Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
Metabolism ; 157: 155940, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38878857

RESUMEN

BACKGROUND AND AIM: Although it is well established that hormones like glucagon stimulates gluconeogenesis via the PKA-mediated phosphorylation of CREB and dephosphorylation of the cAMP-regulated CREB coactivators CRTC2, the role of neural signals in the regulation of gluconeogenesis remains uncertain. METHODS AND RESULTS: Here, we characterize the noradrenergic bundle architecture in mouse liver; we show that the sympathoexcitation induced by acute cold exposure promotes hyperglycemia and upregulation of gluconeogenesis via triggering of the CREB/CRTC2 pathway. Following its induction by dephosphorylation, CRTC2 translocates to the nucleus and drives the transcription of key gluconeogenic genes. Rodents submitted to different models of sympathectomy or knockout of CRTC2 do not activate gluconeogenesis in response to cold. Norepinephrine directly acts in hepatocytes mainly through a Ca2+-dependent pathway that stimulates CREB/CRTC2, leading to activation of the gluconeogenic program. CONCLUSION: Our data demonstrate the importance of the CREB/CRTC2 pathway in mediating effects of hepatic sympathetic inputs on glucose homeostasis, providing new insights into the role of norepinephrine in health and disease.


Asunto(s)
Frío , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Gluconeogénesis , Hígado , Norepinefrina , Factores de Transcripción , Animales , Gluconeogénesis/fisiología , Hígado/metabolismo , Ratones , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Masculino , Norepinefrina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Neuronas Adrenérgicas/metabolismo , Neuronas Adrenérgicas/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/fisiología , Hepatocitos/metabolismo
2.
Food Res Int ; 179: 114021, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38342541

RESUMEN

Sheep milk is rich in fat, protein, vitamins and minerals and is also one of the most important sources of natural bioactives. Several biopeptides in sheep milk have been reported to possess antibacterial, antiviral and anti-inflammatory properties, and they may prevent type 2 diabetes (T2D), disease and cancer. However, the precise mechanism(s) underlying the protective role of sheep milk against T2D development remains unclear. Therefore, in the current study, we investigated the effect of sheep milk on insulin resistance and glucose intolerance in high-fat diet (HFD)-fed mice, by conducting intraperitoneal glucose tolerance tests, metabolic cage studies, genomic sequencing, polymerase chain reaction, and biochemical assays. Hyperinsulinemic-euglycemic clamp-based experiments revealed that mice consuming sheep milk exhibited lower hepatic glucose production than mice in the control group. These findings further elucidate the mechanism by which dietary supplementation with sheep milk alleviates HFD-induced systemic glucose intolerance.


Asunto(s)
Diabetes Mellitus Tipo 2 , Intolerancia a la Glucosa , Resistencia a la Insulina , Ovinos , Ratones , Animales , Dieta Alta en Grasa/efectos adversos , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/prevención & control , Diabetes Mellitus Tipo 2/prevención & control , Leche/metabolismo
3.
Am J Physiol Endocrinol Metab ; 326(4): E428-E442, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38324258

RESUMEN

Glucagon rapidly and profoundly stimulates hepatic glucose production (HGP), but for reasons that are unclear, this effect normally wanes after a few hours, despite sustained plasma glucagon levels. This study characterized the time course of glucagon-mediated molecular events and their relevance to metabolic flux in the livers of conscious dogs. Glucagon was either infused into the hepato-portal vein at a sixfold basal rate in the presence of somatostatin and basal insulin, or it was maintained at a basal level in control studies. In one control group, glucose remained at basal, whereas in the other, glucose was infused to match the hyperglycemia that occurred in the hyperglucagonemic group. Elevated glucagon caused a rapid (30 min) and largely sustained increase in hepatic cAMP over 4 h, a continued elevation in glucose-6-phosphate (G6P), and activation and deactivation of glycogen phosphorylase and synthase activities, respectively. Net hepatic glycogenolysis increased rapidly, peaking at 15 min due to activation of the cAMP/PKA pathway, then slowly returned to baseline over the next 3 h in line with allosteric inhibition by glucose and G6P. Glucagon's stimulatory effect on HGP was sustained relative to the hyperglycemic control group due to continued PKA activation. Hepatic gluconeogenic flux did not increase due to the lack of glucagon's effect on substrate supply to the liver. Global gene expression profiling highlighted glucagon-regulated activation of genes involved in cellular respiration, metabolic processes, and signaling, as well as downregulation of genes involved in extracellular matrix assembly and development.NEW & NOTEWORTHY Glucagon rapidly stimulates hepatic glucose production, but these effects are transient. This study links the molecular and metabolic flux changes that occur in the liver over time in response to a rise in glucagon, demonstrating the strength of the dog as a translational model to couple findings in small animals and humans. In addition, this study clarifies why the rapid effects of glucagon on liver glycogen metabolism are not sustained.


Asunto(s)
Glucagón , Insulina , Humanos , Perros , Animales , Glucagón/metabolismo , Insulina/metabolismo , Transcriptoma , Glucosa/metabolismo , Hígado/metabolismo , Gluconeogénesis/genética , Glucemia/metabolismo
4.
Int J Mol Sci ; 25(4)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38397103

RESUMEN

Hyperglycemia is a hallmark of type 2 diabetes (T2D). Metformin, the first-line drug used to treat T2D, maintains blood glucose within a normal range by suppressing hepatic glucose production (HGP). However, resistance to metformin treatment is developed in most T2D patients over time. Transforming growth factor beta 1 (TGF-ß1) levels are elevated both in the liver and serum of T2D humans and mice. Here, we found that TGF-ß1 treatment impairs metformin action on suppressing HGP via inhibiting AMPK phosphorylation at Threonine 172 (T172). Hepatic TGF-ß1 deficiency improves metformin action on glycemic control in high fat diet (HFD)-induced obese mice. In our hepatic insulin resistant mouse model (hepatic insulin receptor substrate 1 (IRS1) and IRS2 double knockout (DKO)), metformin action on glycemic control was impaired, which is largely improved by further deletion of hepatic TGF-ß1 (TKObeta1) or hepatic Foxo1 (TKOfoxo1). Moreover, blockade of TGF-ß1 signaling by chemical inhibitor of TGF-ß1 type I receptor LY2157299 improves to metformin sensitivity in mice. Taken together, our current study suggests that hepatic TGF-ß1 signaling impairs metformin action on glycemic control, and suppression of TGF-ß1 signaling could serve as part of combination therapy with metformin for T2D treatment.


Asunto(s)
Diabetes Mellitus Tipo 2 , Metformina , Humanos , Ratones , Animales , Metformina/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Control Glucémico , Glucosa/metabolismo
5.
Diabetes Obes Metab ; 26(3): 1033-1039, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38131252

RESUMEN

AIM: To examine the impact of increased hepatic glucose production (HGP) on the decrease in plasma glucose concentration caused by empagliflozin in individuals living with diabetes and in nondiabetic individuals. METHODS: A total of 36 individuals living with diabetes and 34 nondiabetic individuals were randomized to receive, in double-blind fashion, empagliflozin or matching placebo in a 2:1 treatment ratio. Following an overnight fast, HGP was measured with 3-3 H-glucose infusion before, at the start of, and 3 months after therapy with empagliflozin. RESULTS: On Day 1 of empagliflozin administration, the increase in urinary glucose excretion (UGE) in individuals with normal glucose tolerance was smaller than in those with impaired glucose tolerance and those living with diabetes, and was accompanied by an increase in HGP in all three groups. The amount of glucose returned to the systemic circulation as a result of the increase in HGP was smaller than that excreted by the kidney during the first 3 h after empagliflozin administration, resulting in a decrease in fasting plasma glucose (FPG) concentration. After 3 h, the increase in HGP was in excess of UGE, leading to a small increase in plasma glucose concentration, which reached a new steady state. After 12 weeks, the amount of glucose returned to the circulation due to the empagliflozin-induced increase in HGP was comparable with that excreted by the kidney in all three groups. CONCLUSION: The balance between UGE and increase in HGP immediately after sodium-glucose cotransporter-2 (SGLT2) inhibition determined the magnitude of decrease in FPG and the new steady state which was achieved. After 12 weeks, the increase in HGP caused by empagliflozin closely matched the amount of glucose excreted by the kidneys; thus, FPG level remained stable despite the continuous urinary excretion of glucose caused by SGLT2 inhibition.


Asunto(s)
Compuestos de Bencidrilo , Diabetes Mellitus Tipo 2 , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Compuestos de Bencidrilo/uso terapéutico , Glucemia , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucosa/metabolismo , Glucósidos , Hipoglucemiantes , Transportador 2 de Sodio-Glucosa , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
6.
Eur J Pharmacol ; 956: 175984, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37567458

RESUMEN

Glucose metabolism is reported to be regulated by the central nervous system, but it is unclear whether this regulation is altered in diabetes. We investigated whether regulation of glucose metabolism by central dopamine D2 receptors is altered in type 1 and type 2 diabetic models. Intracerebroventricular injections of both the dopamine D2 receptor agonist quinpirole and the antagonist l-sulpiride induced hyperglycemia in control mice, but not in streptozotocin (STZ)-induced diabetic mice, a type 1 diabetic model. Hyperglycemia induced by quinpirole or l-sulpiride was diminished following fasting and these drugs did not affect hyperglycemia in the pyruvate tolerance test. In addition, both quinpirole and l-sulpiride increased hepatic glucose-6-phosphatase (G6Pase) mRNA. In STZ-induced diabetic mice, dopamine and dopamine D2 receptor mRNA in the hypothalamus, which regulates glucose homeostasis, were decreased. Hepatic glycogen and G6Pase mRNA were also decreased in STZ-induced diabetic mice. Neither quinpirole nor l-sulpiride increased hepatic G6Pase mRNA in STZ-induced diabetic mice. In diet-induced obesity mice, a type 2 diabetic model, both quinpirole and l-sulpiride induced hyperglycemia, and hypothalamic dopamine and dopamine D2 receptor mRNA were not altered. These results indicate that (i) stimulation or blockade of dopamine D2 receptors causes hyperglycemia by increasing hepatic glycogenolysis, and (ii) stimulation or blockade of dopamine D2 receptors does not affect glucose levels in type 1 but does so in type 2 diabetic models. Moreover, hypothalamic dopaminergic function and hepatic glycogenolysis are decreased in the type 1 diabetic model, which reduces hyperglycemia induced by stimulation or blockade of dopamine D2 receptors.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Hiperglucemia , Ratones , Animales , Quinpirol/farmacología , Dopamina , Sulpirida/farmacología , Glucemia , Diabetes Mellitus Tipo 1/inducido químicamente , Receptores de Dopamina D2/metabolismo , Agonistas de Dopamina/farmacología , Receptores de Dopamina D1/metabolismo
7.
Trends Pharmacol Sci ; 44(7): 457-473, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37188578

RESUMEN

Metformin is the most prescribed drug for the treatment of type 2 diabetes mellitus (T2DM), but its mechanism of action has not yet been completely elucidated. Classically, the liver has been considered the major site of action of metformin. However, over the past few years, advances have unveiled the gut as an additional important target of metformin, which contributes to its glucose-lowering effect through new mechanisms of action. A better understanding of the mechanistic details of metformin action in the gut and the liver and its relevance in patients remains the challenge of present and future research and may impact drug development for the treatment of T2DM. Here, we offer a critical analysis of the current status of metformin-driven multiorgan glucose-lowering effects.


Asunto(s)
Diabetes Mellitus Tipo 2 , Metformina , Humanos , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Metformina/farmacología , Metformina/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hígado , Glucosa
8.
Diabetologia ; 66(7): 1322-1339, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37202506

RESUMEN

AIMS/HYPOTHESIS: Hyperglucagonaemia-stimulated hepatic glucose production (HGP) contributes to hyperglycaemia during type 2 diabetes. A better understanding of glucagon action is important to enable efficient therapies to be developed for the treatment of diabetes. Here, we aimed to investigate the role of p38 MAPK family members in glucagon-induced HGP and determine the underlying mechanisms by which p38 MAPK regulates glucagon action. METHODS: p38α, ß, γ and δ MAPK siRNAs were transfected into primary hepatocytes, followed by measurement of glucagon-induced HGP. Adeno-associated virus serotype 8 carrying p38α MAPK short hairpin RNA (shRNA) was injected into liver-specific Foxo1 knockout, liver-specific Irs1/Irs2 double knockout and Foxo1S273D knockin mice. Foxo1S273A knockin mice were fed a high-fat diet for 10 weeks. Pyruvate tolerance tests, glucose tolerance tests, glucagon tolerance tests and insulin tolerance tests were carried out in mice, liver gene expression profiles were analysed and serum triglyceride, insulin and cholesterol levels were measured. Phosphorylation of forkhead box protein O1 (FOXO1) by p38α MAPK in vitro was analysed by LC-MS. RESULTS: We found that p38α MAPK, but not the other p38 isoforms, stimulates FOXO1-S273 phosphorylation and increases FOXO1 protein stability, promoting HGP in response to glucagon stimulation. In hepatocytes and mouse models, inhibition of p38α MAPK blocked FOXO1-S273 phosphorylation, decreased FOXO1 levels and significantly impaired glucagon- and fasting-induced HGP. However, the effect of p38α MAPK inhibition on HGP was abolished by FOXO1 deficiency or a Foxo1 point mutation at position 273 from serine to aspartic acid (Foxo1S273D) in both hepatocytes and mice. Moreover, an alanine mutation at position 273 (Foxo1S273A) decreased glucose production, improved glucose tolerance and increased insulin sensitivity in diet-induced obese mice. Finally, we found that glucagon activates p38α through exchange protein activated by cAMP 2 (EPAC2) signalling in hepatocytes. CONCLUSIONS/INTERPRETATION: This study found that p38α MAPK stimulates FOXO1-S273 phosphorylation to mediate the action of glucagon on glucose homeostasis in both health and disease. The glucagon-induced EPAC2-p38α MAPK-pFOXO1-S273 signalling pathway is a potential therapeutic target for the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Proteína Quinasa 14 Activada por Mitógenos , Animales , Ratones , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Glucagón/metabolismo , Gluconeogénesis/genética , Glucosa/metabolismo , Hepatocitos/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Ratones Endogámicos C57BL , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Fosforilación
9.
Eur J Pharmacol ; 944: 175559, 2023 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-36764353

RESUMEN

Elevated plasma glucose concentration, as a consequence of excessive hepatic glucose production, plays a pivotal role in the development of diabetes. A chromogranin A-derived diabetogenic peptide Pancreastatin (PST) enhances hepatic glucose output leading to diabetes. Therefore, here we probed the role of PSTi8, a PST inhibitor in ameliorating diabetes by investigating the effect of high glucose (HG) or PST on glucose metabolism. Further, we also explored the action mechanism of the underlying anti-hyperglycemic effect of PSTi8. PSTi8 treatment rescue cultured L6 and HepG2 cells from HG and PST-induced insulin resistance, respectively. It also enhances insulin receptor kinase activity by interacting with the insulin receptor and enhancing GLUT4 translocation and glucose uptake. Thus, our in-silico and in-vitro data support the PST-dependent and independent activity of PSTi8. Additionally, PSTi8 treatment in streptozotocin-induced diabetic rats improved glucose tolerance by lowering blood glucose and plasma PST levels. Concomitantly, the treated animals exhibited reduced hepatic glucose production accompanied by downregulation of hepatic gluconeogenic genes PEPCK and G6Pase. PSTi8-treated rats also exhibited enhanced hepatic glycogen in line with reduced plasma glucagon concentrations. Consistently, improved plasma insulin levels in PSTi8-treated rats enhanced skeletal muscle glucose disposal via enhanced P-Akt expression. In summary, these findings suggest PSTi8 has anti-hyperglycemic properties with enhanced skeletal muscle glucose disposal and reduced hepatic gluconeogenesis both PST dependent as well as independent.


Asunto(s)
Diabetes Mellitus Experimental , Resistencia a la Insulina , Ratas , Animales , Glucosa/metabolismo , Cromogranina A , Receptor de Insulina , Estreptozocina , Diabetes Mellitus Experimental/metabolismo , Hígado/metabolismo , Hipoglucemiantes , Insulina/metabolismo
11.
J Ethnopharmacol ; 306: 116162, 2023 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-36646159

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Sanhuang Xiexin Decoction (SHXXD) is a classic prescription for the treatment of diabetes. Excessive hepatic glucose production (HGP) is a major determinant of the occurrence and development of diabetes. Inhibition of HGP can significantly improve type 2 diabetes mellitus (T2DM). AIM OF THE STUDY: To investigate the mechanism by which SHXXD inhibits HGP. MATERIALS AND METHODS: First, a mouse model of T2DM was established through high-fat diet (HFD) feeding combined with streptozotocin (STZ) injection to determine the pharmacodynamic effect of SHXXD in T2DM mice. Then, the possible pathways induced by SHXXD in the treatment of T2DM were predicted by network pharmacology combined with transcriptomics (including target prediction, network analysis and enrichment analysis). Finally, the specific mechanism of SHXXD was elucidated by in vitro experiments. RESULTS: In vivo experiments showed that SHXXD reduced fasting blood glucose and alleviated weight loss in T2DM mice. Improved glucose clearance rates and insulin sensitivity improve dyslipidemia, liver tissue structural abnormalities and inflammatory cell infiltration as well as increase glycogen storage in T2DM mice. The results of network pharmacology and transcriptome analysis showed that SHXXD contained 378 compounds and 2625 targets. In total, 292 intersection targets were identified between the differentially expressed genes (DEGs) of the liver tissue insulin resistance (IR) related dataset GSE23343. KEGG enrichment analysis showed that the insulin/PI3K-Akt/FoxO signaling pathway may be related to SHXXD-mediated improvements in T2DM. In vitro experimental results showed that SHXXD increased glucose consumption by HepG2-IR cells and improved their insulin sensitivity. RT‒qPCR and Western blotting results showed that SHXXD inhibited hepatic gluconeogenesis through the insulin/PI3K-Akt/FoxO signaling pathway by promoting IGFIR, PIK3R1 and AKT2 expression and subsequently inhibiting PEPCK and FBP1 expression via phosphorylation of Foxo1. In addition, PI3K/Akt deactivated p-GSK3ß through phosphorylation, thereby promoting GS expression and increasing glycogen synthesis. CONCLUSIONS: SHXXD can target the liver to cooperate with the insulin/PI3K-Akt/FoxO signaling pathway to inhibit HGP to alleviate T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Ratones , Animales , Glucosa/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Resistencia a la Insulina/fisiología , Transducción de Señal , Hígado , Glucógeno/metabolismo
12.
Am J Physiol Endocrinol Metab ; 324(2): E199-E208, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36652399

RESUMEN

It has been proposed that brain glucagon action inhibits glucagon-stimulated hepatic glucose production (HGP), which may explain, at least in part, why glucagon's effect on HGP is transient. However, the pharmacologic off-target effects of glucagon in the brain may have been responsible for previously observed effects. Therefore, the aim of this study was to determine if central glucagon action plays a physiologic role in the regulation of HGP. Insulin was maintained at baseline while glucagon was either infused into the carotid and vertebral arteries or into a peripheral (leg) vein at rates designed to increase glucagon in the head in one group, while keeping glucagon at the liver matched between groups. The extraction rate of glucagon across the head was high (double that of the liver), and hypothalamic cAMP increased twofold, in proportion to the exposure of the brain to increased glucagon, but HGP was not reduced by the increase in brain glucagon signaling, as had been suggested previously (the areas under the curve for HGP were 840 ± 14 vs. 871 ± 36 mg/kg/240 min in head vs. peripheral infusion groups, respectively). Central nervous system glucagon action reduced circulating free fatty acids and glycerol, and this was associated with a modest reduction in net hepatic gluconeogenic flux. However, offsetting autoregulation by the liver (i.e., a reciprocal increase in net hepatic glycogenolysis) prevented a change in HGP. Thus, while physiologic engagement of the brain by glucagon can alter hepatic carbon flux, it does not appear to be responsible for the transient fall in HGP that occurs following the stimulation of HGP during a square wave rise in glucagon.NEW & NOTEWORTHY Glucagon stimulates hepatic glucose production through its direct effects on the liver but may indirectly inhibit this process by acting on the brain. This was tested by delivering glucagon via the cerebral circulatory system. Central nervous system glucagon action reduced liver gluconeogenic flux, but glycogenolysis increased, resulting in no net change in hepatic glucose production. Surprisingly, brain glucagon also appeared to suppress lipolysis (plasma free fatty acid and glycerol levels were reduced).


Asunto(s)
Glucagón , Glucogenólisis , Glucemia/metabolismo , Encéfalo/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Glucagón/metabolismo , Gluconeogénesis , Glucosa/metabolismo , Glicerol/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Animales
13.
Front Immunol ; 13: 998947, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091076

RESUMEN

Hepatic glucose production (HGP) is fine-regulated via glycogenolysis or gluconeogenesis to maintain physiological concentration of blood glucose during fasting-feeding cycle. Aberrant HGP leads to hyperglycemia in obesity-associated diabetes. Adipose tissue cooperates with the liver to regulate glycolipid metabolism. During these processes, adipose tissue macrophages (ATMs) change their profiles with various physio-pathological settings, producing diverse effects on HGP. Here, we briefly review the distinct phenotypes of ATMs under different nutrition states including feeding, fasting or overnutrition, and detail their effects on HGP. We discuss several pathways by which ATMs regulate hepatic gluconeogenesis or glycogenolysis, leading to favorable or unfavorable metabolic consequences. Furthermore, we summarize emerging therapeutic targets to correct metabolic disorders in morbid obesity or diabetes based on ATM-HGP axis. This review puts forward the importance and flexibility of ATMs in regulating HGP, proposing ATM-based HGP modulation as a potential therapeutic approach for obesity-associated metabolic dysfunction.


Asunto(s)
Diabetes Mellitus , Glucosa , Tejido Adiposo/metabolismo , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Humanos , Hígado/metabolismo , Macrófagos/metabolismo , Obesidad/metabolismo
14.
Cell Rep ; 40(8): 111259, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-36001955

RESUMEN

Excessive hepatic glucose production contributes to the development of hyperglycemia and is a key feature of type 2 diabetes. Here, we report that activation of hepatocyte Rap1a suppresses gluconeogenic gene expression and glucose production, whereas Rap1a silencing stimulates them. Rap1a activation is suppressed in obese mouse liver, and restoring its activity improves glucose intolerance. As Rap1a's membrane localization and activation depends on its geranylgeranylation, which is inhibited by statins, we show that statin-treated hepatocytes and the human liver have lower active-Rap1a levels. Similar to Rap1a inhibition, statins stimulate hepatic gluconeogenesis and increase fasting blood glucose in obese mice. Geranylgeraniol treatment, which acts as the precursor for geranylgeranyl isoprenoids, restores Rap1a activity and improves statin-mediated glucose intolerance. Mechanistically, Rap1a activation induces actin polymerization, which suppresses gluconeogenesis by Akt-mediated FoxO1 inhibition. Thus, Rap1a regulates hepatic glucose homeostasis, and blocking its activity, via lowering geranylgeranyl isoprenoids, contributes to statin-induced glucose intolerance.


Asunto(s)
Diabetes Mellitus Tipo 2 , Intolerancia a la Glucosa , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Hiperglucemia , Animales , Diabetes Mellitus Tipo 2/metabolismo , Gluconeogénesis/genética , Glucosa/metabolismo , Intolerancia a la Glucosa/metabolismo , Hepatocitos/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hiperglucemia/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/metabolismo , Terpenos/metabolismo , Proteínas de Unión al GTP rap1/metabolismo
15.
Artículo en Inglés | MEDLINE | ID: mdl-36011893

RESUMEN

Pea protein is considered to be a high quality dietary protein source, but also it is an ideal raw material for the production of bioactive peptides. Although the hypoglycemic effect of pea protein hydrolysate (PPH) has been previously reported, the underlying mechanisms, in particular its effect on the hepatic gluconeogenesis, remain to be elucidated. In the present study, we found that PPH suppressed glucose production in mouse liver cell-line AML-12 cells. Although both of the gluconeogenic and insulin signaling pathways in the AML-12 cells could be regulated by PPH, the suppression of glucose production was dependent on the inhibition of the cAMP response element-binding protein (CREB)-mediated signaling in the gluconeogenic pathway, but not the activation of insulin signaling. Findings from the present study have unveiled a novel role of PPH underlying its anti-diabetic activity, which could be helpful to accelerate the development of functional foods and nutraceuticals using PPH as a starting material.


Asunto(s)
Leucemia Mieloide Aguda , Proteínas de Guisantes , Animales , Gluconeogénesis , Glucosa/metabolismo , Hepatocitos , Insulina/metabolismo , Hígado , Ratones , Proteínas de Guisantes/metabolismo
16.
J Cell Physiol ; 237(8): 3421-3432, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35822903

RESUMEN

Hepatic glucose production (HGP) is crucial for the maintenance of normal glucose homeostasis. Although hepatic insulin resistance contributes to excessive glucose production, its mechanism is not well understood. Here, we show that inositol polyphosphate multikinase (IPMK), a key enzyme in inositol polyphosphate biosynthesis, plays a role in regulating hepatic insulin signaling and gluconeogenesis both in vitro and in vivo. IPMK-deficient hepatocytes exhibit decreased insulin-induced activation of Akt-FoxO1 signaling. The expression of messenger RNA levels of phosphoenolpyruvate carboxykinase 1 (Pck1) and glucose 6-phosphatase (G6pc), key enzymes mediating gluconeogenesis, are increased in IPMK-deficient hepatocytes compared to wild type hepatocytes. Importantly, re-expressing IPMK restores insulin sensitivity and alleviates glucose production in IPMK-deficient hepatocytes. Moreover, hepatocyte-specific IPMK deletion exacerbates hyperglycemia and insulin sensitivity in mice fed a high-fat diet, accompanied by an increase in HGP during pyruvate tolerance test and reduction in Akt phosphorylation in IPMK deficient liver. Our results demonstrate that IPMK mediates insulin signaling and gluconeogenesis and may be potentially targeted for treatment of diabetes.


Asunto(s)
Glucosa , Resistencia a la Insulina , Insulina , Hígado , Fosfotransferasas (Aceptor de Grupo Alcohol) , Animales , Proteína Forkhead Box O1/metabolismo , Glucosa/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Hepatocitos/metabolismo , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Ratones , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
17.
Eur J Pharmacol ; 928: 175108, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-35718128

RESUMEN

We investigated whether (E)-5-hydroxy-7-methoxy-3-(2-hydroxybenzyl)-4-chromanone (HM-chromanone) could suppress the transcription factors expression and enzymes involved in glucose production by activating AMPK in hepatocytes. HepG2 cells were treated with a medium containing HM-chromanone (5-100 µM), compound C (10 µM) and insulin (100 nM). Glucose production and glycogen synthesis assay were determined using a glucose assay kit and glycogen assay kit, respectively. Activities of AMP-activated protein kinase (AMPK), acetyl CoA carboxylase (ACC), cAMP response element-binding protein (CREB), PPAR coactivator-1α (PGC1α), CREB-regulated transcription coactivator 2 (CRTC2), Glycogen synthase kinase (GSK3ß), Phosphoenolpyruvate carboxykinase (PEPCK), glycogen synthase (GS), Glucose 6-phosphatase (G6pase) and ß-actin were determined by Western blot analysis. HM-chromanone significantly inhibited hepatic glucose production and increased glycogen synthesis by activating glycogen synthase. HM-chromanone induced the phosphorylation of CRTC2 and GSK-3ß by phosphorylating AMPK in HepG2 cells, which was confirmed by compound C. Furthermore, it significantly decreased the phosphorylation of CREB in a time- and concentration-dependent manner, and the effect was reversed in the presence of compound C. Therefore, the complex formation of CRTC2 and CREB was inhibited. HM-chromanone inhibited the expression of PGC-1α, PEPCK, and G6Pase genes involved in production of hepatic glucose. The results showed that HM-chromanone activates AMPK in a time and concentration dependent manner, thus suppressing hepatic glucose production and increasing glycogen synthesis in HepG2 cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Glucosa , Proteínas Quinasas Activadas por AMP/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Gluconeogénesis , Glucosa/metabolismo , Glucógeno/metabolismo , Glucógeno Sintasa/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Células Hep G2 , Humanos , Hipoglucemiantes/farmacología , Isoflavonas , Hígado/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosforilación
18.
J Tradit Complement Med ; 12(2): 195-205, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35528476

RESUMEN

Chronic insulin resistance suppresses muscle and liver response to insulin, which is partially due to impaired vesicle trafficking. We report here that a formula consisting of resveratrol, ferulic acid and epigallocatechin-3-O-gallate is more effective in ameliorating muscle and hepatic insulin resistance than the anti-diabetic drugs, metformin and AICAR. The formula enhanced glucose transporter-4 (GLUT4) translocation to the plasma membrane in the insulin-resistant muscle cells by regulating both insulin-independent (calcium and AMPK) and insulin-dependent (PI3K) signaling molecules. Particularly, it regulated the subcellular location of GLUT4 through endosomes to increase glucose uptake under insulin-resistant condition. Meanwhile, this phytochemicals combination increased glycogen synthesis and decreased glucose production in the insulin-resistant liver cells. On the other hand, this formula also showed anti-diabetic potential by the reduction of lipid content in the myotubes, hepatocytes, and adipocytes. This study demonstrated that the three phenolic compounds in the formula could work in distinct mechanisms and enhance both insulin-dependent and independent vesicles trafficking and glucose transport mechanisms to improve carbohydrate and lipid metabolism.

19.
Metabolism ; 131: 155199, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35390439

RESUMEN

AIMS/HYPOTHESIS: Roux-en Y gastric bypass surgery (GB) and sleeve gastrectomy (SG) alter prandial glucose metabolism, producing lower nadir glucose values and predisposing susceptible individuals to prandial hypoglycemia. The glycemic phenotype of GB or SG is associated with prandial hyperinsulinemia and hyperglucagonemia along with an increased influx of ingested glucose. Following insulin-induced hypoglycemia, glucagon is the most important stimulus for hepatic glucose production (HGP). It is unclear whether prandial hyperglucagonemia after GB or SG changes HGP under hyperinsulinemic hypoglycemia conditions. This study examined the hypothesis that prandial glucose production is reduced after GB and SG during hypoglycemia. METHODS: Glucose kinetics and islet-cell and gut hormone secretion during hyperinsulinemic (120 mU.m-2.min-1) hypoglycemic clamp (~3.2 mM) were measured before and after mixed meal ingestion in 9 non-diabetic subjects with GB, 7 with SG, and 5 matched non-operated controls (CN). RESULTS: Systemic appearance of ingested glucose was faster in GB compared to SG, and in SG compared to CN (p < 0.05). Subjects with GB and SG had greater plasma glucagon levels after eating (AUCGlucagon) compared to CN (p < 0.05). But prandial HGP response during insulin-induced hypoglycemia (AUCHGP) was smaller and shorter in duration in surgical groups (p < 0.05). In the absence of meal stimuli, however, glucose counterregulatory response to hypoglycemia was comparable among the 3 groups during hyperinsulinemic clamp. CONCLUSION: After bariatric surgery, prandial glucose counterregulatory response to hypoglycemia is impaired. Considering post-meal hyperglucagonemia after GB or SG the blunted HGP response suggests a lower sensitivity of liver to glucagon that can predispose to hypoglycemia in this population.


Asunto(s)
Derivación Gástrica , Hiperinsulinismo , Hipoglucemia , Glucemia/metabolismo , Gastrectomía/efectos adversos , Glucagón , Glucosa/metabolismo , Humanos , Hiperinsulinismo/etiología , Hipoglucemia/etiología , Insulina/metabolismo , Hígado/metabolismo
20.
Curr Diabetes Rev ; 18(9): e100222201016, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35142270

RESUMEN

Diabetes mellitus is the most prevalent malady, becoming a leading public health concern around the world. It is a chronic endocrine metabolic disturbance that is accompanied by the commencement of a sequence of complications. The liver primarily serves as the body's glucose or fuel reserve and also maintains standard blood sugar concentration. Hepatic gluconeogenesis, glycolysis and glycogenesis are the key contributors to fasting or post-prandial hyperglycaemia in type 2 diabetes mellitus subjects. So, regulating these channels could be a viable approach for mitigating hyperglycaemia in type 2 diabetes mellitus. Few potential synthetic drugs that precisely target hepatic glucose-producing metabolic pathways are presently available, but they have some serious negative effects like hypoglycaemia, hepatosteatosis and lactic acidosis. Therefore, scientists have veered to herbal products because of their edible nature, costeffectiveness and fewer side effects. Natural products and their isolated phytochemicals are progressively being employed to manage hyperglycaemia by modulating the enzyme's activity and regulating transcription factors concerned with hepatic glucose synthesis. We reviewed the potential effects of isolated bioactive phytochemicals on interesting targets that affect hepatic glucose homeostasis in diabetes. This study illustrates the benefit and feasibility of developing liver-specific drugs through secondary metabolites to restore hyperglycaemia.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperglucemia , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Humanos , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Hígado/metabolismo , Fitoquímicos/farmacología , Fitoquímicos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA