazoline, an alpha(2A)-adrenoreceptor agonist, thus suggesting the involvement of a low sympathetic tone in insulin hypersecretion in response to glucose in HF rats. In conclusion, the HF diet rapidly results in an increased GIIS, at least in part related to a decreased sympathetic tone, which can be the first step of a cascade of events leading to impaired glucose homeostasis.


Subject(s)
Dietary Fats/pharmacology , Insulin/metabolism , Sympathetic Nervous System/physiology , Animals , Blood Glucose/metabolism , Body Weight , Eating , Hyperglycemia/physiopathology , Hyperinsulinism/physiopathology , Hypothalamus/physiology , Insulin/blood , Insulin Secretion , Liver/physiology , Male , Norepinephrine/metabolism , Oxymetazoline/pharmacology , Rats , Rats, Wistar , Sympathetic Nervous System/drug effects , Sympathomimetics/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...