Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 25(5): 1213-1220, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36597795

RESUMEN

AIMS: To examine association of liver-expressed antimicrobial peptide 2 (LEAP2), an endogenous ghrelin antagonist with anorexiant effects, to key cardiometabolic risk factors in people with overweight and obesity. METHODS: In this cross-sectional study, we sought to identify associations between LEAP2 levels and cardiometabolic risk factors, including body composition (dual X-ray absorptiometry), insulin and glucose metabolism (oral and intravenous glucose tolerance tests and hyperinsulinaemic-euglycaemic clamps), plasma lipids and inflammation markers (ELISA and multiplex assays). RESULTS: In 65 participants with overweight or obesity (63.1% male, mean age 31.3 ± 8.5 years), LEAP2 levels were associated with total body fat, but not with body mass index or waist-hip ratio in both univariable and age- and sex-adjusted models (P < 0.05). Higher LEAP2 level was also positively associated with higher insulin secretion in univariable (P = 0.047) and multivariable models adjusted for age, sex and body fat (P = 0.03), but not with fasting glucose levels (P ≥ 0.05). Higher LEAP2 levels were associated insulin resistance (P = 0.07) after adjustment for age and sex, but the association disappeared after an additional adjustment for body fat (P = 0.2). There was an inverse association between LEAP2 levels and nuclear factor kappa-B (NFκB) activity in the peripheral blood mononuclear cells in age-, sex- and body fat-adjusted models (P = 0.04). There were no associations with cardiovascular risk factors (lipids, blood pressure) or other inflammation markers. CONCLUSIONS: These results provide important insights into the association between LEAP2 and cardiometabolic health in a high-risk population of individuals with overweight and obesity. This is a first report of an association between LEAP2 and insulin secretion, insulin sensitivity and NFκB activity. LEAP2 may represent an important potential therapeutic target to promote insulin secretion in people with type 2 diabetes and obesity.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Obesidad , Sobrepeso , Adulto , Femenino , Humanos , Masculino , Adulto Joven , Estudios Transversales , Diabetes Mellitus Tipo 2/complicaciones , Hepcidinas/metabolismo , Inflamación/complicaciones , Insulina/metabolismo , Secreción de Insulina , Leucocitos Mononucleares/metabolismo , Lípidos , Obesidad/complicaciones , Sobrepeso/complicaciones
2.
FASEB J ; : fj201800634R, 2018 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-29932868

RESUMEN

Hunger-sensing agouti-related peptide (AgRP) neurons ensure survival by adapting metabolism and behavior to low caloric environments. This adaption is accomplished by consolidating food intake, suppressing energy expenditure, and maximizing fat storage (nutrient partitioning) for energy preservation. The intracellular mechanisms responsible are unknown. Here we report that AgRP carnitine acetyltransferase (Crat) knockout (KO) mice exhibited increased fatty acid utilization and greater fat loss after 9 d of calorie restriction (CR). No differences were seen in mice with ad libitum food intake. Eleven days ad libitum feeding after CR resulted in greater food intake, rebound weight gain, and adiposity in AgRP Crat KO mice compared with wild-type controls, as KO mice act to restore pre-CR fat mass. Collectively, this study highlights the importance of Crat in AgRP neurons to regulate nutrient partitioning and fat mass during chronically reduced caloric intake. The increased food intake, body weight gain, and adiposity in KO mice after CR also highlights the detrimental and persistent metabolic consequence of impaired substrate utilization associated with CR. This finding may have significant implications for postdieting weight management in patients with metabolic diseases.-Reichenbach, A., Stark, R., Mequinion, M., Lockie, S. H., Lemus, M. B., Mynatt, R. L., Luquet, S., Andrews, Z. B. Carnitine acetyltransferase (Crat) in hunger-sensing AgRP neurons permits adaptation to calorie restriction.

3.
J Neurosci ; 36(10): 3049-63, 2016 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-26961958

RESUMEN

Calorie restriction (CR) is neuroprotective in Parkinson's disease (PD) although the mechanisms are unknown. In this study we hypothesized that elevated ghrelin, a gut hormone with neuroprotective properties, during CR prevents neurodegeneration in an 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of PD. CR attenuated the MPTP-induced loss of substantia nigra (SN) dopamine neurons and striatal dopamine turnover in ghrelin WT but not KO mice, demonstrating that ghrelin mediates CR's neuroprotective effect. CR elevated phosphorylated AMPK and ACC levels in the striatum of WT but not KO mice suggesting that AMPK is a target for ghrelin-induced neuroprotection. Indeed, exogenous ghrelin significantly increased pAMPK in the SN. Genetic deletion of AMPKß1 and 2 subunits only in dopamine neurons prevented ghrelin-induced AMPK phosphorylation and neuroprotection. Hence, ghrelin signaling through AMPK in SN dopamine neurons mediates CR's neuroprotective effects. We consider targeting AMPK in dopamine neurons may recapitulate neuroprotective effects of CR without requiring dietary intervention.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Restricción Calórica , Ghrelina/metabolismo , Intoxicación por MPTP/patología , Intoxicación por MPTP/prevención & control , Enfermedad de Parkinson/fisiopatología , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Recuento de Células , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ghrelina/genética , Ghrelina/farmacología , Proteína Ácida Fibrilar de la Glía/metabolismo , Intoxicación por MPTP/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Neuronas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tirosina 3-Monooxigenasa/metabolismo
4.
J Biol Chem ; 289(11): 7257-63, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24497630

RESUMEN

Synthesis of phosphoenolpyruvate (PEP) from oxaloacetate is an absolute requirement for gluconeogenesis from mitochondrial substrates. Generally, this reaction has solely been attributed to the cytosolic isoform of PEPCK (PEPCK-C), although loss of the mitochondrial isoform (PEPCK-M) has never been assessed. Despite catalyzing the same reaction, to date the only significant role reported in mammals for the mitochondrial isoform is as a glucose sensor necessary for insulin secretion. We hypothesized that this nutrient-sensing mitochondrial GTP-dependent pathway contributes importantly to gluconeogenesis. PEPCK-M was acutely silenced in gluconeogenic tissues of rats using antisense oligonucleotides both in vivo and in isolated hepatocytes. Silencing PEPCK-M lowers plasma glucose, insulin, and triglycerides, reduces white adipose, and depletes hepatic glycogen, but raises lactate. There is a switch of gluconeogenic substrate preference to glycerol that quantitatively accounts for a third of glucose production. In contrast to the severe mitochondrial deficiency characteristic of PEPCK-C knock-out livers, hepatocytes from PEPCK-M-deficient livers maintained normal oxidative function. Consistent with its predicted role, gluconeogenesis rates from hepatocytes lacking PEPCK-M are severely reduced for lactate, alanine, and glutamine, but not for pyruvate and glycerol. Thus, PEPCK-M has a direct role in fasted and fed glucose homeostasis, and this mitochondrial GTP-dependent pathway should be reconsidered for its involvement in both normal and diabetic metabolism.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Gluconeogénesis , Péptidos y Proteínas de Señalización Intracelular/fisiología , Hígado/enzimología , Hígado/metabolismo , Mitocondrias/enzimología , Fosfoenolpiruvato Carboxiquinasa (GTP)/fisiología , Alimentación Animal , Animales , Glucemia/metabolismo , Privación de Alimentos , Silenciador del Gen , Glicerol/metabolismo , Glucógeno/metabolismo , Guanosina Trifosfato/metabolismo , Hepatocitos/citología , Homeostasis , Insulina/metabolismo , Isoenzimas/fisiología , Ácido Láctico/metabolismo , Masculino , Mitocondrias/metabolismo , Oligonucleótidos Antisentido/química , Oxígeno/metabolismo , Consumo de Oxígeno , Ratas , Ratas Sprague-Dawley
5.
Biochim Biophys Acta ; 1840(4): 1313-30, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24177027

RESUMEN

BACKGROUND: Plasma glucose levels are tightly regulated within a narrow physiologic range. Insulin-mediated glucose uptake by tissues must be balanced by the appearance of glucose from nutritional sources, glycogen stores, or gluconeogenesis. In this regard, a common pathway regulating both glucose clearance and appearance has not been described. The metabolism of glucose to produce ATP is generally considered to be the primary stimulus for insulin release from beta-cells. Similarly, gluconeogenesis from phosphoenolpyruvate (PEP) is believed to be the primarily pathway via the cytosolic isoform of phosphoenolpyruvate carboxykinase (PEPCK-C). These models cannot adequately explain the regulation of insulin secretion or gluconeogenesis. SCOPE OF REVIEW: A metabolic sensing pathway involving mitochondrial GTP (mtGTP) and PEP synthesis by the mitochondrial isoform of PEPCK (PEPCK-M) is associated with glucose-stimulated insulin secretion from pancreatic beta-cells. Here we examine whether there is evidence for a similar mtGTP-dependent pathway involved in gluconeogenesis. In both islets and the liver, mtGTP is produced at the substrate level by the enzyme succinyl CoA synthetase (SCS-GTP) with a rate proportional to the TCA cycle. In the beta-cell PEPCK-M then hydrolyzes mtGTP in the production of PEP that, unlike mtGTP, can escape the mitochondria to generate a signal for insulin release. Similarly, PEPCK-M and mtGTP might also provide a significant source of PEP in gluconeogenic tissues for the production of glucose. This review will focus on the possibility that PEPCK-M, as a sensor for TCA cycle flux, is a key mechanism to regulate both insulin secretion and gluconeogenesis suggesting conservation of this biochemical mechanism in regulating multiple aspects of glucose homeostasis. Moreover, we propose that this mechanism may be important for regulating insulin secretion and gluconeogenesis compared to canonical nutrient sensing pathways. MAJOR CONCLUSIONS: PEPCK-M, initially believed to be absent in islets, carries a substantial metabolic flux in beta-cells. This flux is intimately involved with the coupling of glucose-stimulated insulin secretion. PEPCK-M activity may have been similarly underestimated in glucose producing tissues and could potentially be an unappreciated but important source of gluconeogenesis. GENERAL SIGNIFICANCE: The generation of PEP via PEPCK-M may occur via a metabolic sensing pathway important for regulating both insulin secretion and gluconeogenesis. This article is part of a Special Issue entitled Frontiers of Mitochondrial Research.


Asunto(s)
Glucosa/metabolismo , Mitocondrias/enzimología , Fosfoenolpiruvato Carboxiquinasa (GTP)/fisiología , Animales , Gluconeogénesis/fisiología , Homeostasis/fisiología , Humanos , Células Secretoras de Insulina/fisiología , Isoenzimas/fisiología , Mitocondrias/metabolismo , Proteínas Mitocondriales/fisiología
6.
J Neuroendocrinol ; 36(6): e13382, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38468186

RESUMEN

Olfaction is the most ancient sense and is needed for food-seeking, danger protection, mating and survival. It is often the first sensory modality to perceive changes in the external environment, before sight, taste or sound. Odour molecules activate olfactory sensory neurons that reside on the olfactory epithelium in the nasal cavity, which transmits this odour-specific information to the olfactory bulb (OB), where it is relayed to higher brain regions involved in olfactory perception and behaviour. Besides odour processing, recent studies suggest that the OB extends its function into the regulation of food intake and energy balance. Furthermore, numerous hormone receptors associated with appetite and metabolism are expressed within the OB, suggesting a neuroendocrine role outside the hypothalamus. Olfactory cues are important to promote food preparatory behaviours and consumption, such as enhancing appetite and salivation. In addition, altered metabolism or energy state (fasting, satiety and overnutrition) can change olfactory processing and perception. Similarly, various animal models and human pathologies indicate a strong link between olfactory impairment and metabolic dysfunction. Therefore, understanding the nature of this reciprocal relationship is critical to understand how olfactory or metabolic disorders arise. This present review elaborates on the connection between olfaction, feeding behaviour and metabolism and will shed light on the neuroendocrine role of the OB as an interface between the external and internal environments. Elucidating the specific mechanisms by which olfactory signals are integrated and translated into metabolic responses holds promise for the development of targeted therapeutic strategies and interventions aimed at modulating appetite and promoting metabolic health.


Asunto(s)
Conducta Alimentaria , Sistemas Neurosecretores , Bulbo Olfatorio , Bulbo Olfatorio/fisiología , Bulbo Olfatorio/metabolismo , Animales , Humanos , Sistemas Neurosecretores/fisiología , Sistemas Neurosecretores/metabolismo , Conducta Alimentaria/fisiología , Olfato/fisiología , Metabolismo Energético/fisiología
7.
Mol Metab ; 78: 101826, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37898450

RESUMEN

OBJECTIVE: The sensory detection of food and food cues suppresses Agouti related peptide (AgRP) neuronal activity prior to consumption with greatest suppression occurring in response to highly caloric food or interoceptive energy need. However, the interoceptive mechanisms priming an appropriate AgRP neural response to external sensory information of food availability remain unexplored. Since hunger increases plasma ghrelin, we hypothesized that ghrelin receptor (GHSR) signalling on AgRP neurons is a key interoceptive mechanism integrating energy need with external sensory cues predicting caloric availability. METHODS: We used in vivo photometry to measure the effects of ghrelin administration or fasting on AgRP neural activity with GCaMP6s and dopamine release in the nucleus accumbens with GRAB-DA in mice lacking ghrelin receptors in AgRP neurons. RESULTS: The deletion of GHSR on AgRP neurons prevented ghrelin-induced food intake, motivation and AgRP activity. The presentation of food (peanut butter pellet) or a wooden dowel suppressed AgRP activity in fasted WT but not mice lacking GHSRs in AgRP neurons. Similarly, peanut butter and a wooden dowel increased dopamine release in the nucleus accumbens after ip ghrelin injection in WT but not mice lacking GHSRs in AgRP neurons. No difference in dopamine release was observed in fasted mice. Finally, ip ghrelin administration did not directly increase dopamine neural activity in the ventral tegmental area. CONCLUSIONS: Our results suggest that AgRP GHSRs integrate an interoceptive state of energy need with external sensory information to produce an optimal change in AgRP neural activity. Thus, ghrelin signalling on AgRP neurons is more than just a feedback signal to increase AgRP activity during hunger.


Asunto(s)
Ingestión de Alimentos , Ghrelina , Ratones , Animales , Ghrelina/metabolismo , Proteína Relacionada con Agouti/metabolismo , Dopamina/metabolismo , Neuronas/metabolismo
8.
Mol Metab ; 77: 101803, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37690518

RESUMEN

OBJECTIVE: An environmental context, which reliably predicts food availability, can increase the appetitive food drive within the same environment context. However, hunger is required for the development of such a context-induced feeding (CIF) response, suggesting the neural circuits sensitive to hunger link an internal energy state with a particular environment context. Since Agouti related peptide (AgRP) neurons are activated by energy deficit, we hypothesised that AgRP neurons are both necessary and sufficient to drive CIF. METHODS: To examine the role of AgRP neurons in the CIF process, we used fibre photometry with GCaMP7f, chemogenetic activation of AgRP neurons, as well as optogenetic control of AgRP neurons to facilitate acute temporal control not permitted with chemogenetics. RESULTS: A CIF response at test was only observed when mice were fasted during context training and AgRP population activity at test showed an attenuated inhibitory response to food, suggesting increased food-seeking and/or decreased satiety signalling drives the increased feeding response at test. Intriguingly, chemogenetic activation of AgRP neurons during context training did not increase CIF, suggesting precise temporal firing properties may be required. Indeed, termination of AgRP neuronal photostimulation during context training (ON-OFF in context), in the presence or absence of food, increased CIF. Moreover, photoinhibition of AgRP neurons during context training in fasted mice was sufficient to drive a subsequent CIF in the absence of food. CONCLUSIONS: Our results suggest that AgRP neurons regulate the acquisition of CIF when the acute inhibition of AgRP activity is temporally matched to context exposure. These results establish acute AgRP inhibition as a salient neural event underscoring the effect of hunger on associative learning.

9.
Biol Psychiatry ; 93(4): 309-321, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36400605

RESUMEN

BACKGROUND: A greater understanding of how the brain controls appetite is fundamental to developing new approaches for treating diseases characterized by dysfunctional feeding behavior, such as obesity and anorexia nervosa. METHODS: By modeling neural network dynamics related to homeostatic state and body mass index, we identified a novel pathway projecting from the medial prefrontal cortex (mPFC) to the lateral hypothalamus (LH) in humans (n = 53). We then assessed the physiological role and dissected the function of this mPFC-LH circuit in mice. RESULTS: In vivo recordings of population calcium activity revealed that this glutamatergic mPFC-LH pathway is activated in response to acute stressors and inhibited during food consumption, suggesting a role in stress-related control over food intake. Consistent with this role, inhibition of this circuit increased feeding and sucrose seeking during mild stressors, but not under nonstressful conditions. Finally, chemogenetic or optogenetic activation of the mPFC-LH pathway is sufficient to suppress food intake and sucrose seeking in mice. CONCLUSIONS: These studies identify a glutamatergic mPFC-LH circuit as a novel stress-sensitive anorexigenic neural pathway involved in the cortical control of food intake.


Asunto(s)
Conducta Alimentaria , Área Hipotalámica Lateral , Corteza Prefrontal , Estrés Psicológico , Animales , Humanos , Ratones , Conducta Alimentaria/fisiología , Área Hipotalámica Lateral/fisiología , Corteza Prefrontal/fisiología , Estrés Psicológico/fisiopatología
10.
Proc Natl Acad Sci U S A ; 106(27): 11288-93, 2009 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-19549853

RESUMEN

Hepatic gluconeogenesis is a major contributing factor to hyperglycemia in the fasting and postprandial states in type 2 diabetes mellitus (T2DM). Because Sirtuin 1 (SirT1) induces hepatic gluconeogenesis during fasting through the induction of phosphoenolpyruvate carboxylase kinase (PEPCK), fructose-1,6-bisphosphatase (FBPase), and glucose-6-phosphatase (G6Pase) gene transcription, we hypothesized that reducing SirT1, by using an antisense oligonucleotide (ASO), would decrease fasting hyperglycemia in a rat model of T2DM. SirT1 ASO lowered both fasting glucose concentration and hepatic glucose production in the T2DM rat model. Whole body insulin sensitivity was also increased in the SirT1 ASO treated rats as reflected by a 25% increase in the glucose infusion rate required to maintain euglycemia during the hyperinsulinemic-euglycemic clamp and could entirely be attributed to increased suppression of hepatic glucose production by insulin. The reduction in basal and clamped rates of glucose production could in turn be attributed to decreased expression of PEPCK, FBPase, and G6Pase due to increased acetylation of signal transducer and activator of transcription 3 (STAT3), forkhead box O1 (FOXO1), and peroxisome proliferator-activated receptor-gamma coactivator 1alpha (PGC-1alpha), known substrates of SirT1. In addition to the effects on glucose metabolism, SirT1 ASO decreased plasma total cholesterol, which was attributed to increased cholesterol uptake and export from the liver. These results indicate that inhibition of hepatic SirT1 may be an attractive approach for treatment of T2DM.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamiento del Gen , Glucosa/biosíntesis , Insulina/metabolismo , Hígado/metabolismo , Sirtuinas/deficiencia , Acetilación/efectos de los fármacos , Animales , Colesterol/sangre , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Hiperinsulinismo/sangre , Hiperinsulinismo/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Oligonucleótidos Antisentido/farmacología , Ratas , Ratas Sprague-Dawley , Sirtuina 1 , Sirtuinas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos
11.
Elife ; 112022 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-35018884

RESUMEN

Agouti-related peptide (AgRP) neurons increase motivation for food, however, whether metabolic sensing of homeostatic state in AgRP neurons potentiates motivation by interacting with dopamine reward systems is unexplored. As a model of impaired metabolic-sensing, we used the AgRP-specific deletion of carnitine acetyltransferase (Crat) in mice. We hypothesised that metabolic sensing in AgRP neurons is required to increase motivation for food reward by modulating accumbal or striatal dopamine release. Studies confirmed that Crat deletion in AgRP neurons (KO) impaired ex vivo glucose-sensing, as well as in vivo responses to peripheral glucose injection or repeated palatable food presentation and consumption. Impaired metabolic-sensing in AgPP neurons reduced acute dopamine release (seconds) to palatable food consumption and during operant responding, as assessed by GRAB-DA photometry in the nucleus accumbens, but not the dorsal striatum. Impaired metabolic-sensing in AgRP neurons suppressed radiolabelled 18F-fDOPA accumulation after ~30 min in the dorsal striatum but not the nucleus accumbens. Impaired metabolic sensing in AgRP neurons suppressed motivated operant responding for sucrose rewards during fasting. Thus, metabolic-sensing in AgRP neurons is required for the appropriate temporal integration and transmission of homeostatic hunger-sensing to dopamine signalling in the striatum.


Asunto(s)
Proteína Relacionada con Agouti/genética , Cuerpo Estriado/fisiología , Dopamina/fisiología , Homeostasis , Neuronas/fisiología , Transducción de Señal , Proteína Relacionada con Agouti/metabolismo , Animales , Ratones , Ratones Noqueados
12.
Am J Physiol Endocrinol Metab ; 301(6): E1174-83, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21862723

RESUMEN

Islet damage from glucose toxicity is implicated in the pathogenesis of type 2 diabetes, but the sequence of events leading to islet cell dysfunction and hyperglycemia remains unclear. To examine the early stages of islet pathology resulting from increased basal glucose loads, normal awake rats were infused with glucose continuously for 10 days. Plasma glucose and markers of islet and liver function were monitored throughout the infusion. After initial hyperglycemia, rats adapted to the infusion and maintained euglycemia for approximately 4 days. Continued infusion led to worsening hyperglycemia in just 5% of rats after 6 days, but 69% after 8 days and 89% after 10 days, despite unchanged basal and stimulated plasma insulin and C-peptide concentrations. In contrast, plasma glucagon concentrations increased fivefold. Endogenous glucose production (EGP) was appropriately suppressed after 4 days (2.8 ± 0.7 vs. 6.1 ± 0.4 mg·kg(-1)·min(-1) on day 0, P < 0.001) but tripled between days 4 and 8 (9.9 ± 1.7 mg·kg(-1)·min(-1), P < 0.01). Surprisingly, the increase in EGP was accompanied by increased mitochondrial phosphoenolpyruvate carboxykinase expression with appropriate suppression of the cytosolic isoform. Infusion of anti-glucagon antibodies normalized plasma glucose to levels identical to those on day 4 and ∼300 mg/dl lower than controls. This improved glycemia was associated with a 60% reduction in EGP. These data support the novel concept that glucose toxicity may first manifest as α-cell dysfunction prior to any measurable deficit in insulin secretion. Such hyperglucagonemia could lead to excessive glucose production overwhelming the capacity of the ß-cell to maintain glucose homeostasis.


Asunto(s)
Glucagón/sangre , Glucosa/administración & dosificación , Hiperglucemia/etiología , Insulina/metabolismo , Animales , Regulación hacia Abajo , Esquema de Medicación , Glucagón/fisiología , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/fisiología , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa/métodos , Hiperglucemia/sangre , Hiperglucemia/inducido químicamente , Bombas de Infusión , Secreción de Insulina , Masculino , Modelos Animales , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Regulación hacia Arriba
13.
Nutrients ; 13(6)2021 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-34200678

RESUMEN

Food homeostatic states (hunger and satiety) influence the cognitive systems regulating impulsive responses, but the direction and specific mechanisms involved in this effect remain elusive. We examined how fasting, and satiety, affect cognitive mechanisms underpinning disinhibition using a novel framework and a gamified test-battery. Thirty-four participants completed the test-battery measuring three cognitive facets of disinhibition: attentional control, information gathering and monitoring of feedback, across two experimental sessions: one after overnight fasting and another after a standardised meal. Homeostatic state was assessed using subjective self-reports and biological markers (i.e., blood-derived liver-expressed antimicrobial protein 2 (LEAP-2), insulin and leptin). We found that participants who experienced greater subjective hunger during the satiety session were more impulsive in the information gathering task; results were not confounded by changes in mood or anxiety. Homeostatic state did not significantly influence disinhibition mechanisms linked to attentional control or feedback monitoring. However, we found a significant interaction between homeostatic state and LEAP-2 on attentional control, with higher LEAP-2 associated with faster reaction times in the fasted condition only. Our findings indicate lingering hunger after eating increases impulsive behaviour via reduced information gathering. These findings identify a novel mechanism that may underpin the tendency to overeat and/or engage in broader impulsive behaviours.


Asunto(s)
Cognición/fisiología , Homeostasis , Hambre/fisiología , Pruebas Neuropsicológicas , Adolescente , Adulto , Péptidos Catiónicos Antimicrobianos/metabolismo , Apetito/fisiología , Atención/fisiología , Proteínas Sanguíneas/metabolismo , Toma de Decisiones , Retroalimentación , Femenino , Hormonas/metabolismo , Humanos , Masculino , Saciedad , Adulto Joven
14.
Elife ; 102021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33779547

RESUMEN

Feeding is critical for survival, and disruption in the mechanisms that govern food intake underlies disorders such as obesity and anorexia nervosa. It is important to understand both food intake and food motivation to reveal mechanisms underlying feeding disorders. Operant behavioral testing can be used to measure the motivational component to feeding, but most food intake monitoring systems do not measure operant behavior. Here, we present a new solution for monitoring both food intake and motivation in rodent home-cages: the Feeding Experimentation Device version 3 (FED3). FED3 measures food intake and operant behavior in rodent home-cages, enabling longitudinal studies of feeding behavior with minimal experimenter intervention. It has a programmable output for synchronizing behavior with optogenetic stimulation or neural recordings. Finally, FED3 design files are open-source and freely available, allowing researchers to modify FED3 to suit their needs.


Obesity and anorexia nervosa are two health conditions related to food intake. Researchers studying these disorders in animal models need to both measure food intake and assess behavioural factors: that is, why animals seek and consume food. Measuring an animal's food intake is usually done by weighing food containers. However, this can be inaccurate due to the small amount of food that rodents eat. As for studying feeding motivation, this can involve calculating the number of times an animal presses a lever to receive a food pellet. These tests are typically conducted in hour-long sessions in temporary testing cages, called operant boxes. Yet, these tests only measure a brief period of a rodent's life. In addition, it takes rodents time to adjust to these foreign environments, which can introduce stress and may alter their feeding behaviour. To address this, Matikainen-Ankney, Earnest, Ali et al. developed a device for monitoring food intake and feeding behaviours around the clock in rodent home cages with minimal experimenter intervention. This 'Feeding Experimentation Device' (FED3) features a pellet dispenser and two 'nose-poke' sensors to measure total food intake, as well as motivation for and learning about food rewards. The battery-powered, wire-free device fits in standard home cages, enabling long-term studies of feeding behaviour with minimal intervention from investigators and less stress on the animals. This means researchers can relate data to circadian rhythms and meal patterns, as Matikainen-Ankney did here. Moreover, the device software is open-source so researchers can customise it to suit their experimental needs. It can also be programmed to synchronise with other instruments used in animal experiments, or across labs running the same behavioural tasks for multi-site studies. Used in this way, it could help improve reproducibility and reliability of results from such studies. In summary, Matikainen-Ankney et al. have presented a new practical solution for studying food-related behaviours in mice and rats. Not only could the device be useful to researchers, it may also be suitable to use in educational settings such as teaching labs and classrooms.


Asunto(s)
Crianza de Animales Domésticos , Condicionamiento Operante , Diseño de Equipo/instrumentación , Conducta Alimentaria , Vivienda para Animales , Roedores/fisiología , Animales , Ingestión de Alimentos , Femenino , Masculino , Ratones
15.
J Biol Chem ; 284(39): 26578-90, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19635791

RESUMEN

Pancreatic beta-cells couple the oxidation of glucose to the secretion of insulin. Apart from the canonical K(ATP)-dependent glucose-stimulated insulin secretion (GSIS), there are important K(ATP)-independent mechanisms involving both anaplerosis and mitochondrial GTP (mtGTP). How mtGTP that is trapped within the mitochondrial matrix regulates the cytosolic calcium increases that drive GSIS remains a mystery. Here we have investigated whether the mitochondrial isoform of phosphoenolpyruvate carboxykinase (PEPCK-M) is the GTPase linking hydrolysis of mtGTP made by succinyl-CoA synthetase (SCS-GTP) to an anaplerotic pathway producing phosphoenolpyruvate (PEP). Although cytosolic PEPCK (PEPCK-C) is absent, PEPCK-M message and protein were detected in INS-1 832/13 cells, rat islets, and mouse islets. PEPCK enzymatic activity is half that of primary hepatocytes and is localized exclusively to the mitochondria. Novel (13)C-labeling strategies in INS-1 832/13 cells and islets measured substantial contribution of PEPCK-M to the synthesis of PEP. As high as 30% of PEP in INS-1 832/13 cells and 41% of PEP in rat islets came from PEPCK-M. The contribution of PEPCK-M to overall PEP synthesis more than tripled with glucose stimulation. Silencing the PEPCK-M gene completely inhibited GSIS underscoring its central role in mitochondrial metabolism-mediated insulin secretion. Given that mtGTP synthesized by SCS-GTP is an indicator of TCA flux that is crucial for GSIS, PEPCK-M is a strong candidate to link mtGTP synthesis with insulin release through anaplerotic PEP cycling.


Asunto(s)
Guanosina Trifosfato/metabolismo , Insulina/metabolismo , Mitocondrias/enzimología , Fosfoenolpiruvato Carboxilasa/metabolismo , Fosfoenolpiruvato/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Ciclo del Ácido Cítrico , Secreción de Insulina , Insulinoma/enzimología , Insulinoma/metabolismo , Insulinoma/patología , Islotes Pancreáticos/citología , Islotes Pancreáticos/enzimología , Islotes Pancreáticos/metabolismo , Ratones , Mitocondrias/metabolismo , Modelos Biológicos , Fosfoenolpiruvato Carboxilasa/genética , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Succinato-CoA Ligasas/metabolismo
16.
Cell Metab ; 32(5): 751-766.e11, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33147485

RESUMEN

The mitochondrial GTP (mtGTP)-dependent phosphoenolpyruvate (PEP) cycle couples mitochondrial PEPCK (PCK2) to pyruvate kinase (PK) in the liver and pancreatic islets to regulate glucose homeostasis. Here, small molecule PK activators accelerated the PEP cycle to improve islet function, as well as metabolic homeostasis, in preclinical rodent models of diabetes. In contrast, treatment with a PK activator did not improve insulin secretion in pck2-/- mice. Unlike other clinical secretagogues, PK activation enhanced insulin secretion but also had higher insulin content and markers of differentiation. In addition to improving insulin secretion, acute PK activation short-circuited gluconeogenesis to reduce endogenous glucose production while accelerating red blood cell glucose turnover. Four-week delivery of a PK activator in vivo remodeled PK phosphorylation, reduced liver fat, and improved hepatic and peripheral insulin sensitivity in HFD-fed rats. These data provide a preclinical rationale for PK activation to accelerate the PEP cycle to improve metabolic homeostasis and insulin sensitivity.


Asunto(s)
Mitocondrias/metabolismo , Fosfoenolpiruvato/metabolismo , Animales , Homeostasis , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piruvato Quinasa/metabolismo , Ratas , Ratas Sprague-Dawley
17.
Cell Rep Med ; 1(7): 100120, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33103129

RESUMEN

Blood-borne factors regulate adult hippocampal neurogenesis and cognition in mammals. We report that elevating circulating unacylated-ghrelin (UAG), using both pharmacological and genetic methods, reduced hippocampal neurogenesis and plasticity in mice. Spatial memory impairments observed in ghrelin-O-acyl transferase-null (GOAT-/-) mice that lack acyl-ghrelin (AG) but have high levels of UAG were rescued by acyl-ghrelin. Acyl-ghrelin-mediated neurogenesis in vitro was dependent on non-cell-autonomous BDNF signaling that was inhibited by UAG. These findings suggest that post-translational acylation of ghrelin is important to neurogenesis and memory in mice. To determine relevance in humans, we analyzed circulating AG:UAG in Parkinson disease (PD) patients diagnosed with dementia (PDD), cognitively intact PD patients, and controls. Notably, plasma AG:UAG was only reduced in PDD. Hippocampal ghrelin-receptor expression remained unchanged; however, GOAT+ cell number was reduced in PDD. We identify UAG as a regulator of hippocampal-dependent plasticity and spatial memory and AG:UAG as a putative circulating diagnostic biomarker of dementia.


Asunto(s)
Aciltransferasas/genética , Ghrelina/análogos & derivados , Ghrelina/genética , Hipocampo/metabolismo , Proteínas de la Membrana/genética , Enfermedad de Parkinson/genética , Parálisis Supranuclear Progresiva/genética , Aciltransferasas/deficiencia , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Cognición/fisiología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Ghrelina/metabolismo , Hipocampo/patología , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis/genética , Plasticidad Neuronal/genética , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Cultivo Primario de Células , Ratas , Transducción de Señal , Memoria Espacial/fisiología , Parálisis Supranuclear Progresiva/metabolismo , Parálisis Supranuclear Progresiva/patología
18.
J Neuroendocrinol ; 31(7): e12696, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30742723

RESUMEN

Information about metabolic status arrives in the brain in the form of a complex milieu of circulating signalling factors, including glucose and fatty acids, ghrelin, leptin and insulin. The specific interactions between humoural factors, brain sites of action and how they influence behaviour are largely unknown. We have previously observed interactions between glucose availability and the actions of ghrelin mediated via the agouti-related peptide neurones of the hypothalamus. In the present study, we examine whether these effects generalise to another ghrelin-sensitive brain nucleus, the ventral tegmental area (VTA). We altered glucose availability by injecting mice with glucose or 2-deoxyglucose i.p. to induce hyperglycaemia and glucopenia, respectively. Thirty minutes later, we injected ghrelin in the VTA. Glucose administration suppressed intra-VTA ghrelin-induced feeding. Leptin, a longer-term signal of positive energy balance, did not affect intra-VTA ghrelin-induced feeding. 2-Deoxyglucose and ghrelin both increased food intake in their own right and, together, they additively increased feeding. These results add support to the idea that calculation of metabolic need depends on multiple signals across multiple brain regions and identifies that VTA circuits are sensitive to the integration of signals reflecting internal homeostatic state and influencing food intake.


Asunto(s)
Ingestión de Alimentos/fisiología , Ghrelina/fisiología , Glucosa/administración & dosificación , Área Tegmental Ventral/fisiología , Animales , Glucemia/efectos de los fármacos , Desoxiglucosa/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Ghrelina/administración & dosificación , Ratones Endogámicos C57BL , Área Tegmental Ventral/efectos de los fármacos
19.
Endocrinology ; 159(11): 3605-3614, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30204871

RESUMEN

Metabolic feedback from the periphery to the brain results from a dynamic physiologic fluctuation of nutrients and hormones, including glucose and fatty acids, ghrelin, leptin, and insulin. The specific interactions between humoral factors and how they influence feeding is largely unknown. We hypothesized that acute glucose availability may alter how the brain responds to ghrelin, a hormonal signal of energy availability. Acute glucose administration suppressed a range of ghrelin-induced behaviors as well as gene expression changes in hypothalamic neuropeptide Y (NPY) and agouti-related peptide (AgRP) neurons after ghrelin administration. Knockdown of the energy-sensing molecule AMP-activated protein kinase (AMPK) in AgRP neurons resulted in loss of the glucose effect, and mice responded as though pretreated with saline. Conversely, 2-deoxyglucose (2-DG), which decreases glucose availability, potentiated ghrelin-induced feeding and increased hypothalamic NPY mRNA levels. AMPK knockdown did not alter the additive effect of 2-DG and ghrelin on feeding. Our findings support the idea that computation of energy status is dynamic, is informed by multiple signals, and responds to acute fluctuations in metabolic state. These observations are broadly relevant to the investigation of neuroendocrine control of feeding and highlight the underappreciated complexity of control within these systems.


Asunto(s)
Proteína Relacionada con Agouti/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Ghrelina/farmacología , Glucosa/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Proteína Relacionada con Agouti/genética , Animales , Antimetabolitos/farmacología , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Desoxiglucosa/farmacología , Técnicas de Silenciamiento del Gen , Hipotálamo/citología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Neuropéptido Y/genética , ARN Mensajero/metabolismo
20.
Endocrinology ; 159(6): 2473-2483, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29697769

RESUMEN

Behavioral adaptation to periods of varying food availability is crucial for survival, and agouti-related protein (AgRP) neurons have been associated with entrainment to temporal restricted feeding. We have shown that carnitine acetyltransferase (Crat) in AgRP neurons enables metabolic flexibility and appropriate nutrient partitioning. In this study, by restricting food availability to 3 h/d during the light phase, we examined whether Crat is a component of a food-entrainable oscillator (FEO) that helps link behavior to food availability. AgRP Crat knockout (KO) mice consumed less food and regained less body weight but maintained blood glucose levels during the 25-day restricted feeding protocol. Importantly, we observed no difference in meal latency, food anticipatory activity (FAA), or brown adipose tissue temperature during the first 13 days of restricted feeding. However, as the restricted feeding paradigm progressed, we noticed an increased FAA in AgRP Crat KO mice. The delayed increase in FAA, which developed during the last 12 days of restricted feeding, corresponded with elevated plasma levels of corticosterone and nonesterified fatty acids, indicating it resulted from greater energy debt incurred by KO mice over the course of the experiment. These experiments highlight the importance of Crat in AgRP neurons in regulating feeding behavior and body weight gain during restricted feeding but not in synchronizing behavior to food availability. Thus, Crat within AgRP neurons forms a component of the homeostatic response to restricted feeding but is not likely to be a molecular component of FEO.


Asunto(s)
Adaptación Fisiológica/genética , Proteína Relacionada con Agouti/metabolismo , Restricción Calórica , Carnitina O-Acetiltransferasa/fisiología , Conducta Alimentaria/fisiología , Homeostasis/genética , Neuronas/metabolismo , Animales , Carnitina O-Acetiltransferasa/genética , Carnitina O-Acetiltransferasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Condicionamiento Físico Animal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA