Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.256
Filter
1.
FASEB J ; 38(15): e23880, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39132919

ABSTRACT

Long noncoding RNAs (lncRNAs) are strongly associated with glucose homeostasis, but their roles remain largely unknown. In this study, the potential role of lncRNA-Snhg3 in glucose metabolism was evaluated both in vitro and in vivo. Here, we found a positive relationship between Snhg3 and hepatic glycogenesis. Glucose tolerance improved in hepatocyte-specific Snhg3 knock-in (Snhg3-HKI) mice, while it worsened in hepatocyte-specific Snhg3 knockout (Snhg3-HKO) mice. Furthermore, hepatic glycogenesis had shown remarkable increase in Snhg3-HKI mice and reduction in Snhg3-HKO mice, respectively. Mechanistically, Snhg3 increased mRNA and protein expression levels of PPP1R3B through inducing chromatin remodeling and promoting the phosphorylation of protein kinase B. Collectively, these results suggested that lncRNA-Snhg3 plays a critical role in hepatic glycogenesis.


Subject(s)
Liver , RNA, Long Noncoding , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mice , Liver/metabolism , Mice, Knockout , Glucose/metabolism , Male , Hepatocytes/metabolism , Mice, Inbred C57BL , Liver Glycogen/metabolism
2.
Nutrients ; 16(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39064628

ABSTRACT

BACKGROUND: In MASLD (formerly called NAFLD) mouse models, oversupply of dietary fat and sugar is more lipogenic than either nutrient alone. Fatty acids suppress de novo lipogenesis (DNL) from sugars, while DNL inhibits fatty acid oxidation. How such factors interact to impact hepatic triglyceride levels are incompletely understood. METHODS: Using deuterated water, we measured DNL in mice fed 18-weeks with standard chow (SC), SC supplemented with 55/45-fructose/glucose in the drinking water at 30% (w/v) (HS), high-fat chow (HF), and HF with HS supplementation (HFHS). Liver glycogen levels and its sources were also measured. For HS and HFHS mice, pentose phosphate (PP) fluxes and fructose contributions to DNL and glycogen were measured using [U-13C]fructose. RESULTS: The lipogenic diets caused significantly higher liver triglyceride levels compared to SC. DNL rates were suppressed in HF compared to SC and were partially restored in HFHS but supplied a minority of the additional triglyceride in HFHS compared to HF. Fructose contributed a significantly greater fraction of newly synthesized saturated fatty acids compared to oleic acid in both HS and HFHS. Glycogen levels were not different between diets, but significant differences in Direct and Indirect pathway contributions to glycogen synthesis were found. PP fluxes were similar in HS and HFHS mice and were insufficient to account for DNL reducing equivalents. CONCLUSIONS: Despite amplifying the lipogenic effects of fat, the fact that sugar-activated DNL per se barely contributes suggests that its role is likely more relevant in the inhibition of fatty acid oxidation. Fructose promotes lipogenesis of saturated over unsaturated fatty acids and contributes to maintenance of glycogen levels. PP fluxes associated with sugar conversion to fat account for a minor fraction of DNL reducing equivalents.


Subject(s)
Diet, High-Fat , Fructose , Lipogenesis , Liver Glycogen , Liver , Mice, Inbred C57BL , Postprandial Period , Triglycerides , Animals , Triglycerides/metabolism , Triglycerides/blood , Lipogenesis/drug effects , Male , Liver/metabolism , Mice , Liver Glycogen/metabolism , Fructose/administration & dosage , Fatty Acids/metabolism , Dietary Sugars/administration & dosage , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Glucose/metabolism
3.
Pak J Pharm Sci ; 37(2(Special)): 459-462, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38822550

ABSTRACT

The purpose of this study was to examine the potential hypoglycemic effects of administering ginger (Zingiber officinale) and garlic (Allium sativum) to rats with induced type 2 diabetes. A total of forty-five male adult albino rats were randomly assigned to five groups. The groups were named Normal Control, Diabetic Control, Ginger group, Garlic group and a combination group of ginger and garlic. Diabetes was produced in all groups, except the normal control group, using an intraperitoneal injection of streptozotocin at a dosage of 60 mg/body weight. During the course of two months, rats were administered varying amounts of ginger and garlic powders as part of their treatment After the experiment concluded, measurements were taken for glycated hemoglobin, serum glucose, insulin, cholesterol, high density protein, low density protein and liver glycogen levels. These groups exhibited considerably greater serum insulin and high-density lipoprotein concentrations (P<0.05) compared to the diabetic control group. Conversely, body weight, fasting blood glucose, total cholesterol, low density lipoprotein, and glycated hemoglobin levels were significantly lower (P<0.05) in all groups compared to the diabetic control group. A statistically significant increase (P<0.05) increase shown in liver glycogen levels. This study proposes that the utilization of ginger and garlic powders improve the condition of type 2 diabetes and maybe reduce the risk of subsequent diabetic complications.


Subject(s)
Blood Glucose , Diabetes Mellitus, Experimental , Garlic , Hypoglycemic Agents , Insulin , Powders , Zingiber officinale , Animals , Garlic/chemistry , Zingiber officinale/chemistry , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/blood , Male , Blood Glucose/drug effects , Blood Glucose/metabolism , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Rats , Insulin/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/blood , Glycated Hemoglobin/metabolism , Plant Extracts/pharmacology , Phytotherapy , Liver Glycogen/metabolism , Streptozocin
4.
Pak J Pharm Sci ; 37(2): 321-326, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38767099

ABSTRACT

Fatigue is a serious disturbance to human health, especially in people who have a severe disease such as cancer, or have been infected with COVID-19. Our research objective is to evaluate the anti-fatigue effect and mechanism of icariin through a mouse experimental model. Mice were treated with icariin for 30 days and anti-fatigue effects were evaluated by the weight-bearing swimming test, serum urea nitrogen test, lactic acid accumulation and clearance test in blood and the amount of liver glycogen. The protein expression levels of adenosine monophosphate-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC1-α) in the skeletal muscle of mice in each group were measured by western blotting. Results showed that icariin prolonged the weight-bearing swimming time of animals, reduced the serum urea nitrogen level after exercise, decreased the blood lactic acid concentration after exercise and increased the liver glycogen content observably. Compared to that in the control group, icariin upregulated AMPK and PGC1-α expression in skeletal muscle. Icariin can improve fatigue resistance in mice and its mechanism may be through improving the AMPK/PGC-1α pathway in skeletal muscle to enhance energy synthesis, decreasing the accumulation of metabolites and slowing glycogen consumption and decomposition.


Subject(s)
Blood Urea Nitrogen , Fatigue , Flavonoids , Lactic Acid , Muscle, Skeletal , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Animals , Flavonoids/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Mice , Male , Lactic Acid/blood , Lactic Acid/metabolism , Fatigue/drug therapy , Fatigue/metabolism , Swimming , AMP-Activated Protein Kinases/metabolism , Glycogen/metabolism , Liver/drug effects , Liver/metabolism , Liver Glycogen/metabolism
5.
Mol Metab ; 84: 101942, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642890

ABSTRACT

OBJECTIVE: Hepatic glucose metabolism is profoundly perturbed by excessive alcohol intake. miR-141/200c expression is significantly induced by chronic ethanol feeding. This study aimed at identifying the role of miR-141/200c in glucose homeostasis during chronic ethanol exposure. METHODS: WT and miR-141/200c KO mice were fed a control or an ethanol diet for 30 days, followed by a single binge of maltose dextrin or ethanol, respectively. Untargeted metabolomics analysis of hepatic primary metabolites was performed along with analyses for liver histology, gene expression, intracellular signaling pathways, and physiological relevance. Primary hepatocytes were used for mechanistic studies. RESULTS: miR-141/200c deficiency rewires hepatic glucose metabolism during chronic ethanol feeding, increasing the abundance of glucose intermediates including G6P, an allosteric activator for GS. miR-141/200c deficiency replenished glycogen depletion during chronic ethanol feeding accompanied by reduced GS phosphorylation in parallel with increased expression of PP1 glycogen targeting subunits. Moreover, miR-141/200c deficiency prevented ethanol-mediated increases in AMPK and CaMKK2 activity. Ethanol treatment reduced glycogen content in WT-hepatocytes, which was reversed by dorsomorphin, a selective AMPK inhibitor, while KO-hepatocytes displayed higher glycogen content than WT-hepatocytes in response to ethanol treatment. Furthermore, treatment of hepatocytes with A23187, a calcium ionophore activating CaMKK2, lowered glycogen content in WT-hepatocytes. Notably, the suppressive effect of A23187 on glycogen deposition was reversed by dorsomorphin, demonstrating that the glycogen depletion by A23187 is mediated by AMPK. KO-hepatocytes exhibited higher glycogen content than WT-hepatocytes in response to A23187. Finally, miR-141/200c deficiency led to improved glucose tolerance and insulin sensitivity during chronic ethanol feeding. CONCLUSIONS: miR-141/200c deficiency replenishes ethanol-mediated hepatic glycogen depletion through the regulation of GS activity and calcium signaling coupled with the AMPK pathway, improving glucose homeostasis and insulin sensitivity. These results underscore miR-141/200c as a potential therapeutic target for the management of alcohol intoxication.


Subject(s)
Ethanol , Hepatocytes , Liver Glycogen , Liver , Mice, Knockout , MicroRNAs , Animals , Ethanol/pharmacology , Mice , MicroRNAs/metabolism , MicroRNAs/genetics , Hepatocytes/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Male , Mice, Inbred C57BL , Glucose/metabolism
6.
Int J Biol Macromol ; 268(Pt 1): 131741, 2024 May.
Article in English | MEDLINE | ID: mdl-38649083

ABSTRACT

Glycogen, a complex branched glucose polymer, is responsible for sugar storage in blood glucose homeostasis. It comprises small ß particles bound together into composite α particles. In diabetic livers, α particles are fragile, breaking apart into smaller particles in dimethyl sulfoxide, DMSO; they are however stable in glycogen from healthy animals. We postulate that the bond between ß particles in α particles involves hydrogen bonding. Liver-glycogen fragility in normal and db/db mice (an animal model for diabetes) is compared using various hydrogen-bond breakers (DMSO, guanidine and urea) at different temperatures. The results showed different degrees of α-particle disruption. Disrupted glycogen showed changes in the mid-infra-red spectrum that are related to hydrogen bonds. While glycogen α-particles are only fragile under harsh, non-physiological conditions, these results nevertheless imply that the bonding between ß particles in α particles is different in diabetic livers compared to healthy, and is probably associated with hydrogen bonding.


Subject(s)
Hydrogen Bonding , Animals , Mice , Dimethyl Sulfoxide/chemistry , Liver Glycogen/metabolism , Urea/chemistry , Guanidine/chemistry , Guanidine/pharmacology , Liver/metabolism , Male
7.
Nutrients ; 16(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38674881

ABSTRACT

Anorexia nervosa (AN) induces organ dysfunction caused by malnutrition, including liver damage leading to a rise in transaminases due to hepatocyte damage. The underlying pathophysiology of starvation-induced liver damage is poorly understood. We investigate the effect of a 25% body weight reduction on murine livers in a mouse model and examine possible underlying mechanisms of starvation-induced liver damage. Female mice received a restricted amount of food with access to running wheels until a 25% weight reduction was achieved. This weight reduction was maintained for two weeks to mimic chronic starvation. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were measured spectrophotometrically. Liver fat content was analyzed using an Oil Red O stain, and liver glycogen was determined using a Periodic acid-Schiff (PAS) stain. Immunohistochemical stains were used to investigate macrophages, proliferation, apoptosis, and autophagy. Starvation led to an elevation of AST and ALT values, a decreased amount of liver fat, and reduced glycogen deposits. The density of F4/80+ macrophage numbers as well as proliferating KI67+ cells were decreased by starvation, while apoptosis was not altered. This was paralleled by an increase in autophagy-related protein staining. Increased transaminase values suggest the presence of liver damage in the examined livers of starved mice. The observed starvation-induced liver damage may be attributed to increased autophagy. Whether other mechanisms play an additional role in starvation-induced liver damage remains to be investigated.


Subject(s)
Alanine Transaminase , Aspartate Aminotransferases , Autophagy , Liver , Starvation , Animals , Female , Liver/metabolism , Liver/pathology , Mice , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Liver Diseases/etiology , Liver Diseases/pathology , Disease Models, Animal , Apoptosis , Macrophages/metabolism , Mice, Inbred C57BL , Liver Glycogen/metabolism
8.
Acta Pharm ; 74(1): 117-130, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38554383

ABSTRACT

Statin treatment may increase the risk of diabetes; there is insufficient data on how statins affect glucose regulation and glycemic control and the effects of statins on liver enzymes related to carbohydrate metabolism have not been fully studied. Therefore, we aimed to compare the effects of the statin derivatives, pravastatin, and rosuvastatin, on carbohydrate metabolism in an experimental diabetic rat model. Female Wistar albino rats were used and diabetes was induced by intraperitoneal injection of streptozotocin. Thereafter, 10 and 20 mg kg-1 day-1 doses of both pravastatin and rosuvastatin were administered by oral gavage to the diabetic rats for 8 weeks. At the end of the experiment, body masses, the levels of fasting blood glucose, serum insulin, insulin resistance (HOMA-IR), liver glycogen, and liver enzymes related to carbohydrate metabolism were measured. Both doses of pravastatin significantly in creased the body mass in diabetic rats, however, rosuvastatin, especially at the dose of 20 mg kg-1 day-1 reduced the body mass signi ficantly. Pravastatin, especially at a dose of 20 mg kg-1 day-1, caused significant increases in liver glycogen synthase and glucose 6-phosphate dehydrogenase levels but significant decreases in the levels of glycogen phosphorylase, lactate dehydrogenase, and glucose-6-phosphatase. Hence, pravastatin partially ameliorated the adverse changes in liver enzymes caused by diabetes and, especially at the dose of 20 mg kg-1 day-1, reduced the fasting blood glucose level and increased the liver glycogen content. However, rosuvastatin, especially at the dose of 20 mg kg-1 day-1, significantly reduced the liver glycogen synthase and pyruvate kinase levels, but increased the glycogen phosphorylase level in diabetic rats. Rosuvastatin, 20 mg kg-1 day-1 dose, caused significant decreases in the body mass and the liver glycogen content of diabetic rats. It can be concluded that pravastatin, especially at the dose of 20 mg kg-1 day-1 is more effective in ameliorating the negative effects of diabetes by modulating carbohydrate metabolism.


Subject(s)
Diabetes Mellitus, Experimental , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Female , Rats , Animals , Blood Glucose , Rats, Wistar , Rosuvastatin Calcium/adverse effects , Pravastatin/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Glycogen Synthase/metabolism , Glycogen Synthase/pharmacology , Liver Glycogen/adverse effects , Liver Glycogen/metabolism , Glycated Hemoglobin , Glucose/metabolism , Carbohydrate Metabolism , Glycogen Phosphorylase/metabolism , Glycogen Phosphorylase/pharmacology , Liver/metabolism , Insulin/pharmacology
9.
Science ; 383(6684): eadi3332, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38359126

ABSTRACT

The identification of mechanisms to store glucose carbon in the form of glycogen rather than fat in hepatocytes has important implications for the prevention of nonalcoholic fatty liver disease (NAFLD) and other chronic metabolic diseases. In this work, we show that glycogenesis uses its intermediate metabolite uridine diphosphate glucose (UDPG) to antagonize lipogenesis, thus steering both mouse and human hepatocytes toward storing glucose carbon as glycogen. The underlying mechanism involves transport of UDPG to the Golgi apparatus, where it binds to site-1 protease (S1P) and inhibits S1P-mediated cleavage of sterol regulatory element-binding proteins (SREBPs), thereby inhibiting lipogenesis in hepatocytes. Consistent with this mechanism, UDPG administration is effective at treating NAFLD in a mouse model and human organoids. These findings indicate a potential opportunity to ameliorate disordered fat metabolism in the liver.


Subject(s)
Lipogenesis , Liver Glycogen , Liver , Proprotein Convertases , Serine Endopeptidases , Uridine Diphosphate Glucose , Animals , Humans , Male , Mice , Carbon/metabolism , Glucose/metabolism , HEK293 Cells , Hepatocytes/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Proprotein Convertases/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Uridine Diphosphate Glucose/administration & dosage , Uridine Diphosphate Glucose/metabolism
10.
PLoS One ; 19(1): e0293627, 2024.
Article in English | MEDLINE | ID: mdl-38206915

ABSTRACT

INTRODUCTION: We evaluated the effect of Tucum-do-Cerrado on glucose metabolism homeostasis and its relationship with redox-inflammatory responses in a high-fat (HF) diet-induced obesity model. RESULTS: The HF diet increased energy intake, feed efficiency, body weight, muscle and hepatic glycogen, insulin, homeostatic model assessment of insulin resistance (HOMA IR) and beta (ß)-cell function, and gut catalase (CAT) activity, and decreased food intake, hepatic glutathione reductase (GR), glutathione peroxidase (GPX), glutathione S-transferase (GST), and superoxide dismutase (SOD) activities, hepatic phosphoenolpyruvate carboxykinase 1 (Pck1), and intestinal solute carrier family 5 member 1 (Slc5a1) mRNA levels compared with the control diet. However, the HF diet with Tucum-do-Cerrado decreased hepatic glycogen, and increased hepatic GR activity, hepatic Slc2a2 mRNA levels and serum Tnfa compared with the HF diet. Tucum-do-Cerrado decreased muscle glycogen, intestinal CAT and GPX activities, muscle PFK-1 and HK activities, and increased hepatic protein (CARB) and intestinal lipid (MDA) oxidation, hepatic GST activity, serum antioxidant potential, hepatic phosphofructokinase-1 (PFK-1) activity, intestinal solute carrier family 2 member 2 (Slc2a2), tumor necrosis factor (Tnf), interleukin-1 beta (Il1b), muscle protein kinase AMP-activated alpha 1 (Prkaa1), solute carrier family 2 member 2 (Slc2a2) mRNA levels, and serum interleukin-6 (IL-6) levels, regardless of diet type. CONCLUSION: Tucum-do-Cerrado consumption may ameliorate impaired glucose utilization in a HF diet-induced obesity model by increasing liver and muscle glucose uptake and oxidation. These data suggest that Tucum-do-Cerrado consumption improves muscle glucose oxidation in non-obese and obese rats. This response may be related to the improvement in the total antioxidant capacity of rats.


Subject(s)
Arecaceae , Glucose , Rats , Animals , Glucose/metabolism , Antioxidants/metabolism , Diet, High-Fat/adverse effects , Liver Glycogen/metabolism , Obesity/etiology , Obesity/metabolism , Liver/metabolism , Arecaceae/metabolism , RNA, Messenger/metabolism
11.
Int J Biol Macromol ; 260(Pt 1): 129491, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38228202

ABSTRACT

In this study, the impact of prenatal exposure to Epigallocatechin gallate (EGCG) on the liver of adult offspring mice was investigated. While EGCG is known for its health benefits, its effects of prenatal exposure on the liver remain unclear. Pregnant C57BL/6 J mice were exposed to 1 mg/kg of EGCG for 16 days to assess hepatotoxicity effects of adult offspring. Transcriptomics and metabolomics were employed to elucidate the hepatotoxicity mechanisms. The findings revealed that prenatal EGCG exposure led to a decrease in liver somatic index, enhanced inflammatory responses and disrupted liver function through increased glycogen accumulation in adult mice. The integrated omics analysis revealed significant alterations in key pathways involved in liver glucose lipid metabolism, such as gluconeogenesis, dysregulation of insulin signaling, and induction of liver inflammation. Furthermore, the study found a negative correlation between the promoter methylation levels of Ppara and their mRNA levels, suggesting that EGCG could reduce hepatic lipid content through epigenetic modifications. The findings suggest that prenatal EGCG exposure can have detrimental impacts on the liver among adult individuals and emphasize the need for a comprehensive evaluation of the potential risks associated with EGCG consumption during pregnancy.


Subject(s)
Catechin , Catechin/analogs & derivatives , Chemical and Drug Induced Liver Injury , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Female , Mice , Animals , Liver Glycogen/metabolism , Liver Glycogen/pharmacology , Lipid Metabolism , Prenatal Exposure Delayed Effects/metabolism , Mice, Inbred C57BL , Liver , Catechin/pharmacology , Catechin/metabolism , Gluconeogenesis , Chemical and Drug Induced Liver Injury/metabolism
12.
J Clin Invest ; 134(7)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38290087

ABSTRACT

In response to a meal, insulin drives hepatic glycogen synthesis to help regulate systemic glucose homeostasis. The mechanistic target of rapamycin complex 1 (mTORC1) is a well-established insulin target and contributes to the postprandial control of liver lipid metabolism, autophagy, and protein synthesis. However, its role in hepatic glucose metabolism is less understood. Here, we used metabolomics, isotope tracing, and mouse genetics to define a role for liver mTORC1 signaling in the control of postprandial glycolytic intermediates and glycogen deposition. We show that mTORC1 is required for glycogen synthase activity and glycogenesis. Mechanistically, hepatic mTORC1 activity promotes the feeding-dependent induction of Ppp1r3b, a gene encoding a phosphatase important for glycogen synthase activity whose polymorphisms are linked to human diabetes. Reexpression of Ppp1r3b in livers lacking mTORC1 signaling enhances glycogen synthase activity and restores postprandial glycogen content. mTORC1-dependent transcriptional control of Ppp1r3b is facilitated by FOXO1, a well characterized transcriptional regulator involved in the hepatic response to nutrient intake. Collectively, we identify a role for mTORC1 signaling in the transcriptional regulation of Ppp1r3b and the subsequent induction of postprandial hepatic glycogen synthesis.


Subject(s)
Glycogen Synthase , Liver Glycogen , Mechanistic Target of Rapamycin Complex 1 , Protein Phosphatase 1 , Animals , Humans , Mice , Glycogen/genetics , Glycogen/metabolism , Glycogen Synthase/metabolism , Insulin/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Phosphatase 1/metabolism , Postprandial Period
13.
Article in English | MEDLINE | ID: mdl-37898361

ABSTRACT

Brandt's vole (Lasiopodomys brandtii) is a species with hypoxia tolerance, and glucose serves as the primary energy substrate under hypoxia. However, the glucose supply in Brandt's voles under hypoxia has not been studied. This study aimed to investigate characteristics in physiological indices and liver gene expression associated with glucose supply in Brandt's voles under hypoxia. Serum glucose of Brandt's voles remained stable under 10% O2, increased under 7.5% O2, and decreased under 5% O2. Serum lactate increased under 10% O2, decreased under 7.5% O2, increased at 6 h and decreased at 12 h under 5% O2. Liver glycogen increased under 10% O2, remained constant under 7.5% O2, and reduced under 5% O2. Pepck and G6pase expression associated with gluconeogenesis decreased under 10% O2, while Pepck expression decreased and G6pase expression increased under 7.5% and 5% O2. Regarding genes related to glycogen metabolism, Gys expression decreased at all oxygen concentrations, Phk expression increased under 5% O2, and Gp expression increased under 7.5% and 5% O2. The alterations in glucose, lactate, liver glycogen, and gene expression related to glycogenolysis in Kunming mice (Mus musculus, control species) are similar to discovery of Brandt's voles under 7.5% O2, but gene expression involved in gluconeogenesis and glycogen synthesis increased. The findings suggest that Brandt's voles are more tolerant to hypoxia than Kunming mice, and their physiological indices and liver gene expression related to glucose supply exhibit species- and oxygen concentration-specific responses to hypoxia. This research offers novel insights for studying hypoxia tolerance of Brandt's voles.


Subject(s)
Glucose , Liver Glycogen , Mice , Animals , Glucose/metabolism , Liver Glycogen/metabolism , Liver , Arvicolinae/genetics , Lactates/metabolism , Gene Expression , Oxygen/metabolism
14.
Poult Sci ; 103(1): 103193, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37931402

ABSTRACT

Animal growth is closely related to glycolipid metabolism, and the liver is the main organ for glycogen storage and fat synthesis in birds, but whether monochromatic light affects glycogen and lipid synthesis in the liver is unclear. Therefore, in this study, a total of 96 Arbor Acre (AA) broilers at posthatching d 0 (P0) were raised under 4 kinds of light-emitting diode (LED) lights, white light (WL), red light (RL), green light (GL), and blue light (BL), to posthatching d 21 (P21) and 35 (P35). The results showed that the liver, abdominal fat, and abdominal fat indices gradually increased with increasing age under monochromatic light treatments. The liver glycogen and triglyceride (TG) contents also showed an increasing trend. Furthermore, compared with those at P21, the mRNA levels of glycogen synthase (GS), glycogen synthase kinase-3ß (GSK-3ß), and protein kinase B (AKT1) in the liver were increased in the WL and RL groups at P35, and the mRNA levels of acetyl-CoA carboxylase (ACC) and apolipoprotein B (APOB) increased in all groups at P35. At the same time, the total antioxidant capacity (T-AOC) and liver superoxide dismutase (SOD) contents increased in all groups at P35 compared with those at P21. In addition, at P21, compared with WL, GL and BL promoted the serum glucose (GLU) and TG contents by increasing the mRNA levels of GS, GSK-3ß, glucose-6-phosphatase (G6PC), ACC, and fatty acid synthase (FAS), but no effect on the proliferative ability and damage of hepatocytes. At P35, RL promoted the hepatic glycogen and TG contents by increasing GSK-3ß, AKT1, ACC, and APOB mRNA levels, and the serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were increased than in the WL group. These results suggest that the effects of light color on liver glycogen and lipid synthesis in broilers changed with age, and also provide a theoretical guidance for scientific use of color of light information to improve productive performance in broilers.


Subject(s)
Chickens , Liver Glycogen , Animals , Liver Glycogen/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Lipid Metabolism , RNA, Messenger/metabolism , Apolipoproteins B/metabolism , Lipids , Liver/metabolism
15.
Mol Metab ; 79: 101838, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37995884

ABSTRACT

OBJECTIVE: Carbohydrate Response Element Binding Protein (ChREBP) is a glucose 6-phosphate (G6P)-sensitive transcription factor that acts as a metabolic switch to maintain intracellular glucose and phosphate homeostasis. Hepatic ChREBP is well-known for its regulatory role in glycolysis, the pentose phosphate pathway, and de novo lipogenesis. The physiological role of ChREBP in hepatic glycogen metabolism and blood glucose regulation has not been assessed in detail, and ChREBP's contribution to carbohydrate flux adaptations in hepatic Glycogen Storage Disease type 1 (GSD I) requires further investigation. METHODS: The current study aimed to investigate the role of ChREBP as a regulator of glycogen metabolism in response to hepatic G6P accumulation, using a model for acute hepatic GSD type Ib. The immediate biochemical and regulatory responses to hepatic G6P accumulation were evaluated upon G6P transporter inhibition by the chlorogenic acid S4048 in mice that were either treated with a short hairpin RNA (shRNA) directed against ChREBP (shChREBP) or a scrambled shRNA (shSCR). Complementary stable isotope experiments were performed to quantify hepatic carbohydrate fluxes in vivo. RESULTS: ShChREBP treatment normalized the S4048-mediated induction of hepatic ChREBP target genes to levels observed in vehicle- and shSCR-treated controls. In parallel, hepatic shChREBP treatment in S4048-infused mice resulted in a more pronounced accumulation of hepatic glycogen and further reduction of blood glucose levels compared to shSCR treatment. Hepatic ChREBP knockdown modestly increased glucokinase (GCK) flux in S4048-treated mice while it enhanced UDP-glucose turnover as well as glycogen synthase and phosphorylase fluxes. Hepatic GCK mRNA and protein levels were induced by shChREBP treatment in both vehicle- and S4048-treated mice, while glycogen synthase 2 (GYS2) and glycogen phosphorylase (PYGL) mRNA and protein levels were reduced. Finally, knockdown of hepatic ChREBP expression reduced starch domain binding protein 1 (STBD1) mRNA and protein levels while it inhibited acid alpha-glucosidase (GAA) activity, suggesting reduced capacity for lysosomal glycogen breakdown. CONCLUSIONS: Our data show that ChREBP activation controls hepatic glycogen and blood glucose levels in acute hepatic GSD Ib through concomitant regulation of glucose phosphorylation, glycogenesis, and glycogenolysis. ChREBP-mediated control of GCK enzyme levels aligns with corresponding adaptations in GCK flux. In contrast, ChREBP activation in response to acute hepatic GSD Ib exerts opposite effects on GYS2/PYGL enzyme levels and their corresponding fluxes, indicating that GYS2/PYGL expression levels are not limiting to their respective fluxes under these conditions.


Subject(s)
Blood Glucose , Glycogen Storage Disease Type I , Animals , Mice , Carbohydrate Metabolism , Disease Models, Animal , Glucose/metabolism , Glucose-6-Phosphate/metabolism , Glycogen/metabolism , Glycogen Synthase/metabolism , Liver Glycogen/metabolism , Phosphates , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
J Appl Physiol (1985) ; 136(2): 233-243, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38126089

ABSTRACT

The carotid bodies (CBs) have been implicated in glucose abnormalities in obesity via elevation of activity of the sympathetic nervous system. Obesity-induced hypertension is mediated by insulin receptor (INSR) signaling and by leptin, which binds to the leptin receptor (LEPRb) in CB and activates transient receptor potential channel subfamily M member 7 (TRPM7). We hypothesize that in mice with diet-induced obesity, hyperglycemia, glucose intolerance, and insulin resistance will be attenuated by the CB denervation (carotid sinus nerve dissection, CSND) and by knockdown of Leprb, Trpm7, and Insr gene expression in CB. In series of experiments in 75 male diet-induced obese (DIO) mice, we performed either CSND (vs. sham) surgeries or shRNA-induced suppression of Leprb, Trpm7, or Insr gene expression in CB, followed by blood pressure telemetry, intraperitoneal glucose tolerance and insulin tolerance tests, and measurements of fasting plasma insulin, leptin, corticosterone, glucagon and free fatty acids (FFAs) levels, hepatic expression of gluconeogenesis enzymes phosphoenolpyruvate carboxykinase (PEPCK) and glucose 6-phosphatase (G-6-Pase) mRNA and liver glycogen levels. CSND decreased blood pressure, fasting blood glucose levels and improved glucose tolerance without any effect on insulin resistance. CSND did not affect any hormone levels and gluconeogenesis enzymes, but increased liver glycogen level. Genetic knockdown of CB Leprb, Trpm7, and Insr had no effect on glucose metabolism. We conclude that CB contributes to hyperglycemia of obesity, probably by modulation of the glycogen-glucose equilibrium. Diabetogenic effects of obesity on CB in mice do not occur via activation of CB Leprb, Trpm7, and Insr.NEW & NOTEWORTHY This paper provides first evidence that carotid body denervation abolishes hypertension and improves fasting blood glucose levels and glucose tolerance in mice with diet-induced obesity. Furthermore, we have shown that this phenomenon is associated with increased liver glycogen content, whereas insulin sensitivity and enzymes of gluconeogenesis were not affected.


Subject(s)
Carotid Body , Hyperglycemia , Hypertension , Insulin Resistance , Insulins , TRPM Cation Channels , Male , Mice , Animals , Leptin , Blood Glucose/metabolism , Carotid Body/metabolism , Mice, Obese , TRPM Cation Channels/metabolism , Liver Glycogen/metabolism , Hyperglycemia/metabolism , Obesity/metabolism , Glucose/metabolism , Hypertension/metabolism , Denervation , Insulins/metabolism
17.
Fish Physiol Biochem ; 50(2): 785-796, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38108936

ABSTRACT

To elucidate the underlying mechanism of the energy metabolism in largemouth bass (Micropterus salmoides), cultured fish (initial body weight: 77.57 ± 0.75 g) in the present study were starved for 0 h, 12 h, 24 h, 48 h, 96 h and 192 h, respectively. The proximate composition analysis showed that short-term starvation induced a significant up-regulation in crude protein proportion in hepatic of cultured fish (P < 0.05). However, short-term starvation significantly decreased the hepatosomatic index and the viscerosomatic index of cultured fish (P < 0.05). The exact hepatic glycogen content in the group starved for 92 h presented remarkable decrease (P < 0.05). Meanwhile, compared with the weight change of lipid and protein (mg) in hepatic (y = 0.0007x2 - 0.2827x + 49.402; y = 0.0013x2 - 0.5666x + 165.31), the decreasing trend of weight in glycogen (mg) was more pronounced (y = 0.0032x2 - 1.817x + 326.52), which suggested the preferential utilization of hepatic glycogen as energy substrates under short-term starvation. Gene expression analysis revealed that the starvation down-regulated the expression of insulin-like growth factor 1 and genes of TOR pathway, such as target of rapamycin (tor) and ribosomal protein S6 (s6) (P < 0.05). In addition, the starvation significantly enhanced expression of lipolysis-related genes, including hormone-sensitive lipase (hsl) and carnitine palmitoyl transferase I (cpt1), but down-regulated lipogenesis as indicated by the inhibited expression of fatty acids synthase (fas), acetyl-CoA carboxylase 1 (acc1) and acetyl-CoA carboxylase 2 (acc2) (P < 0.05). Starvation of 24 h up-regulated the expression of glycolysis genes, glucokinase (gk), phosphofructokinase liver type (pfkl) and pyruvate kinase (pk), and then their expression returned to the normal level. Meanwhile, the expression of gluconeogenesis genes, such as glucose-6-phosphatase catalytic subunit (g6pc), fructose-1,6-bisphosphatase-1 (fbp1) and phosphoenolpyruvate carboxy kinase (pepck), was significantly inhibited with the short-term starvation (P < 0.05). In conclusion, short-term starvation induced an overall decline in growth performance, but it could deplete the hepatic glycogen accumulation and mobilize glycogen for energy effectively.


Subject(s)
Bass , Animals , Liver Glycogen/metabolism , Acetyl-CoA Carboxylase/metabolism , Lipogenesis , Glycogen/metabolism , Proteins/metabolism , Liver/metabolism
18.
Commun Biol ; 6(1): 1226, 2023 12 04.
Article in English | MEDLINE | ID: mdl-38049528

ABSTRACT

The maternal liver is challenged by metabolic demands throughout pregnancy. However, hepatocyte dynamics and their physiological significance in pregnancy remain unclear. Here, we show in mice that hepatocyte proliferation is spatiotemporally regulated in each liver lobular zone during pregnancy, with transient proliferation of periportal and pericentral hepatocytes during mid and late gestation, respectively. Using adeno-associated virus (AAV)-8-mediated expression of the cell cycle inhibitor p21 in hepatocytes, we show that inhibition of hepatocyte proliferation during mid, but not late, gestation impairs liver growth. Transcriptionally, genes involved in glucose/glycogen metabolism are downregulated in late pregnancy when midgestational hepatocyte proliferation is attenuated. In addition, hepatic glycogen storage is abolished, with concomitant elevated blood glucose concentrations, glucose intolerance, placental glycogen deposition, and fetal overgrowth. Laser capture microdissection and RNA-seq analysis of each liver lobular zone show zone-specific changes in the transcriptome during pregnancy and identify genes that are periportally expressed at midgestation, including the hyaluronan-mediated motility receptor (Hmmr). Knockdown of Hmmr in hepatocytes by AAV8-shHmmr suppresses periportal hepatocyte proliferation at midgestation and induces impaired hepatic glycogen storage, glucose intolerance, placental glycogen deposition and fetal overgrowth. Our results suggest that periportal hepatocyte proliferation during midgestation is critical for maternal glycogen metabolism and fetal size.


Subject(s)
Diabetes, Gestational , Glucose Intolerance , Humans , Mice , Pregnancy , Female , Animals , Liver Glycogen/metabolism , Placenta/metabolism , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Fetal Macrosomia/metabolism , Glucose/metabolism , Glycogen/metabolism , Hepatocytes/metabolism , Homeostasis , Cell Proliferation
19.
J Biol Chem ; 299(11): 105322, 2023 11.
Article in English | MEDLINE | ID: mdl-37805137

ABSTRACT

The liver is critical in maintaining metabolic homeostasis, regulating both anabolic and catabolic processes. Scaffold protein IQ motif-containing GTPase activating protein 2 (IQGAP2) is highly expressed in the liver and implicated in fatty acid uptake. However, its role in coordinating either fed or fasted responses is not well understood. Here we report that IQGAP2 is widely expressed in the liver that is pronounced in the pericentral region. Although control and IQGAP2 knockout mouse model showed comparable hepatic gene expression in the fasted state, we found significant defects in fed state responses. Glycogen levels were reduced in the periportal region when IQGAP2 was deleted. Consistently, we observed a decrease in phosphorylated glycogen synthase kinase 3α and total glycogen synthase protein in the fed IQGAP2 knockout mice which suggest inadequate glycogen synthesis. Moreover, immunoprecipitation of IQGAP2 revealed its interaction with GSK3 and GYS. Furthermore, our study demonstrated that knocking down IQGAP2 in vitro significantly decreased the phosphorylation of AKT and forkhead box O3 proteins downstream of insulin signaling. These findings suggest that IQGAP2 contributes to liver fed state metabolism by interacting with glycogen synthesis regulators and affecting the phosphorylation of insulin pathway components. Our results suggest that IQGAP2 plays a role in regulating fed state metabolism.


Subject(s)
Insulin , Liver Glycogen , Animals , Mice , Glycogen Synthase Kinase 3/metabolism , Insulin/metabolism , Liver/metabolism , Liver Glycogen/metabolism , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
20.
Peptides ; 168: 171076, 2023 10.
Article in English | MEDLINE | ID: mdl-37572792

ABSTRACT

Neprilysin is a peptidase that cleaves glucoregulatory peptides, including glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK). Some studies suggest that its inhibition in diabetes and/or obesity improves glycemia, and that this is associated with enhanced insulin secretion, glucose tolerance and insulin sensitivity. Whether reduced neprilysin activity also improves hepatic glucose metabolism has not been explored. We sought to determine whether genetic deletion of neprilysin suppresses hepatic glucose production (HGP) in high fat-fed mice. Nep+/+ and Nep-/- mice were fed high fat diet for 16 weeks, and then underwent a pyruvate tolerance test (PTT) to assess hepatic gluconeogenesis. Since glycogen breakdown in liver can also yield glucose, we assessed liver glycogen content in fasted and fed mice. In Nep-/- mice, glucose excursion during the PTT was reduced when compared to Nep+/+ mice. Further, liver glycogen levels were significantly greater in fasted but not fed Nep-/- versus Nep+/+ mice. Since gut-derived factors modulate HGP, we tested whether gut-selective inhibition of neprilysin could recapitulate the suppression of hepatic gluconeogenesis observed with whole-body inhibition, and this was indeed the case. Finally, the gut-derived neprilysin substrates, GLP-1 and CCK, are well-known to suppress HGP. Having previously demonstrated elevated plasma GLP-1 levels in Nep-/- mice, we now measured plasma CCK bioactivity and reveal an increase in Nep-/- versus Nep+/+ mice, suggesting GLP-1 and/or CCK may play a role in reducing HGP under conditions of neprilysin deficiency. In sum, neprilysin modulates hepatic gluconeogenesis and strategies to inhibit its activity may reduce HGP in type 2 diabetes and obesity.


Subject(s)
Diabetes Mellitus, Type 2 , Gluconeogenesis , Mice , Animals , Gluconeogenesis/genetics , Neprilysin , Diabetes Mellitus, Type 2/metabolism , Liver Glycogen/metabolism , Glucose/metabolism , Liver/metabolism , Glucagon-Like Peptide 1/metabolism , Obesity/metabolism , Insulin/metabolism , Blood Glucose/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL