181 results on '"G. Mithieux"'
Search Results
2. Intestinal gluconeogenesis controls the neonatal development of hypothalamic feeding circuits.
- Author
-
Estrada-Meza J, Videlo J, Bron C, Duchampt A, Saint-Béat C, Zergane M, Silva M, Rajas F, Bouret SG, Mithieux G, and Gautier-Stein A
- Subjects
- Animals, Mice, Agouti-Related Protein metabolism, Glucose-6-Phosphatase metabolism, Glucose-6-Phosphatase genetics, Female, Male, Mice, Inbred C57BL, Pro-Opiomelanocortin metabolism, Energy Metabolism, Intestines growth & development, Intestines innervation, Intestines metabolism, Adipose Tissue metabolism, Leptin metabolism, Hypothalamus metabolism, Gluconeogenesis, Animals, Newborn
- Abstract
Objective: Intestinal gluconeogenesis (IGN) regulates adult energy homeostasis in part by controlling the same hypothalamic targets as leptin. In neonates, leptin exhibits a neonatal surge controlling axonal outgrowth between the different hypothalamic nuclei involved in feeding circuits and autonomic innervation of peripheral tissues involved in energy and glucose homeostasis. Interestingly, IGN is induced during this specific time-window. We hypothesized that the neonatal pic of IGN also regulates the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues., Methods: We genetically induced neonatal IGN by overexpressing G6pc1 the catalytic subunit of glucose-6-phosphatase (the mandatory enzyme of IGN) at birth or at twelve days after birth. The neonatal development of hypothalamic feeding circuits was studied by measuring Agouti-related protein (AgRP) and Pro-opiomelanocortin (POMC) fiber density in hypothalamic nuclei of 20-day-old pups. The effect of the neonatal induction of intestinal G6pc1 on sympathetic innervation of the adipose tissues was studied via tyrosine hydroxylase (TH) quantification. The metabolic consequences of the neonatal induction of intestinal G6pc1 were studied in adult mice challenged with a high-fat/high-sucrose (HFHS) diet for 2 months., Results: Induction of intestinal G6pc1 at birth caused a neonatal reorganization of AgRP and POMC fiber density in the paraventricular nucleus of the hypothalamus, increased brown adipose tissue tyrosine hydroxylase levels, and protected against high-fat feeding-induced metabolic disorders. In contrast, inducing intestinal G6pc1 12 days after birth did not impact AgRP/POMC fiber densities, adipose tissue innervation or adult metabolism., Conclusion: These findings reveal that IGN at birth but not later during postnatal life controls the development of hypothalamic feeding circuits and sympathetic innervation of adipose tissues, promoting a better management of metabolism in adulthood., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Loss of atrial natriuretic peptide signaling causes insulin resistance, mitochondrial dysfunction, and low endurance capacity.
- Author
-
Carper D, Lac M, Coue M, Labour A, Märtens A, Banda JAA, Mazeyrie L, Mechta M, Ingerslev LR, Elhadad M, Petit JV, Maslo C, Monbrun L, Del Carmine P, Sainte-Marie Y, Bourlier V, Laurens C, Mithieux G, Joanisse DR, Coudray C, Feillet-Coudray C, Montastier E, Viguerie N, Tavernier G, Waldenberger M, Peters A, Wang-Sattler R, Adamski J, Suhre K, Gieger C, Kastenmüller G, Illig T, Lichtinghagen R, Seissler J, Mounier R, Hiller K, Jordan J, Barrès R, Kuhn M, Pesta D, and Moro C
- Subjects
- Animals, Mice, Humans, Physical Endurance, Male, Mice, Knockout, Oxidative Phosphorylation, Insulin Resistance, Signal Transduction, Atrial Natriuretic Factor metabolism, Receptors, Atrial Natriuretic Factor metabolism, Receptors, Atrial Natriuretic Factor genetics, Muscle, Skeletal metabolism, Diabetes Mellitus, Type 2 metabolism, Diabetes Mellitus, Type 2 pathology, Mitochondria metabolism
- Abstract
Type 2 diabetes (T2D) and obesity are strongly associated with low natriuretic peptide (NP) plasma levels and a down-regulation of NP guanylyl cyclase receptor-A (GCA) in skeletal muscle and adipose tissue. However, no study has so far provided evidence for a causal link between atrial NP (ANP)/GCA deficiency and T2D pathogenesis. Here, we show that both systemic and skeletal muscle ANP/GCA deficiencies in mice promote metabolic disturbances and prediabetes. Skeletal muscle insulin resistance is further associated with altered mitochondrial function and impaired endurance running capacity. ANP/GCA-deficient mice exhibit increased proton leak and reduced content of mitochondrial oxidative phosphorylation proteins. We further show that GCA is related to several metabolic traits in T2D and positively correlates with markers of oxidative capacity in human skeletal muscle. Together, these results indicate that ANP/GCA signaling controls muscle mitochondrial integrity and oxidative capacity in vivo and plays a causal role in the development of prediabetes.
- Published
- 2024
- Full Text
- View/download PDF
4. Intestinal gluconeogenesis: A translator of nutritional information needed for glycemic and emotional balance.
- Author
-
Gautier-Stein A, Vily-Petit J, Rajas F, and Mithieux G
- Subjects
- Humans, Animals, Emotions physiology, Intestines physiology, Blood Glucose metabolism, Intestinal Mucosa metabolism, Glucose metabolism, Dietary Fiber metabolism, Gluconeogenesis
- Abstract
At the interface between the outside world and the self, the intestine is the first organ receiving nutritional information. One intestinal function, gluconeogenesis, is activated by various nutrients, particularly diets enriched in fiber or protein, and thus results in glucose production in the portal vein in the post-absorptive period. The detection of portal glucose induces a nervous signal controlling the activity of the central nuclei involved in the regulation of metabolism and emotional behavior. Induction of intestinal gluconeogenesis is necessary for the beneficial effects of fiber or protein-enriched diets on metabolism and emotional behavior. Through its ability to translate nutritional information from the diet to the brain's regulatory centers, intestinal gluconeogenesis plays an essential role in maintaining physiological balance., (Copyright © 2024 Elsevier B.V. and Société Française de Biochimie et Biologie Moléculaire (SFBBM). All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. Long-term trajectories of weight loss and health outcomes: protocol of the SCOOP-RNPC nationwide observational study.
- Author
-
Fabre O, Bailly S, Mithieux G, Legrand R, Costentin C, Astrup A, and Pépin JL
- Subjects
- Humans, Prospective Studies, France, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 therapy, Hypertension therapy, Weight Reduction Programs methods, Sleep Apnea, Obstructive therapy, Research Design, Female, Observational Studies as Topic, Male, Multicenter Studies as Topic, Cardiovascular Diseases prevention & control, Weight Loss, Obesity complications, Obesity therapy
- Abstract
Introduction: Behavioural weight loss programmes are generally accepted as being beneficial in reducing cardiometabolic risk and improving patient-reported outcomes. However, prospective data from large real-world cohorts are scarce concerning the mid-term and long-term impact of such interventions. The objective of this large prospective cohort study (n>10 000 participants) is to demonstrate the effectiveness of the standardised Nutritional and Psycho-Behavioural Rehabilitation programme (RNPC Programme) in reducing the percentage of subjects requiring insulin and/or other diabetes drug therapy, antihypertensive drugs, lipid-lowering therapies and continuous positive airway pressure therapy for obstructive sleep apnoea after the end of the intervention. The rate of remission of hypertension, type 2 diabetes and sleep apnoea will also be prospectively assessed., Methods: This is a prospective multicentre observational study carried out in 92 RNPC centres in France. Participants will follow the standardised RNPC Programme. The prospective dataset will include clinical, anthropometric and biochemical data, comorbidities, medications, body composition, patient-reported outcome questionnaire responses, sleep study data with objective measurements of sleep apnoea severity and surrogate markers of cardiovascular risk (ie, blood pressure and arterial stiffness). About 10 000 overweight or obese participants will be included over 2 years with a follow-up duration of up to 5 years., Ethics and Dissemination: Ethical approval for this study has been granted by the Ethics Committee (Comité de protection des personnes Sud-Est I) of Saint-Etienne University Hospital, France (SI number: 23.00174.000237). Results will be submitted for publication in peer-review journals, presented at conferences and inform the design of a future randomised controlled trial in the specific population identified as good responders to the RNPC Programme., Trial Registration Number: NCT05857319., Competing Interests: Competing interests: OF is employed by the 'Groupe Éthique et Santé', RL is the Founding President of 'Groupe Éthique et Santé', AA is the president of the Scientific Committee of the Groupe Éthique et Santé. SB, GM and J-LP have no conflicts of interest to disclose in relation to this study., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
6. Absence of the Peptide Transporter 1 Induces a Prediabetic and Depressive-Like Phenotype in Mice.
- Author
-
Vily-Petit J, Taki A, Sinet F, Soty M, Guiard B, Zemdegs J, Malleret G, Stefanutti A, Mithieux G, and Gautier-Stein A
- Abstract
Introduction: Protein-enriched diets improve glycemic control in diabetes or emotional behavior in depressive patients. In mice, these benefits depend on intestinal gluconeogenesis activation by di-/tripeptides. Intestinal di-/tripeptides absorption is carried out by the peptide transporter 1, PEPT1. The lack of PEPT1 might thus alter glucose and emotional balance., Methods: To determine the effects of PEPT1 deficiency under standard dietary conditions or during a dietary challenge known to promote both metabolic and cognitive dysfunction, insulin sensitivity, anxiety, and depressive-like traits, hippocampal serotonin (5-HT) and insulin signaling pathway were measured in wild-type (WT) and Pept1-/- mice fed either a chow or a high-fat high-sucrose (HF-HS) diet., Results: Pept1-/- mice exhibited slight defects in insulin sensitivity and emotional behavior, which were aggravated by an HF-HS diet. Pept1-/- mice fed a chow diet had lower hippocampal 5-HT levels and exhibited cerebral insulin resistance under HF-HS diet. These defects were independent of intestinal gluconeogenesis but might be linked to increased plasma amino acids levels., Conclusion: Pept1-/- mice develop prediabetic and depressive-like traits and could thus be used to develop strategies to prevent or cure both diseases., (© 2024 S. Karger AG, Basel.)
- Published
- 2024
- Full Text
- View/download PDF
7. Antiobesity effects of intestinal gluconeogenesis are mediated by the brown adipose tissue sympathetic nervous system.
- Author
-
Vily-Petit J, Soty-Roca M, Silva M, Micoud M, Evrard F, Bron C, Raffin M, Beiroa D, Nogueiras R, Roussel D, Gautier-Stein A, Rajas F, Cota D, and Mithieux G
- Subjects
- Humans, Animals, Mice, Obesity complications, Adipose Tissue, White metabolism, Glucose metabolism, Sympathetic Nervous System metabolism, Thermogenesis, Energy Metabolism, Adipose Tissue, Brown metabolism, Gluconeogenesis genetics
- Abstract
Objective: Intestinal gluconeogenesis (IGN), via the initiation of a gut-brain nervous circuit, accounts for the metabolic benefits linked to dietary proteins or fermentable fiber in rodents and has been positively correlated with the rapid amelioration of body weight after gastric bypass surgery in humans with obesity. In particular, the activation of IGN moderates the development of hepatic steatosis accompanying obesity. In this study, we investigated the specific effects of IGN on adipose tissue metabolism, independent of its induction by nutritional manipulation., Methods: We used two transgenic mouse models of suppression or overexpression of G6pc1, the catalytic subunit of glucose-6 phosphatase, which is the key enzyme of endogenous glucose production specifically in the intestine., Results: Under a hypercaloric diet, mice overexpressing IGN showed lower adiposity and higher thermogenic capacities than wild-type mice, featuring marked browning of white adipose tissue (WAT) and prevention of the whitening of brown adipose tissue (BAT). Sympathetic denervation restricted to BAT caused the loss of the antiobesity effects associated with IGN. Conversely, IGN-deficient mice exhibited an increase in adiposity under a standard diet, which was associated with decreased expression of markers of thermogenesis in both BAT and WAT., Conclusions: IGN is sufficient to activate the sympathetic nervous system and prevent the expansion and the metabolic alterations of BAT and WAT metabolism under a high-calorie diet, thereby preventing the development of obesity. These data increase knowledge of the mechanisms of weight reduction in gastric bypass surgery and pave the way for new approaches to prevent or cure obesity., (© 2024 The Obesity Society.)
- Published
- 2024
- Full Text
- View/download PDF
8. Fish oil minimises feed intake and improves insulin sensitivity in Zucker fa/fa rats.
- Author
-
Corporeau C, Le Foll C, Cruciani-Guglielmacci C, Le Stunff H, Mithieux G, Magnan C, and Delarue J
- Subjects
- Humans, Male, Rats, Animals, Fish Oils pharmacology, Rats, Zucker, Blood Glucose metabolism, Insulin metabolism, Obesity metabolism, Glucose pharmacology, Eating, Weight Gain, Fatty Acids, Unsaturated pharmacology, Corn Oil pharmacology, Insulin Resistance physiology, Fatty Acids, Omega-3 pharmacology
- Abstract
Long-chain n -3 PUFA (LC n -3 PUFA) prevent, in rodents, insulin resistance (IR) induced by a high-fat and/or fructose diet but not IR induced by glucocorticoids. In humans, contrasting effects have also been reported. We investigated their effects on insulin sensitivity, feed intake (FI) and body weight gain in genetically insulin resistant male obese (fa/fa) Zucker (ZO) rats during the development of obesity. ZO rats were fed a diet supplemented with 7 % fish oil (FO) + 1 % corn oil (CO) (wt/wt) (ZO
FO ), while the control group was fed a diet containing 8 % fat from CO (wt/wt) (ZOCO ). Male lean Zucker (ZL) rats fed either FO (ZLFO ) or CO (ZLCO ) diet were used as controls. FO was a marine-derived TAG oil containing EPA 90 mg/g + DHA 430 mg/g. During an oral glucose tolerance test, glucose tolerance remained unaltered by FO while insulin response was reduced in ZOFO only. Liver insulin sensitivity (euglycaemic-hyperinsulinaemic clamp + 2 deoxyglucose) was improved in ZOFO rats, linked to changes in phosphoenolpyruvate carboxykinase expression, activity and glucose-6-phosphatase activity. FI in response to intra-carotid insulin/glucose infusion was decreased similarly in ZOFO and ZOCO . Hypothalamic ceramides levels were lower in ZOFO than in ZOCO . Our study demonstrates that LC n -3 PUFA can minimise weight gain, possibly by alleviating hypothalamic lipotoxicity, and liver IR in genetically obese Zucker rats.- Published
- 2024
- Full Text
- View/download PDF
9. Improvement of energy metabolism associated with NUTRIOSE® soluble fiber, a dietary ingredient exhibiting prebiotic properties, requires intestinal gluconeogenesis.
- Author
-
Vily-Petit J, Soty M, Silva M, Micoud M, Bron C, Guérin-Deremaux L, and Mithieux G
- Subjects
- Humans, Mice, Animals, Gluconeogenesis, Diet, Energy Metabolism, Dietary Fiber metabolism, Obesity prevention & control, Obesity metabolism, Prebiotics, Non-alcoholic Fatty Liver Disease prevention & control
- Abstract
While the prevalence of obesity progresses worldwide, the consumption of sugars and dietary fiber increases and decreases, respectively. In this context, NUTRIOSE® soluble fiber is a plant-based food ingredient with beneficial effects in Humans. Here, we studied in mice the mechanisms involved, particularly the involvement of intestinal gluconeogenesis (IGN), the essential function in the beneficial effects of dietary fibers. To determine whether NUTRIOSE® exerts its beneficial effects via the activation of IGN, we studied the effects of dietary NUTRIOSE® on the development of obesity, diabetes and non-alcoholic fatty liver disease (NAFLD), which IGN is able to prevent. To assert the role of IGN in the observed effects, we studied wild-type (WT) and IGN-deficient mice. In line with our hypothesis, NUTRIOSE® exerts metabolic benefits in WT mice, but not in IGN-deficient mice. Indeed, WT mice are protected from body weight gain and NAFLD induced by a high calorie diet. In addition, our data suggests that NUTRIOSE® may improve energy balance by activating a browning process in subcutaneous white adipose tissue. While the gut microbiota composition changes with NUTRIOSE®, this is not sufficient in itself to account for the benefits observed. On the contrary, IGN is obligatory in the NUTRIOSE® benefits, since no benefit take place in absence of IGN. In conclusion, IGN plays a crucial and essential role in the set-up of the beneficial effects of NUTRIOSE®, highlighting the interest of the supplementation of food with healthy ingredients in the context of the current obesity epidemic., Competing Interests: Declaration of Competing Interest L.G-D is an employee of the Roquette company. The other authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Normalization of hepatic ChREBP activity does not protect against liver disease progression in a mouse model for Glycogen Storage Disease type Ia.
- Author
-
Rutten MGS, Lei Y, Hoogerland JH, Bloks VW, Yang H, Bos T, Krishnamurthy KA, Bleeker A, Koster MH, Thomas RE, Wolters JC, van den Bos H, Mithieux G, Rajas F, Mardinoglu A, Spierings DCJ, de Bruin A, van de Sluis B, and Oosterveer MH
- Abstract
Background: Glycogen storage disease type 1a (GSD Ia) is an inborn error of metabolism caused by a defect in glucose-6-phosphatase (G6PC1) activity, which induces severe hepatomegaly and increases the risk for liver cancer. Hepatic GSD Ia is characterized by constitutive activation of Carbohydrate Response Element Binding Protein (ChREBP), a glucose-sensitive transcription factor. Previously, we showed that ChREBP activation limits non-alcoholic fatty liver disease (NAFLD) in hepatic GSD Ia. As ChREBP has been proposed as a pro-oncogenic molecular switch that supports tumour progression, we hypothesized that ChREBP normalization protects against liver disease progression in hepatic GSD Ia., Methods: Hepatocyte-specific G6pc knockout (L-G6pc
-/- ) mice were treated with AAV-shChREBP to normalize hepatic ChREBP activity., Results: Hepatic ChREBP normalization in GSD Ia mice induced dysplastic liver growth, massively increased hepatocyte size, and was associated with increased hepatic inflammation. Furthermore, nuclear levels of the oncoprotein Yes Associated Protein (YAP) were increased and its transcriptional targets were induced in ChREBP-normalized GSD Ia mice. Hepatic ChREBP normalization furthermore induced DNA damage and mitotic activity in GSD Ia mice, while gene signatures of chromosomal instability, the cytosolic DNA-sensing cGAS-STING pathway, senescence, and hepatocyte dedifferentiation emerged., Conclusions: In conclusion, our findings indicate that ChREBP activity limits hepatomegaly while decelerating liver disease progression and protecting against chromosomal instability in hepatic GSD Ia. These results disqualify ChREBP as a therapeutic target for treatment of liver disease in GSD Ia. In addition, they underline the importance of establishing the context-specific roles of hepatic ChREBP to define its therapeutic potential to prevent or treat advanced liver disease., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
11. A caveolin-1 dependent glucose-6-phosphatase trafficking contributes to hepatic glucose production.
- Author
-
Gautier-Stein A, Chilloux J, Soty M, Thorens B, Place C, Zitoun C, Duchampt A, Da Costa L, Rajas F, Lamaze C, and Mithieux G
- Subjects
- Animals, Mice, Caveolin 1 metabolism, Cholesterol metabolism, Liver metabolism, Glucose metabolism, Glucose-6-Phosphatase metabolism
- Abstract
Objective: Deregulation of hepatic glucose production is a key driver in the pathogenesis of diabetes, but its short-term regulation is incompletely deciphered. According to textbooks, glucose is produced in the endoplasmic reticulum by glucose-6-phosphatase (G6Pase) and then exported in the blood by the glucose transporter GLUT2. However, in the absence of GLUT2, glucose can be produced by a cholesterol-dependent vesicular pathway, which remains to be deciphered. Interestingly, a similar mechanism relying on vesicle trafficking controls short-term G6Pase activity. We thus investigated whether Caveolin-1 (Cav1), a master regulator of cholesterol trafficking, might be the mechanistic link between glucose production by G6Pase in the ER and glucose export through a vesicular pathway., Methods: Glucose production from fasted mice lacking Cav1, GLUT2 or both proteins was measured in vitro in primary culture of hepatocytes and in vivo by pyruvate tolerance tests. The cellular localization of Cav1 and the catalytic unit of glucose-6-phosphatase (G6PC1) were studied by western blotting from purified membranes, immunofluorescence on primary hepatocytes and fixed liver sections and by in vivo imaging of chimeric constructs overexpressed in cell lines. G6PC1 trafficking to the plasma membrane was inhibited by a broad inhibitor of vesicular pathways or by an anchoring system retaining G6PC1 specifically to the ER membrane., Results: Hepatocyte glucose production is reduced at the step catalyzed by G6Pase in the absence of Cav1. In the absence of both GLUT2 and Cav1, gluconeogenesis is nearly abolished, indicating that these pathways can be considered as the two major pathways of de novo glucose production. Mechanistically, Cav1 colocalizes but does not interact with G6PC1 and controls its localization in the Golgi complex and at the plasma membrane. The localization of G6PC1 at the plasma membrane is correlated to glucose production. Accordingly, retaining G6PC1 in the ER reduces glucose production by hepatic cells., Conclusions: Our data evidence a pathway of glucose production that relies on Cav1-dependent trafficking of G6PC1 to the plasma membrane. This reveals a new cellular regulation of G6Pase activity that contributes to hepatic glucose production and glucose homeostasis., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Amandine Gautier-Stein reports financial support was provided by French National Research Agency., (Copyright © 2023 The Author(s). Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Transcription factor p63, a member of the p53 family of tumour suppressors, regulates hepatic glucose metabolism.
- Author
-
Mithieux G
- Subjects
- Humans, Glucose metabolism, Tumor Suppressor Protein p53 metabolism, Neoplasms, Transcription Factors metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2023
- Full Text
- View/download PDF
13. Intestinal gluconeogenesis: metabolic benefits make sense in the light of evolution.
- Author
-
Gautier-Stein A and Mithieux G
- Subjects
- Animals, Humans, Glucose metabolism, Homeostasis physiology, Liver metabolism, Gluconeogenesis physiology, Intestines
- Abstract
The intestine, like the liver and kidney, in various vertebrates and humans is able to carry out gluconeogenesis and release glucose into the blood. In the fed post-absorptive state, intestinal glucose is sensed by the gastrointestinal nervous system. The latter initiates a signal to the brain regions controlling energy homeostasis and stress-related behaviour. Intestinal gluconeogenesis (IGN) is activated by several complementary mechanisms, in particular nutritional situations (for example, when food is enriched in protein or fermentable fibre and after gastric bypass surgery in obesity). In these situations, IGN has several metabolic and behavioural benefits. As IGN is activated by nutrients capable of fuelling systemic gluconeogenesis, IGN could be a signal to the brain that food previously ingested is suitable for maintaining plasma glucose for a while. This process might account for the benefits observed. Finally, in this Perspective, we discuss how the benefits of IGN in fasting and fed states could explain why IGN emerged and was maintained in vertebrates by natural selection., (© 2022. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
14. Editorial: Human microbiota: A key player in the etiology and pathophysiology of cardiovascular and metabolic diseases.
- Author
-
Velmurugan G, Ramprasath T, and Mithieux G
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2022
- Full Text
- View/download PDF
15. Portal Glucose Infusion, Afferent Nerve Fibers, and Glucose and Insulin Tolerance of Insulin-Resistant Rats.
- Author
-
Joly-Amado A, Soty M, Philippe E, Lacombe A, Castel J, Pillot B, Vily-Petit J, Zitoun C, Mithieux G, and Magnan C
- Subjects
- Animals, Blood Glucose metabolism, Capsaicin metabolism, Capsaicin pharmacology, Emulsions metabolism, Glucose metabolism, Insulin, Regular, Human pharmacology, Liver metabolism, Male, Nerve Fibers metabolism, Portal Vein metabolism, Rats, Rats, Wistar, Triglycerides metabolism, Insulin metabolism, Insulin Resistance
- Abstract
Background: The role of hepatoportal glucose sensors is poorly understood in the context of insulin resistance., Objectives: We assessed the effects of glucose infusion in the portal vein on insulin tolerance in 2 rat models of insulin resistance, and the role of capsaicin sensitive nerves in this signal., Methods: Male Wistar rats, 8 weeks old, weighing 250-275 g, were used. Insulin and glucose tolerance were assessed following a 4-hour infusion of either glucose or saline through catheterization in the portal vein in 3 paradigms. In experiment 1, for diet-induced insulin resistance, rats were fed either a control diet (energy content: proteins = 22.5%, carbohydrates = 64.1%, and lipids = 13.4%) or a high-fat diet (energy content: proteins = 15.3%, carbohydrates = 40.3%, and lipids =44.4%) for 4 months. In experiment 2, for centrally induced peripheral insulin resistance, catheters were inserted in the carotid artery to deliver either an emulsion of triglycerides [intralipid (IL)] or saline towards the brain for 24 hours. In experiment 3, for testing the role of capsaicin-sensitive nerves, experiment 2 was repeated following a periportal treatment with capsaicin or vehicle., Results: In experiment 1, when compared to rats fed the control diet, rats fed the high-fat diet exhibited decreased insulin and glucose tolerance (P ≤ 0.05) that was restored with a glucose infusion in the portal vein (P ≤ 0.05). In experiment 2, infusion of a triglyceride emulsion towards the brain (IL rats) decreased insulin and glucose tolerance and increased hepatic endogenous production when compared to saline-infused rats (P ≤ 0.05). Glucose infusion in the portal vein in IL rats restored insulin and glucose tolerance, as well as hepatic glucose production, to controls levels (P ≤ 0.05). In experiment 3, portal infusion of glucose did not increase insulin tolerance in IL rats that received a periportal pretreatment with capsaicin., Conclusions: Stimulation of hepatoportal glucose sensors increases insulin tolerance in rat models of insulin resistance and requires the presence of capsaicin-sensitive nerves., (© The Author(s) 2022. Published by Oxford University Press on behalf of the American Society for Nutrition.)
- Published
- 2022
- Full Text
- View/download PDF
16. A hypometabolic defense strategy against malaria.
- Author
-
Ramos S, Ademolue TW, Jentho E, Wu Q, Guerra J, Martins R, Pires G, Weis S, Carlos AR, Mahú I, Seixas E, Duarte D, Rajas F, Cardoso S, Sousa AGG, Lilue J, Paixão T, Mithieux G, Nogueira F, and Soares MP
- Subjects
- Glucose, Humans, Plasmodium falciparum, Hypoglycemia, Malaria, Malaria, Falciparum parasitology
- Abstract
Hypoglycemia is a clinical hallmark of severe malaria, the often-lethal outcome of Plasmodium falciparum infection. Here, we report that malaria-associated hypoglycemia emerges from a non-canonical resistance mechanism, whereby the infected host reduces glycemia to starve Plasmodium. This hypometabolic response is elicited by labile heme, a byproduct of hemolysis that induces illness-induced anorexia and represses hepatic glucose production. While transient repression of hepatic glucose production prevents unfettered immune-mediated inflammation, organ damage, and anemia, when sustained over time it leads to hypoglycemia, compromising host energy expenditure and adaptive thermoregulation. The latter arrests the development of asexual stages of Plasmodium via a mechanism associated with parasite mitochondrial dysfunction. In response, Plasmodium activates a transcriptional program associated with the reduction of virulence and sexual differentiation toward the generation of transmissible gametocytes. In conclusion, malaria-associated hypoglycemia represents a trade-off of a hypometabolic-based defense strategy that balances parasite virulence versus transmission., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
17. The gut microbiota: stable bioreactor of variable composition?
- Author
-
Mithieux G
- Subjects
- Bioreactors, Fatty Acids, Volatile metabolism, Humans, Gastrointestinal Microbiome
- Abstract
The gut microbiota plays a crucial role in host health, providing energy and vitamins from food undigested by the gut enzymes of the host. Bacterial metabolites, such as short-chain fatty acids (SCFAs), are essentially metabolized by the gut mucosa. The importance to metabolic health of gut microbiota composition versus function is discussed., Competing Interests: Declaration of interests The author has no conflict of interest to disclose relating to this article., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. Increased atherosclerosis in a mouse model of glycogen storage disease type 1a.
- Author
-
La Rose AM, Groenen AG, Halmos B, Bazioti V, Rutten MGS, Krishnamurthy KA, Koster MH, Kloosterhuis NJ, Smit M, Havinga R, Mithieux G, Rajas F, Kuipers F, Oosterveer MH, and Westerterp M
- Abstract
Glycogen storage disease type 1a (GSD Ia) is an inborn error of carbohydrate metabolism. Despite severe hyperlipidemia, GSD Ia patients show limited atherogenesis compared to age-and-gender matched controls. Employing a GSD Ia mouse model that resembles the severe hyperlipidemia in patients, we here found increased atherogenesis in GSD Ia. These data provide a rationale for investigating atherogenesis in GSD Ia in a larger patient cohort., Competing Interests: None., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
19. Cellular and metabolic effects of renin-angiotensin system blockade on glycogen storage disease type I nephropathy.
- Author
-
Monteillet L, Labrune P, Hochuli M, Do Cao J, Tortereau A, Miliano AC, Ardon-Zitoun C, Duchampt A, Silva M, Verzieux V, Mithieux G, and Rajas F
- Subjects
- Animals, Female, Glucose metabolism, Humans, Lipids, Male, Mice, Renin-Angiotensin System genetics, Retrospective Studies, Glycogen Storage Disease Type I complications, Glycogen Storage Disease Type I drug therapy, Glycogen Storage Disease Type I genetics, Renal Insufficiency, Chronic drug therapy, Renal Insufficiency, Chronic genetics
- Abstract
Glycogen Storage Disease Type I (GSDI) is an inherited disease caused by glucose-6 phosphatase (G6Pase) deficiency, leading to a loss of endogenous glucose production and severe hypoglycemia. Moreover, most GSDI patients develop a chronic kidney disease (CKD) due to lipid accumulation in the kidney. Similar to diabetic CKD, activation of renin-angiotensin system (RAS) promotes renal fibrosis in GSDI. Here, we investigated the physiological and molecular effects of RAS blockers in GSDI patients and mice. A retrospective analysis of renal function was performed in 21 GSDI patients treated with RAS blockers. Cellular and metabolic impacts of RAS blockade were analyzed in K.G6pc-/- mice characterized by G6pc1 deletion in kidneys. GSDI patients started RAS blocker treatment at a median age of 21 years and long-term treatment reduced the progression of CKD in about 50% of patients. However, CKD progressed to kidney failure in 20% of treated patients, requiring renal transplantation. In K.G6pc-/- mice, CKD was associated with an impairment of autophagy and ER stress. RAS blockade resulted in a rescue of autophagy and decreased ER stress, concomitantly with decreased fibrosis and improved renal function, but without impact on glycogen and lipid contents. In conclusion, these data confirm the partial beneficial effect of RAS blockers in the prevention of CKD in GSDI. Mechanistically, we show that these effects are linked to a reduction of cell stress, without affecting metabolism., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
20. Intestinal gluconeogenesis shapes gut microbiota, fecal and urine metabolome in mice with gastric bypass surgery.
- Author
-
Vily-Petit J, Barataud A, Zitoun C, Gautier-Stein A, Serino M, and Mithieux G
- Subjects
- Actinobacteria classification, Actinobacteria genetics, Actinobacteria isolation & purification, Animals, DNA, Bacterial genetics, Enterobacteriaceae classification, Enterobacteriaceae genetics, Enterobacteriaceae isolation & purification, Enterococcaceae classification, Enterococcaceae genetics, Enterococcaceae isolation & purification, Fatty Acids, Volatile metabolism, Firmicutes classification, Firmicutes genetics, Firmicutes isolation & purification, Intestines microbiology, Male, Mice, Mice, Inbred C57BL, Phylogeny, Proteobacteria classification, Proteobacteria genetics, Proteobacteria isolation & purification, Stomach microbiology, Stomach surgery, Gastric Bypass methods, Gastrointestinal Microbiome physiology, Gluconeogenesis physiology, Intestines metabolism, Metabolome, Stomach metabolism
- Abstract
Intestinal gluconeogenesis (IGN), gastric bypass (GBP) and gut microbiota positively regulate glucose homeostasis and diet-induced dysmetabolism. GBP modulates gut microbiota, whether IGN could shape it has not been investigated. We studied gut microbiota and microbiome in wild type and IGN-deficient mice, undergoing GBP or not, and fed on either a normal chow (NC) or a high-fat/high-sucrose (HFHS) diet. We also studied fecal and urine metabolome in NC-fed mice. IGN and GBP had a different effect on the gut microbiota of mice fed with NC and HFHS diet. IGN inactivation increased abundance of Deltaproteobacteria on NC and of Proteobacteria such as Helicobacter on HFHS diet. GBP increased abundance of Firmicutes and Proteobacteria on NC-fed WT mice and of Firmicutes, Bacteroidetes and Proteobacteria on HFHS-fed WT mice. The combined effect of IGN inactivation and GBP increased abundance of Actinobacteria on NC and the abundance of Enterococcaceae and Enterobacteriaceae on HFHS diet. A reduction was observed in the amounf of short-chain fatty acids in fecal (by GBP) and in both fecal and urine (by IGN inactivation) metabolome. IGN and GBP, separately or combined, shape gut microbiota and microbiome on NC- and HFHS-fed mice, and modify fecal and urine metabolome., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. [Intestinal gluconeogenesis: an insulin-mimetic function].
- Author
-
Mithieux G
- Subjects
- Animals, Dietary Fiber metabolism, Glucose metabolism, Homeostasis, Humans, Insulin metabolism, Intestinal Mucosa metabolism, Mice, Obesity metabolism, Gluconeogenesis physiology, Insulin Resistance
- Abstract
Intestinal gluconeogenesis (IGN) is a regulatory function of energy homeostasis. IGN-produced glucose is sensed by the gastrointestinal nervous system and sends a signal to regions of the brain regulating food intake and glucose control. IGN is activated by dietary protein and dietary fibre, and by gastric bypass surgery of obesity. Glutamine, propionate and succinate are the main substrates used for glucose production by IGN. Activation of IGN accounts for the well-known satiety effect of protein-enriched diets and the anti-obesity and anti-diabetes effects associated with fibre feeding and gastric bypass surgery. Genetic activation of IGN in mice shows the same beneficial effects, independently of any nutritional manipulation, including a marked prevention of hepatic steatosis under hypercaloric feeding. The activation of IGN could thus be the basis for new approaches to prevent or correct metabolic diseases in humans., (© Société de Biologie, 2022.)
- Published
- 2022
- Full Text
- View/download PDF
22. Hepatocyte-specific glucose-6-phosphatase deficiency disturbs platelet aggregation and decreases blood monocytes upon fasting-induced hypoglycemia.
- Author
-
La Rose AM, Bazioti V, Hoogerland JA, Svendsen AF, Groenen AG, van Faassen M, Rutten MGS, Kloosterhuis NJ, Dethmers-Ausema B, Nijland JH, Mithieux G, Rajas F, Kuipers F, Lukens MV, Soehnlein O, Oosterveer MH, and Westerterp M
- Subjects
- Animals, Disease Models, Animal, Female, Glycogen Storage Disease Type I pathology, Hepatocytes pathology, Hypoglycemia pathology, Ice, Male, Mice, Knockout, Mice, Transgenic, Monocytes pathology, Platelet Aggregation, Mice, Fasting, Glycogen Storage Disease Type I metabolism, Hepatocytes metabolism, Hypoglycemia metabolism, Monocytes metabolism
- Abstract
Objective: Glycogen storage disease type 1a (GSD Ia) is a rare inherited metabolic disorder caused by mutations in the glucose-6-phosphatase (G6PC1) gene. When untreated, GSD Ia leads to severe fasting-induced hypoglycemia. Although current intensive dietary management aims to prevent hypoglycemia, patients still experience hypoglycemic events. Poor glycemic control in GSD Ia is associated with hypertriglyceridemia, hepatocellular adenoma and carcinoma, and also with an increased bleeding tendency of unknown origin., Methods: To evaluate the effect of glycemic control on leukocyte levels and coagulation in GSD Ia, we employed hepatocyte-specific G6pc1 deficient (L-G6pc
-/- ) mice under fed or fasted conditions, to match good or poor glycemic control in GSD Ia, respectively., Results: We found that fasting-induced hypoglycemia in L-G6pc-/- mice decreased blood leukocytes, specifically proinflammatory Ly6Chi monocytes, compared to controls. Refeeding reversed this decrease. The decrease in Ly6Chi monocytes was accompanied by an increase in plasma corticosterone levels and was prevented by the glucocorticoid receptor antagonist mifepristone. Further, fasting-induced hypoglycemia in L-G6pc-/- mice prolonged bleeding time in the tail vein bleeding assay, with reversal by refeeding. This could not be explained by changes in coagulation factors V, VII, or VIII, or von Willebrand factor. While the prothrombin and activated partial thromboplastin time as well as total platelet counts were not affected by fasting-induced hypoglycemia in L-G6pc-/- mice, ADP-induced platelet aggregation was disturbed., Conclusions: These studies reveal a relationship between fasting-induced hypoglycemia, decreased blood monocytes, and disturbed platelet aggregation in L-G6pc-/- mice. While disturbed platelet aggregation likely accounts for the bleeding phenotype in GSD Ia, elevated plasma corticosterone decreases the levels of proinflammatory monocytes. These studies highlight the necessity of maintaining good glycemic control in GSD Ia., (Copyright © 2021 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
23. Pain sensing neurons promote tissue regeneration in adult mice.
- Author
-
Rabiller L, Labit E, Guissard C, Gilardi S, Guiard BP, Moulédous L, Silva M, Mithieux G, Pénicaud L, Lorsignol A, Casteilla L, and Dromard C
- Abstract
Tissue repair after injury in adult mammals, usually results in scarring and loss of function in contrast to lower vertebrates such as the newt and zebrafish that regenerate. Understanding the regulatory processes that guide the outcome of tissue repair is therefore a concerning challenge for regenerative medicine. In multiple regenerative animal species, the nerve dependence of regeneration is well established, but the nature of the innervation required for tissue regeneration remains largely undefined. Using our model of induced adipose tissue regeneration in adult mice, we demonstrate here that nociceptive nerves promote regeneration and their removal impairs tissue regeneration. We also show that blocking the receptor for the nociceptive neuropeptide calcitonin gene-related peptide (CGRP) inhibits regeneration, whereas CGRP administration induces regeneration. These findings reveal that peptidergic nociceptive neurons are required for adult mice tissue regeneration., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
24. Driving regeneration, instead of healing, in adult mammals: the decisive role of resident macrophages through efferocytosis.
- Author
-
Rabiller L, Robert V, Arlat A, Labit E, Ousset M, Salon M, Coste A, Da Costa-Fernandes L, Monsarrat P, Ségui B, André M, Guissard C, Renoud ML, Silva M, Mithieux G, Raymond-Letron I, Pénicaud L, Lorsignol A, Casteilla L, Dromard Berthézène C, and Cousin B
- Abstract
Tissue repair after lesion usually leads to scar healing and thus loss of function in adult mammals. In contrast, other adult vertebrates such as amphibians have the ability to regenerate and restore tissue homeostasis after lesion. Understanding the control of the repair outcome is thus a concerning challenge for regenerative medicine. We recently developed a model of induced tissue regeneration in adult mice allowing the comparison of the early steps of regenerative and scar healing processes. By using studies of gain and loss of function, specific cell depletion approaches, and hematopoietic chimeras we demonstrate here that tissue regeneration in adult mammals depends on an early and transient peak of granulocyte producing reactive oxygen species and an efficient efferocytosis specifically by tissue-resident macrophages. These findings highlight key and early cellular pathways able to drive tissue repair towards regeneration in adult mammals., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
25. Hypothalamic bile acid-TGR5 signaling protects from obesity.
- Author
-
Castellanos-Jankiewicz A, Guzmán-Quevedo O, Fénelon VS, Zizzari P, Quarta C, Bellocchio L, Tailleux A, Charton J, Fernandois D, Henricsson M, Piveteau C, Simon V, Allard C, Quemener S, Guinot V, Hennuyer N, Perino A, Duveau A, Maitre M, Leste-Lasserre T, Clark S, Dupuy N, Cannich A, Gonzales D, Deprez B, Mithieux G, Dombrowicz D, Bäckhed F, Prevot V, Marsicano G, Staels B, Schoonjans K, and Cota D
- Subjects
- Animals, Body Weight genetics, Energy Metabolism genetics, HEK293 Cells, Humans, Hypothalamus metabolism, Mice, Mice, Inbred C57BL, Mice, Obese, Mice, Transgenic, Obesity genetics, Obesity prevention & control, Receptors, G-Protein-Coupled genetics, Signal Transduction physiology, Bile Acids and Salts metabolism, Obesity metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
Bile acids (BAs) improve metabolism and exert anti-obesity effects through the activation of the Takeda G protein-coupled receptor 5 (TGR5) in peripheral tissues. TGR5 is also found in the brain hypothalamus, but whether hypothalamic BA signaling is implicated in body weight control and obesity pathophysiology remains unknown. Here we show that hypothalamic BA content is reduced in diet-induced obese mice. Central administration of BAs or a specific TGR5 agonist in these animals decreases body weight and fat mass by activating the sympathetic nervous system, thereby promoting negative energy balance. Conversely, genetic downregulation of hypothalamic TGR5 expression in the mediobasal hypothalamus favors the development of obesity and worsens established obesity by blunting sympathetic activity. Lastly, hypothalamic TGR5 signaling is required for the anti-obesity action of dietary BA supplementation. Together, these findings identify hypothalamic TGR5 signaling as a key mediator of a top-down neural mechanism that counteracts diet-induced obesity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
26. Impaired Very-Low-Density Lipoprotein catabolism links hypoglycemia to hypertriglyceridemia in Glycogen Storage Disease type Ia.
- Author
-
Hoogerland JA, Peeks F, Hijmans BS, Wolters JC, Kooijman S, Bos T, Bleeker A, van Dijk TH, Wolters H, Gerding A, van Eunen K, Havinga R, Pronk ACM, Rensen PCN, Mithieux G, Rajas F, Kuipers F, Reijngoud DJ, Derks TGJ, and Oosterveer MH
- Subjects
- Adult, Aged, Animals, Disease Models, Animal, Fatty Liver etiology, Female, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I metabolism, Hepatocytes metabolism, Humans, Hypertriglyceridemia prevention & control, Hypoglycemia metabolism, Lipid Metabolism, Male, Mice, Middle Aged, Glucose metabolism, Glycogen Storage Disease Type I complications, Hypertriglyceridemia etiology, Hypoglycemia etiology, Lipoproteins, VLDL metabolism, Triglycerides metabolism
- Abstract
Prevention of hypertriglyceridemia is one of the biomedical targets in Glycogen Storage Disease type Ia (GSD Ia) patients, yet it is unclear how hypoglycemia links to plasma triglyceride (TG) levels. We analyzed whole-body TG metabolism in normoglycemic (fed) and hypoglycemic (fasted) hepatocyte-specific glucose-6-phosphatase deficient (L-G6pc
-/- ) mice. De novo fatty acid synthesis contributed substantially to hepatic TG accumulation in normoglycemic L-G6pc-/- mice. In hypoglycemic conditions, enhanced adipose tissue lipolysis was the main driver of liver steatosis, supported by elevated free fatty acid concentrations in GSD Ia mice and GSD Ia patients. Plasma very-low-density lipoprotein (VLDL) levels were increased in GSD Ia patients and in normoglycemic L-G6pc-/- mice, and further elevated in hypoglycemic L-G6pc-/- mice. VLDL-TG secretion rates were doubled in normo- and hypoglycemic L-G6pc-/- mice, while VLDL-TG catabolism was selectively inhibited in hypoglycemic L-G6pc-/- mice. In conclusion, fasting-induced hypoglycemia in L-G6pc-/- mice promotes adipose tissue lipolysis and arrests VLDL catabolism. This mechanism likely contributes to aggravated liver steatosis and dyslipidemia in GSD Ia patients with poor glycemic control and may explain clinical heterogeneity in hypertriglyceridemia between GSD Ia patients., (© 2021 The Authors. Journal of Inherited Metabolic Disease published by John Wiley & Sons Ltd on behalf of SSIEM.)- Published
- 2021
- Full Text
- View/download PDF
27. Tamoxifen Treatment in the Neonatal Period Affects Glucose Homeostasis in Adult Mice in a Sex-Dependent Manner.
- Author
-
Estrada-Meza J, Videlo J, Bron C, Saint-Béat C, Silva M, Duboeuf F, Peyruchaud O, Rajas F, Mithieux G, and Gautier-Stein A
- Subjects
- Animals, Body Composition drug effects, Body Weight drug effects, Diet, High-Fat adverse effects, Female, Insulin Resistance physiology, Male, Mice, Mice, Inbred C57BL, Obesity etiology, Physical Conditioning, Animal, Selective Estrogen Receptor Modulators pharmacology, Animals, Newborn metabolism, Blood Glucose metabolism, Energy Metabolism drug effects, Homeostasis drug effects, Sex Factors, Tamoxifen pharmacology
- Abstract
Tamoxifen is a selective estrogen receptor modulator used to activate the CREERT2 recombinase, allowing tissue-specific and temporal control of the somatic mutagenesis to generate transgenic mice. Studies integrating development and metabolism require a genetic modification induced by a neonatal tamoxifen administration. Here, we investigate the effects of a neonatal tamoxifen administration on energy homeostasis in adult male and female C57BL/6J mice. C57BL/6J male and female mouse pups received a single injection of tamoxifen 1 day after birth (NTT) and were fed a high-fat/high-sucrose diet at 6 weeks of age. We measured weight, body composition, glucose and insulin tolerance, basal metabolism, and tibia length and weight in adult mice. The neonatal tamoxifen administration exerted long-term, sex-dependent effects on energy homeostasis. NTT female mice became overweight and developed impaired glucose control in comparison to vehicle-treated littermates. NTT females exhibited 60% increased fat mass, increased food intake, decreased physical activity and energy expenditure, impaired glucose and insulin tolerance, and fasting hyperglycemia and hyperinsulinemia. In contrast, NTT male mice exhibited a modest amelioration of glucose and insulin tolerance and long-term decreased lean mass linked to decreased bone weight. These results suggest that the neonatal tamoxifen administration exerted a marked and sex-dependent influence on adult energy homeostasis and bone weight and must therefore be used with caution for the development of transgenic mouse models regarding studies on energy homeostasis and bone biology., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
28. mRNA therapy restores euglycemia and prevents liver tumors in murine model of glycogen storage disease.
- Author
-
Cao J, Choi M, Guadagnin E, Soty M, Silva M, Verzieux V, Weisser E, Markel A, Zhuo J, Liang S, Yin L, Frassetto A, Graham AR, Burke K, Ketova T, Mihai C, Zalinger Z, Levy B, Besin G, Wolfrom M, Tran B, Tunkey C, Owen E, Sarkis J, Dousis A, Presnyak V, Pepin C, Zheng W, Ci L, Hard M, Miracco E, Rice L, Nguyen V, Zimmer M, Rajarajacholan U, Finn PF, Mithieux G, Rajas F, Martini PGV, and Giangrande PH
- Subjects
- Animals, Cell Line, Tumor, Cytokines blood, Cytokines metabolism, Glucose-6-Phosphatase metabolism, Glycogen metabolism, Glycogen Storage Disease genetics, Glycogen Storage Disease pathology, HeLa Cells, Humans, Liver metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, Nanoparticles administration & dosage, Nanoparticles chemistry, RNA, Messenger administration & dosage, RNA, Messenger chemistry, Treatment Outcome, Triglycerides metabolism, Mice, Disease Models, Animal, Genetic Therapy methods, Glucose-6-Phosphatase genetics, Glycogen Storage Disease therapy, RNA, Messenger genetics
- Abstract
Glycogen Storage Disease 1a (GSD1a) is a rare, inherited metabolic disorder caused by deficiency of glucose 6-phosphatase (G6Pase-α). G6Pase-α is critical for maintaining interprandial euglycemia. GSD1a patients exhibit life-threatening hypoglycemia and long-term liver complications including hepatocellular adenomas (HCAs) and carcinomas (HCCs). There is no treatment for GSD1a and the current standard-of-care for managing hypoglycemia (Glycosade
® /modified cornstarch) fails to prevent HCA/HCC risk. Therapeutic modalities such as enzyme replacement therapy and gene therapy are not ideal options for patients due to challenges in drug-delivery, efficacy, and safety. To develop a new treatment for GSD1a capable of addressing both the life-threatening hypoglycemia and HCA/HCC risk, we encapsulated engineered mRNAs encoding human G6Pase-α in lipid nanoparticles. We demonstrate the efficacy and safety of our approach in a preclinical murine model that phenotypically resembles the human condition, thus presenting a potential therapy that could have a significant therapeutic impact on the treatment of GSD1a.- Published
- 2021
- Full Text
- View/download PDF
29. Intestinal gluconeogenesis and protein diet: future directions.
- Author
-
Gautier-Stein A, Rajas F, and Mithieux G
- Subjects
- Animals, Glucose, Mice, Obesity prevention & control, Satiation, Gluconeogenesis, Intestines
- Abstract
High-protein meals and foods are promoted for their beneficial effects on satiety, weight loss and glucose homeostasis. However, the mechanisms involved and the long-term benefits of such diets are still debated. We here review how the characterisation of intestinal gluconeogenesis (IGN) sheds new light on the mechanisms by which protein diets exert their beneficial effects on health. The small intestine is the third organ (in addition to the liver and kidney) contributing to endogenous glucose production via gluconeogenesis. The particularity of glucose produced by the intestine is that it is detected in the portal vein and initiates a nervous signal to the hypothalamic nuclei regulating energy homeostasis. In this context, we demonstrated that protein diets initiate their satiety effects indirectly via IGN and portal glucose sensing. This induction results in the activation of brain areas involved in the regulation of food intake. The μ-opioid-antagonistic properties of protein digests, exerted in the portal vein, are a key link between IGN induction and protein-enriched diet in the control of satiety. From our results, IGN can be proposed as a mandatory link between nutrient sensing and the regulation of whole-body homeostasis. The use of specific mouse models targeting IGN should allow us to identify several metabolic functions that could be controlled by protein diets. This will lead to the characterisation of the mechanisms by which protein diets improve whole-body homeostasis. These data could be the basis of novel nutritional strategies targeting the serious metabolic consequences of both obesity and diabetes.
- Published
- 2021
- Full Text
- View/download PDF
30. The absence of hepatic glucose-6 phosphatase/ChREBP couple is incompatible with survival in mice.
- Author
-
Rajas F, Dentin R, Cannella Miliano A, Silva M, Raffin M, Levavasseur F, Gautier-Stein A, Postic C, and Mithieux G
- Subjects
- Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Glucose metabolism, Glucose-6-Phosphatase metabolism, Glucose-6-Phosphate metabolism, Hepatocytes metabolism, Hydrolysis, Lipids physiology, Liver pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Non-alcoholic Fatty Liver Disease physiopathology, Non-alcoholic Fatty Liver Disease prevention & control, Phosphoric Monoester Hydrolases genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Lipid Metabolism physiology, Phosphoric Monoester Hydrolases metabolism
- Abstract
Objective: Glucose production in the blood requires the expression of glucose-6 phosphatase (G6Pase), a key enzyme that allows glucose-6 phosphate (G6P) hydrolysis into free glucose and inorganic phosphate. We previously reported that the hepatic suppression of G6Pase leads to G6P accumulation and to metabolic reprogramming in hepatocytes from liver G6Pase-deficient mice (L.G6pc
-/- ). Interestingly, the activity of the transcription factor carbohydrate response element-binding protein (ChREBP), central for de novo lipid synthesis, is markedly activated in L.G6pc-/- mice, which consequently rapidly develop NAFLD-like pathology. In the current work, we assessed whether a selective deletion of ChREBP could prevent hepatic lipid accumulation and NAFLD initiation in L.G6pc-/- mice., Methods: We generated liver-specific ChREBP (L.Chrebp-/- )- and/or G6Pase (L.G6pc-/- )-deficient mice using a Cre-lox strategy in B6.SACreERT2 mice. Mice were fed a standard chow diet or a high-fat diet for 10 days. Markers of hepatic metabolism and cellular stress were analysed in the liver of control, L. G6pc-/- , L. Chrebp-/- and double knockout (i.e., L.G6pc-/- .Chrebp-/- ) mice., Results: We observed that there was a dramatic decrease in lipid accumulation in the liver of L.G6pc-/- .Chrebp-/- mice. At the mechanistic level, elevated G6P concentrations caused by lack of G6Pase are rerouted towards glycogen synthesis. Importantly, this exacerbated glycogen accumulation, leading to hepatic water retention and aggravated hepatomegaly. This caused animal distress and hepatocyte damage, characterised by ballooning and moderate fibrosis, paralleled with acute endoplasmic reticulum stress., Conclusions: Our study reveals the crucial role of the ChREBP-G6Pase duo in the regulation of G6P-regulated pathways in the liver., (Copyright © 2020 The Authors. Published by Elsevier GmbH.. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
31. Calcitonin Gene-Related Peptide-Induced Phosphorylation of STAT3 in Arcuate Neurons Is a Link in the Metabolic Benefits of Portal Glucose.
- Author
-
Soty M, Vily-Petit J, Castellanos-Jankiewicz A, Guzman-Quevedo O, Raffin M, Clark S, Silva M, Gautier-Stein A, Cota D, and Mithieux G
- Subjects
- Animals, Behavior, Animal drug effects, Behavior, Animal physiology, Calcitonin Gene-Related Peptide deficiency, Eating drug effects, Eating physiology, Glucose administration & dosage, Infusions, Intravenous, Leptin deficiency, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Neurons metabolism, Phosphorylation physiology, Portal Vein, Arcuate Nucleus of Hypothalamus metabolism, Calcitonin Gene-Related Peptide metabolism, Gluconeogenesis physiology, Glucose pharmacology, Intestines metabolism, Leptin metabolism, STAT3 Transcription Factor metabolism
- Abstract
Introduction: Intestinal gluconeogenesis (IGN) exerts metabolic benefits in energy homeostasis via the neural sensing of portal glucose., Objective: The aim of this work was to determine central mechanisms involved in the effects of IGN on the control of energy homeostasis., Methods: We investigated the effects of glucose infusion into the portal vein, at a rate that mimics IGN, in conscious wild-type, leptin-deficient Ob/Ob and calcitonin gene-related peptide (CGRP)-deficient mice., Results: We report that portal glucose infusion decreases food intake and plasma glucose and induces in the hypothalamic arcuate nucleus (ARC) the phosphorylation of STAT3, the classic intracellular messenger of leptin signaling. This notably takes place in POMC-expressing neurons. STAT3 phosphorylation does not require leptin, since portal glucose effects are observed in leptin-deficient Ob/Ob mice. We hypothesized that the portal glucose effects could require CGRP, a neuromediator previously suggested to suppress hunger. In line with this hypothesis, neither the metabolic benefits nor the phosphorylation of STAT3 in the ARC take place upon portal glucose infusion in CGRP-deficient mice. Moreover, intracerebroventricular injection of CGRP activates hypothalamic phosphorylation of STAT3 in mice, and CGRP does the same in hypothalamic cells. Finally, no metabolic benefit of dietary fibers (known to depend on the induction of IGN), takes place in CGRP-deficient mice., Conclusions: CGRP-induced phosphorylation of STAT3 in the ARC is part of the neural chain determining the hunger-modulating and glucose-lowering effects of IGN/portal glucose., (© 2020 S. Karger AG, Basel.)
- Published
- 2021
- Full Text
- View/download PDF
32. Dietary Fibers and Proteins Modulate Behavior via the Activation of Intestinal Gluconeogenesis.
- Author
-
Sinet F, Soty M, Zemdegs J, Guiard B, Estrada J, Malleret G, Silva M, Mithieux G, and Gautier-Stein A
- Subjects
- Animals, Disease Models, Animal, Mice, Anxiety metabolism, Behavior, Animal physiology, Depression metabolism, Dietary Fiber metabolism, Dietary Proteins metabolism, Gluconeogenesis physiology, Intestine, Small metabolism
- Abstract
Introduction: Several studies have suggested that diet, especially the one enriched in microbiota-fermented fibers or fat, regulates behavior. The underlying mechanisms are currently unknown. We previously reported that certain macronutrients (fermentable fiber and protein) regulate energy homeostasis via the activation of intestinal gluconeogenesis (IGN), which generates a neural signal to the brain. We hypothesized that these nutriments might control behavior using the same gut-brain circuit., Methods: Wild-type and IGN-deficient mice were fed chow or diets enriched in protein or fiber. Changes in their behavior were assessed using suited tests. Hippocampal neurogenesis, extracellular levels of serotonin, and protein expression levels were assessed by immunofluorescence, in vivo dialysis, and Western blotting, respectively. IGN was rescued by infusing glucose into the portal vein of IGN-deficient mice., Results: We show here that both fiber- and protein-enriched diets exert beneficial actions on anxiety-like and depressive-like behaviors. These benefits do not occur in mice lacking IGN. Consistently, IGN-deficient mice display hallmarks of depressive-like disorders, including decreased hippocampal neurogenesis, basal hyperactivity, and deregulation of the hypothalamic-pituitary-adrenal axis, which are associated with increased expression of the precursor of corticotropin-releasing hormone in the hypothalamus and decreased expression of the glucocorticoid receptor in the hippocampus. These neurobiological alterations are corrected by portal glucose infusion mimicking IGN., Conclusion: IGN translates nutritional information, allowing the brain to finely coordinate energy metabolism and behavior., (© 2021 S. Karger AG, Basel.)
- Published
- 2021
- Full Text
- View/download PDF
33. Intestinal gluconeogenesis prevents obesity-linked liver steatosis and non-alcoholic fatty liver disease.
- Author
-
Vily-Petit J, Soty-Roca M, Silva M, Raffin M, Gautier-Stein A, Rajas F, and Mithieux G
- Subjects
- Animals, Chemokine CCL2 metabolism, Diet, High-Fat, Interleukin-6 metabolism, Liver innervation, Liver metabolism, Mice, Knockout, Mice, Transgenic, Neurons metabolism, Tumor Necrosis Factor-alpha metabolism, Tyrosine 3-Monooxygenase metabolism, Gastrointestinal Tract metabolism, Gluconeogenesis physiology, Non-alcoholic Fatty Liver Disease prevention & control, Obesity physiopathology
- Abstract
Objective: Hepatic steatosis accompanying obesity is a major health concern, since it may initiate non-alcoholic fatty liver disease (NAFLD) and associated complications like cirrhosis or cancer. Intestinal gluconeogenesis (IGN) is a recently described function that contributes to the metabolic benefits of specific macronutrients as protein or soluble fibre, via the initiation of a gut-brain nervous signal triggering brain-dependent regulations of peripheral metabolism. Here, we investigate the effects of IGN on liver metabolism, independently of its induction by the aforementioned macronutrients., Design: To study the specific effects of IGN on hepatic metabolism, we used two transgenic mouse lines: one is knocked down for and the other overexpresses glucose-6-phosphatase, the key enzyme of endogenous glucose production, specifically in the intestine., Results: We report that mice with a genetic overexpression of IGN are notably protected from the development of hepatic steatosis and the initiation of NAFLD on a hypercaloric diet. The protection relates to a diminution of de novo lipogenesis and lipid import, associated with benefits at the level of inflammation and fibrosis and linked to autonomous nervous system. Conversely, mice with genetic suppression of IGN spontaneously exhibit increased hepatic triglyceride storage associated with activated lipogenesis pathway, in the context of standard starch-enriched diet. The latter is corrected by portal glucose infusion mimicking IGN., Conclusion: We conclude that IGN per se has the capacity of preventing hepatic steatosis and its eventual evolution toward NAFLD., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
34. Hepatic Carbohydrate Response Element Binding Protein Activation Limits Nonalcoholic Fatty Liver Disease Development in a Mouse Model for Glycogen Storage Disease Type 1a.
- Author
-
Lei Y, Hoogerland JA, Bloks VW, Bos T, Bleeker A, Wolters H, Wolters JC, Hijmans BS, van Dijk TH, Thomas R, van Weeghel M, Mithieux G, Houtkooper RH, de Bruin A, Rajas F, Kuipers F, and Oosterveer MH
- Subjects
- Adipose Tissue, White metabolism, Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Dependovirus genetics, Disease Models, Animal, Gene Knockdown Techniques, Genetic Vectors administration & dosage, Genetic Vectors genetics, Glucose-6-Phosphatase genetics, Glycogen metabolism, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I metabolism, Glycolysis, Hepatocytes, Humans, Lipogenesis, Lipoproteins, VLDL metabolism, Male, Mice, Mice, Knockout, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease pathology, RNA, Small Interfering genetics, Triglycerides metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Glycogen Storage Disease Type I complications, Liver pathology, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Background and Aims: Glycogen storage disease (GSD) type 1a is an inborn error of metabolism caused by defective glucose-6-phosphatase catalytic subunit (G6PC) activity. Patients with GSD 1a exhibit severe hepatomegaly due to glycogen and triglyceride (TG) accumulation in the liver. We have shown that the activity of carbohydrate response element binding protein (ChREBP), a key regulator of glycolysis and de novo lipogenesis, is increased in GSD 1a. In the current study, we assessed the contribution of ChREBP to nonalcoholic fatty liver disease (NAFLD) development in a mouse model for hepatic GSD 1a., Approach and Results: Liver-specific G6pc-knockout (L-G6pc
-/- ) mice were treated with adeno-associated viruses (AAVs) 2 or 8 directed against short hairpin ChREBP to normalize hepatic ChREBP activity to levels observed in wild-type mice receiving AAV8-scrambled short hairpin RNA (shSCR). Hepatic ChREBP knockdown markedly increased liver weight and hepatocyte size in L-G6pc-/- mice. This was associated with hepatic accumulation of G6P, glycogen, and lipids, whereas the expression of glycolytic and lipogenic genes was reduced. Enzyme activities, flux measurements, hepatic metabolite analysis and very low density lipoprotein (VLDL)-TG secretion assays revealed that hepatic ChREBP knockdown reduced downstream glycolysis and de novo lipogenesis but also strongly suppressed hepatic VLDL lipidation, hence promoting the storage of "old fat." Interestingly, enhanced VLDL-TG secretion in shSCR-treated L-G6pc-/- mice associated with a ChREBP-dependent induction of the VLDL lipidation proteins microsomal TG transfer protein and transmembrane 6 superfamily member 2 (TM6SF2), the latter being confirmed by ChIP-qPCR., Conclusions: Attenuation of hepatic ChREBP induction in GSD 1a liver aggravates hepatomegaly because of further accumulation of glycogen and lipids as a result of reduced glycolysis and suppressed VLDL-TG secretion. TM6SF2, critical for VLDL formation, was identified as a ChREBP target in mouse liver. Altogether, our data show that enhanced ChREBP activity limits NAFLD development in GSD 1a by balancing hepatic TG production and secretion., (© 2020 The Authors. Hepatology published by Wiley Periodicals, LLC., on behalf of American Association for the Study of Liver Diseases.)- Published
- 2020
- Full Text
- View/download PDF
35. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing.
- Author
-
Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB, Beal MF, Bergersen LH, Brinton RD, de la Monte S, Eckert A, Harvey J, Jeggo R, Jhamandas JH, Kann O, la Cour CM, Martin WF, Mithieux G, Moreira PI, Murphy MP, Nave KA, Nuriel T, Oliet SHR, Saudou F, Mattson MP, Swerdlow RH, and Millan MJ
- Subjects
- Animals, Glycolysis physiology, Humans, Oxidative Phosphorylation, Aging physiology, Brain physiology, Energy Metabolism physiology, Neurodegenerative Diseases physiopathology
- Abstract
The brain requires a continuous supply of energy in the form of ATP, most of which is produced from glucose by oxidative phosphorylation in mitochondria, complemented by aerobic glycolysis in the cytoplasm. When glucose levels are limited, ketone bodies generated in the liver and lactate derived from exercising skeletal muscle can also become important energy substrates for the brain. In neurodegenerative disorders of ageing, brain glucose metabolism deteriorates in a progressive, region-specific and disease-specific manner - a problem that is best characterized in Alzheimer disease, where it begins presymptomatically. This Review discusses the status and prospects of therapeutic strategies for countering neurodegenerative disorders of ageing by improving, preserving or rescuing brain energetics. The approaches described include restoring oxidative phosphorylation and glycolysis, increasing insulin sensitivity, correcting mitochondrial dysfunction, ketone-based interventions, acting via hormones that modulate cerebral energetics, RNA therapeutics and complementary multimodal lifestyle changes.
- Published
- 2020
- Full Text
- View/download PDF
36. Short chain fatty acids in human gut and metabolic health.
- Author
-
Blaak EE, Canfora EE, Theis S, Frost G, Groen AK, Mithieux G, Nauta A, Scott K, Stahl B, van Harsselaar J, van Tol R, Vaughan EE, and Verbeke K
- Subjects
- Animals, Carbohydrate Metabolism, Diabetes Mellitus, Type 2 prevention & control, Host Microbial Interactions, Humans, Obesity prevention & control, Prebiotics, Fatty Acids, Volatile metabolism, Gastrointestinal Diseases prevention & control, Gastrointestinal Microbiome, Gastrointestinal Tract immunology, Gastrointestinal Tract microbiology
- Abstract
Evidence is accumulating that short chain fatty acids (SCFA) play an important role in the maintenance of gut and metabolic health. The SCFA acetate, propionate and butyrate are produced from the microbial fermentation of indigestible carbohydrates and appear to be key mediators of the beneficial effects elicited by the gut microbiome. Microbial SCFA production is essential for gut integrity by regulating the luminal pH, mucus production, providing fuel for epithelial cells and effects on mucosal immune function. SCFA also directly modulate host metabolic health through a range of tissue-specific mechanisms related to appetite regulation, energy expenditure, glucose homeostasis and immunomodulation. Therefore, an increased microbial SCFA production can be considered as a health benefit, but data are mainly based on animal studies, whereas well-controlled human studies are limited. In this review an expert group by ILSI Europe's Prebiotics Task Force discussed the current scientific knowledge on SCFA to consider the relationship between SCFA and gut and metabolic health with a particular focus on human evidence. Overall, the available mechanistic data and limited human data on the metabolic consequences of elevated gut-derived SCFA production strongly suggest that increasing SCFA production could be a valuable strategy in the preventing gastro-intestinal dysfunction, obesity and type 2 diabetes mellitus. Nevertheless, there is an urgent need for well controlled longer term human SCFA intervention studies, including measurement of SCFA fluxes and kinetics, the heterogeneity in response based on metabolic phenotype, the type of dietary fibre and fermentation site in fibre intervention studies and the control for factors that could shape the microbiome like diet, physical activity and use of medication.
- Published
- 2020
- Full Text
- View/download PDF
37. Atrial Natriuretic Peptide Orchestrates a Coordinated Physiological Response to Fuel Non-shivering Thermogenesis.
- Author
-
Carper D, Coué M, Nascimento EBM, Barquissau V, Lagarde D, Pestourie C, Laurens C, Petit JV, Soty M, Monbrun L, Marques MA, Jeanson Y, Sainte-Marie Y, Mairal A, Déjean S, Tavernier G, Viguerie N, Bourlier V, Lezoualc'h F, Carrière A, Saris WHM, Astrup A, Casteilla L, Mithieux G, van Marken Lichtenbelt W, Langin D, Schrauwen P, and Moro C
- Subjects
- Animals, Humans, Male, Mice, Mice, Knockout, Atrial Natriuretic Factor metabolism, Thermogenesis physiology
- Abstract
Atrial natriuretic peptide (ANP) is a cardiac hormone controlling blood volume and pressure in mammals. It is still unclear whether ANP controls cold-induced thermogenesis in vivo. Here, we show that acute cold exposure induces cardiac ANP secretion in mice and humans. Genetic inactivation of ANP promotes cold intolerance and suppresses half of cold-induced brown adipose tissue (BAT) activation in mice. While white adipocytes are resistant to ANP-mediated lipolysis at thermoneutral temperature in mice, cold exposure renders white adipocytes fully responsive to ANP to activate lipolysis and a thermogenic program, a physiological response that is dramatically suppressed in ANP null mice. ANP deficiency also blunts liver triglycerides and glycogen metabolism, thus impairing fuel availability for BAT thermogenesis. ANP directly increases mitochondrial uncoupling and thermogenic gene expression in human white and brown adipocytes. Together, these results indicate that ANP is a major physiological trigger of BAT thermogenesis upon cold exposure in mammals., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
38. [Sensing of blood glucose by the nervous system: why, where, how?]
- Author
-
Mithieux G
- Subjects
- Animals, Blood Glucose metabolism, Energy Metabolism physiology, Humans, Metabolic Networks and Pathways physiology, Nervous System Physiological Phenomena, Blood Glucose physiology, Nervous System metabolism
- Abstract
Maintaining plasma glucose concentration is a critical requirement for the body, making the detection of blood glucose by the nervous system essential. The reason generally given is that glucose is the main source of energy for living cells. In fact, other reasons make that glucose is so crucial. Glucose feeds two non-oxidative metabolic pathways, glycolysis and the pentose pathway, which allow the synthesis of molecules essential for cell survival and division, such as amino acids or nucleotides. The purpose of this review is to argue this statement and discuss the processes of glucose sensing by the central and peripheral nervous systems.
- Published
- 2020
- Full Text
- View/download PDF
39. Glycogen storage disease type 1a is associated with disturbed vitamin A metabolism and elevated serum retinol levels.
- Author
-
Saeed A, Hoogerland JA, Wessel H, Heegsma J, Derks TGJ, van der Veer E, Mithieux G, Rajas F, Oosterveer MH, and Faber KN
- Subjects
- Adolescent, Adult, Animals, Diterpenes metabolism, Fatty Liver metabolism, Female, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I blood, Glycogen Storage Disease Type I enzymology, Glycogen Storage Disease Type I pathology, Humans, Inflammation genetics, Inflammation metabolism, Liver pathology, Male, Mice, Mice, Knockout, Osteoporosis metabolism, Retinoic Acid 4-Hydroxylase genetics, Retinoic Acid 4-Hydroxylase metabolism, Retinol-Binding Proteins, Plasma genetics, Retinyl Esters, Vitamin A analogs & derivatives, Vitamin A metabolism, Glucose-6-Phosphatase metabolism, Glycogen Storage Disease Type I metabolism, Liver metabolism, Retinol-Binding Proteins, Plasma metabolism, Vitamin A blood
- Abstract
Glycogen storage disease type 1a (GSD Ia) is an inborn error of metabolism caused by mutations in the G6PC gene, encoding the catalytic subunit of glucose-6-phosphatase. Early symptoms include severe fasting intolerance, failure to thrive and hepatomegaly, biochemically associated with nonketotic hypoglycemia, fasting hyperlactidemia, hyperuricemia and hyperlipidemia. Dietary management is the cornerstone of treatment aiming at maintaining euglycemia, prevention of secondary metabolic perturbations and long-term complications, including liver (hepatocellular adenomas and carcinomas), kidney and bone disease (hypovitaminosis D and osteoporosis). As impaired vitamin A homeostasis also associates with similar symptoms and is coordinated by the liver, we here analysed whether vitamin A metabolism is affected in GSD Ia patients and liver-specific G6pc-/- knock-out mice. Serum levels of retinol and retinol binding protein 4 (RBP4) were significantly increased in both GSD Ia patients and L-G6pc-/- mice. In contrast, hepatic retinol levels were significantly reduced in L-G6pc-/- mice, while hepatic retinyl palmitate (vitamin A storage form) and RBP4 levels were not altered. Transcript and protein analyses indicate an enhanced production of retinol and reduced conversion the retinoic acids (unchanged LRAT, Pnpla2/ATGL and Pnpla3 up, Cyp26a1 down) in L-G6pc-/- mice. Aberrant expression of genes involved in vitamin A metabolism was associated with reduced basal messenger RNA levels of markers of inflammation (Cd68, Tnfα, Nos2, Il-6) and fibrosis (Col1a1, Acta2, Tgfβ, Timp1) in livers of L-G6pc-/- mice. In conclusion, GSD Ia is associated with elevated serum retinol and RBP4 levels, which may contribute to disease symptoms, including osteoporosis and hepatic steatosis., (© The Author(s) 2019. Published by Oxford University Press.)
- Published
- 2020
- Full Text
- View/download PDF
40. Jejunal Insulin Signalling Is Increased in Morbidly Obese Subjects with High Insulin Resistance and Is Regulated by Insulin and Leptin.
- Author
-
Gutierrez-Repiso C, Ho-Plagaro A, Santiago-Fernandez C, Garcia-Serrano S, Rodríguez-Pacheco F, Valdes S, Garrido-Sanchez L, Rodríguez-Díaz C, López-Gómez C, Moreno-Ruiz FJ, Alcain-Martinez G, Gautier-Stein A, Mithieux G, and Garcia-Fuentes E
- Abstract
Little is known about the jejunal insulin signalling pathways in insulin resistance/diabetes states and their possible regulation by insulin/leptin. We study in jejunum the relation between insulin signalling and insulin resistance in morbidly obese subjects with low (MO-low-IR) or with high insulin resistance (MO-high-IR), and with type 2 diabetes treated with metformin (MO-metf-T2DM)), and the effect of insulin/leptin on intestinal epithelial cells (IEC). Insulin receptor substrate-1 (IRS1) and the catalytic p110β subunit (p110β) of phosphatidylinositol 3-kinase (PI3K) were higher in MO-high-IR than in MO-low-IR. The regulatory p85α subunit of PI3K (p85α)/p110β ratio was lower in MO-high-IR and MO-metf-T2DM than in MO-low-IR. Akt-phosphorylation in Ser473 was reduced in MO-high-IR compared with MO-low-IR. IRS1 and p110-β were associated with insulin and leptin levels. The improvement of body mass index (BMI) and HOMA-IR (homeostasis model assessment of insulin resistance index) after bariatric surgery was associated with a higher IRS1 and a lower p85α/p110β ratio. IEC (intestinal epithelial cells) incubation with a high glucose + insulin dose produced an increase of p85α and p110β. High dose of leptin produced an increase of IRS1, p85α and p110β. In conclusion, despite the existence of insulin resistance, the jejunal expression of genes involved in insulin signalling was increased in MO-high-IR. Their expressions were regulated mainly by leptin. IRS1 and p85α/p110β ratio was associated with the evolution of insulin resistance after bariatric surgery., Competing Interests: The authors declare that they have no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
41. Metabolic benefits of gastric bypass surgery in the mouse: The role of fecal losses.
- Author
-
Barataud A, Vily-Petit J, Goncalves D, Zitoun C, Duchampt A, Philippe E, Gautier-Stein A, and Mithieux G
- Subjects
- Animals, Body Weight, Male, Mice, Mice, Inbred C57BL, Weight Loss, Gastric Bypass, Obesity metabolism, Obesity surgery
- Abstract
Objective: Roux-en-Y gastric surgery (RYGB) promotes a rapid and sustained weight loss and amelioration of glucose control in obese patients. A high number of molecular hypotheses were previously tested using duodenal-jejunal bypass (DJB) performed in various genetic models of mice with knockouts for various hormones or receptors. The data were globally negative or inconsistent. Therefore, the mechanisms remained elusive. Intestinal gluconeogenesis is a gut function that has been suggested to contribute to the metabolic benefits of RYGB in obese patients., Methods: We studied the effects of DJB on body weight and glucose control in obese mice fed a high fat-high sucrose diet. Wild type mice and mice with a genetic suppression of intestinal gluconeogenesis were studied in parallel using glucose- and insulin-tolerance tests. Fecal losses, including excretion of lipids, were studied from the feces recovered in metabolic cages., Results: DJB induced a dramatic decrease in body weight and improvement in glucose control (glucose- and insulin-tolerance) in obese wild type mice fed a high calorie diet, for 25 days after the surgery. The DJB-induced decrease in food intake was transient and resumed to normal in 7-8 days, suggesting that decreased food intake could not account for the benefits. Total fecal losses were about 5 times and lipid losses 7 times higher in DJB-mice than in control (sham-operated and pair-fed) mice, and could account for the weight loss of mice. The results were comparable in mice with suppression of intestinal gluconeogenesis. There was no effect of DJB on food intake, body weight or fecal loss in lean mice fed a normal chow diet., Conclusions: DJB in obese mice fed a high calorie diet promotes dramatic fecal loss, which could account for the dramatic weight loss and metabolic benefits observed. This could dominate the effects of the mouse genotype/phenotype. Thus, fecal energy loss should be considered as an essential process contributing to the metabolic benefits of DJB in obese mice., (Copyright © 2019 The Author(s). Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
42. Glucose-6-Phosphate Regulates Hepatic Bile Acid Synthesis in Mice.
- Author
-
Hoogerland JA, Lei Y, Wolters JC, de Boer JF, Bos T, Bleeker A, Mulder NL, van Dijk TH, Kuivenhoven JA, Rajas F, Mithieux G, Haeusler RA, Verkade HJ, Bloks VW, Kuipers F, and Oosterveer MH
- Subjects
- Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors physiology, Cholesterol metabolism, Humans, Intestinal Mucosa metabolism, Male, Mice, Mice, Inbred C57BL, Steroid 12-alpha-Hydroxylase physiology, Bile Acids and Salts biosynthesis, Glucose-6-Phosphate physiology, Liver metabolism
- Abstract
It is well established that, besides facilitating lipid absorption, bile acids act as signaling molecules that modulate glucose and lipid metabolism. Bile acid metabolism, in turn, is controlled by several nutrient-sensitive transcription factors. Altered intrahepatic glucose signaling in type 2 diabetes associates with perturbed bile acid synthesis. We aimed to characterize the regulatory role of the primary intracellular metabolite of glucose, glucose-6-phosphate (G6P), on bile acid metabolism. Hepatic gene expression patterns and bile acid composition were analyzed in mice that accumulate G6P in the liver, that is, liver-specific glucose-6-phosphatase knockout (L-G6pc
-/- ) mice, and mice treated with a pharmacological inhibitor of the G6P transporter. Hepatic G6P accumulation induces sterol 12α-hydroxylase (Cyp8b1) expression, which is mediated by the major glucose-sensitive transcription factor, carbohydrate response element-binding protein (ChREBP). Activation of the G6P-ChREBP-CYP8B1 axis increases the relative abundance of cholic-acid-derived bile acids and induces physiologically relevant shifts in bile composition. The G6P-ChREBP-dependent change in bile acid hydrophobicity associates with elevated plasma campesterol/cholesterol ratio and reduced fecal neutral sterol loss, compatible with enhanced intestinal cholesterol absorption. Conclusion: We report that G6P, the primary intracellular metabolite of glucose, controls hepatic bile acid synthesis. Our work identifies hepatic G6P-ChREBP-CYP8B1 signaling as a regulatory axis in control of bile acid and cholesterol metabolism., (© 2019 The Authors. Hepatology published by Wiley Periodicals, Inc., on behalf of American Association for the Study of Liver Diseases.)- Published
- 2019
- Full Text
- View/download PDF
43. Glucose-6 Phosphate, A Central Hub for Liver Carbohydrate Metabolism.
- Author
-
Rajas F, Gautier-Stein A, and Mithieux G
- Abstract
: Cells efficiently adjust their metabolism according to the abundance of nutrients and energy. The ability to switch cellular metabolism between anabolic and catabolic processes is critical for cell growth. Glucose-6 phosphate is the first intermediate of glucose metabolism and plays a central role in the energy metabolism of the liver. It acts as a hub to metabolically connect glycolysis, the pentose phosphate pathway, glycogen synthesis, de novo lipogenesis, and the hexosamine pathway. In this review, we describe the metabolic fate of glucose-6 phosphate in a healthy liver and the metabolic reprogramming occurring in two pathologies characterized by a deregulation of glucose homeostasis, namely type 2 diabetes, which is characterized by fasting hyperglycemia; and glycogen storage disease type I, where patients develop severe hypoglycemia during short fasting periods. In these two conditions, dysfunction of glucose metabolism results in non-alcoholic fatty liver disease, which may possibly lead to the development of hepatic tumors. Moreover, we also emphasize the role of the transcription factor carbohydrate response element-binding protein (ChREBP), known to link glucose and lipid metabolisms. In this regard, comparing these two metabolic diseases is a fruitful approach to better understand the key role of glucose-6 phosphate in liver metabolism in health and disease.
- Published
- 2019
- Full Text
- View/download PDF
44. Pathogenesis of Hepatic Tumors following Gene Therapy in Murine and Canine Models of Glycogen Storage Disease.
- Author
-
Kang HR, Gjorgjieva M, Smith SN, Brooks ED, Chen Z, Burgess SM, Chandler RJ, Waskowicz LR, Grady KM, Li S, Mithieux G, Venditti CP, Rajas F, and Koeberl DD
- Abstract
Glycogen storage disease type Ia (GSD Ia) is caused by mutations in the glucose-6-phosphatase (G6Pase) catalytic subunit gene ( G6PC ). GSD Ia complications include hepatocellular adenomas (HCA) with a risk for hepatocellular carcinoma (HCC) formation. Genome editing with adeno-associated virus (AAV) vectors containing a zinc-finger nuclease (ZFN) and a G6PC donor transgene was evaluated in adult mice with GSD Ia. Although mouse livers expressed G6Pase, HCA and HCC occurred following AAV vector administration. Interestingly, vector genomes were almost undetectable in the tumors but remained relatively high in adjacent liver (p < 0.01). G6Pase activity was decreased in tumors, in comparison with adjacent liver (p < 0.01). Furthermore, AAV-G6Pase vector-treated dogs with GSD Ia developed HCC with lower G6Pase activity (p < 0.01) in comparison with adjacent liver. AAV integration and tumor marker analysis in mice revealed that tumors arose from the underlying disorder, not from vector administration. Similarly to human GSD Ia-related HCA and HCC, mouse and dog tumors did not express elevated α-fetoprotein. Taken together, these results suggest that AAV-mediated gene therapy not only corrects hepatic G6Pase deficiency, but also has potential to suppress HCA and HCC in the GSD Ia liver., (© 2019 The Authors.)
- Published
- 2019
- Full Text
- View/download PDF
45. Challenges of Gene Therapy for the Treatment of Glycogen Storage Diseases Type I and Type III.
- Author
-
Jauze L, Monteillet L, Mithieux G, Rajas F, and Ronzitti G
- Subjects
- Animals, Clinical Trials as Topic, Dependovirus metabolism, Disease Models, Animal, Gene Transfer Techniques, Genetic Vectors chemistry, Genetic Vectors metabolism, Glucose-6-Phosphatase metabolism, Glycogen biosynthesis, Glycogen Storage Disease Type I enzymology, Glycogen Storage Disease Type I genetics, Glycogen Storage Disease Type I pathology, Glycogen Storage Disease Type III enzymology, Glycogen Storage Disease Type III genetics, Glycogen Storage Disease Type III pathology, Hepatocytes enzymology, Hepatocytes pathology, Humans, Hypoglycemia enzymology, Hypoglycemia genetics, Hypoglycemia pathology, Liver enzymology, Liver pathology, Transgenes, Dependovirus genetics, Genetic Therapy methods, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I therapy, Glycogen Storage Disease Type III therapy, Hypoglycemia therapy
- Abstract
Glycogen storage diseases (GSDs) type I (GSDI) and type III (GSDIII), the most frequent hepatic GSDs, are due to defects in glycogen metabolism, mainly in the liver. In addition to hypoglycemia and liver pathology, renal, myeloid, or muscle complications affect GSDI and GSDIII patients. Currently, patient management is based on dietary treatment preventing severe hypoglycemia and increasing the lifespan of patients. However, most of the patients develop long-term pathologies. In the past years, gene therapy for GSDI has generated proof of concept for hepatic GSDs. This resulted in a recent clinical trial of adeno-associated virus (AAV)-based gene replacement for GSDIa. However, the current limitations of AAV-mediated gene transfer still represent a challenge for successful gene therapy in GSDI and GSDIII. Indeed, transgene loss over time was observed in GSDI liver, possibly due to the degeneration of hepatocytes underlying the physiopathology of both GSDI and GSDIII and leading to hepatic tumor development. Moreover, multitissue targeting requires high vector doses to target nonpermissive tissues such as muscle and kidney. Interestingly, recent pharmacological interventions or dietary regimen aiming at the amelioration of the hepatocyte abnormalities before the administration of gene therapy demonstrated improved efficacy in GSDs. In this review, we describe the advances in gene therapy and the limitations to be overcome to achieve efficient and safe gene transfer in GSDs.
- Published
- 2019
- Full Text
- View/download PDF
46. Hepatic stress associated with pathologies characterized by disturbed glucose production.
- Author
-
Gjorgjieva M, Mithieux G, and Rajas F
- Abstract
The liver is an organ with many facets, including a role in energy production and metabolic balance, detoxification and extraordinary capacity of regeneration. Hepatic glucose production plays a crucial role in the maintenance of normal glucose levels in the organism i.e. between 0.7 to 1.1 g/l. The loss of this function leads to a rare genetic metabolic disease named glycogen storage disease type I (GSDI), characterized by severe hypoglycemia during short fasts. On the contrary, type 2 diabetes is characterized by chronic hyperglycemia, partly due to an overproduction of glucose by the liver. Indeed, diabetes is characterized by increased uptake/production of glucose by hepatocytes, leading to the activation of de novo lipogenesis and the development of a non-alcoholic fatty liver disease. In GSDI, the accumulation of glucose-6 phosphate, which cannot be hydrolyzed into glucose, leads to an increase of glycogen stores and the development of hepatic steatosis. Thus, in these pathologies, hepatocytes are subjected to cellular stress mainly induced by glucotoxicity and lipotoxicity. In this review, we have compared hepatic cellular stress induced in type 2 diabetes and GSDI, especially oxidative stress, autophagy deregulation, and ER-stress. In addition, both GSDI and diabetic patients are prone to the development of hepatocellular adenomas (HCA) that occur on a fatty liver in the absence of cirrhosis. These HCA can further acquire malignant traits and transform into hepatocellular carcinoma. This process of tumorigenesis highlights the importance of an optimal metabolic control in both GSDI and diabetic patients in order to prevent, or at least to restrain, tumorigenic activity during disturbed glucose metabolism pathologies., Competing Interests: Conflict of interest: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
47. Chronic intake of 4-Methylimidazole induces Hyperinsulinemia and Hypoglycaemia via Pancreatic Beta Cell Hyperplasia and Glucose Dyshomeostasis.
- Author
-
Rekha B, Velmurugan G, Freddy AJ, Anusha S, Ramprasath T, Karthik KV, Suresh S, Kulshrestha P, Mithieux G, Lyon AR, Selvam GS, and Ramasamy S
- Subjects
- Animals, Apoptosis drug effects, Female, Food Coloring Agents administration & dosage, Food Coloring Agents toxicity, Humans, Hyperplasia pathology, Insulin blood, Insulin-Secreting Cells pathology, Lipid Metabolism drug effects, Mice, Mice, Inbred BALB C, Oxidative Stress drug effects, Blood Glucose metabolism, Homeostasis drug effects, Hyperinsulinism chemically induced, Hypoglycemia chemically induced, Imidazoles administration & dosage, Imidazoles toxicity, Insulin-Secreting Cells metabolism
- Abstract
Caramel colours are the preferential food colouring agent globally, reaches wide age groups through eatables. Colas, a sweetened carbonated drink are most common caramel coloured beverage and its consumption is linked with diabetes, obesity, pancreatic cancer and other endocrine disorders. A major by-product produced during caramelization is 4-methylimidazole (4-MEI) that is detected in noteworthy concentrations in colas and other beverages. Previous studies revealed the neurotoxic and carcinogenic potential of 4-MEI in animals at higher doses but the effect of 4-MEI at theoretical maximum daily intake dose on glucose homeostasis is unexplored. Here, mice treated with 4-MEI (32 µg/kg bodyweight/day) for seven weeks exhibited severe hypoglycaemia and hyperinsulinemia mediated by hyperplasia of pancreatic beta cells and induces metabolic alterations. On combinatorial treatment, 4-MEI suppressed the glucogenic potential of non-artificial sweeteners and promotes lipogenesis. Furthermore, increased levels of C-peptide, LDL-cholesterol and triglycerides were observed in the humans with regular intake of 4-MEI containing beverages. In summary, 4-MEI induced pancreatic beta cell hyperplasia and leads to disruption of glucose and lipid homeostasis. This study suggests the need for further assessment and reconsideration of the wide usage of 4-MEI containing caramels as food additives.
- Published
- 2018
- Full Text
- View/download PDF
48. Polycystic kidney features of the renal pathology in glycogen storage disease type I: possible evolution to renal neoplasia.
- Author
-
Gjorgjieva M, Monteillet L, Calderaro J, Mithieux G, and Rajas F
- Subjects
- Animals, Disease Models, Animal, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I genetics, Hepatocyte Nuclear Factor 1-beta genetics, Humans, Kidney Neoplasms etiology, Kidney Neoplasms pathology, Mice, Mice, Knockout, Polycystic Kidney Diseases genetics, Polycystic Kidney Diseases pathology, Renal Insufficiency, Chronic pathology, Glycogen Storage Disease Type I complications, Polycystic Kidney Diseases etiology, Renal Insufficiency, Chronic etiology
- Abstract
Glycogen storage disease type I (GSDI) is a rare genetic pathology characterized by glucose-6 phosphatase (G6Pase) deficiency, translating in hypoglycemia during short fasts. Besides metabolic perturbations, GSDI patients develop long-term complications, especially chronic kidney disease (CKD). In GSDI patients, CKD is characterized by an accumulation of glycogen and lipids in kidneys, leading to a gradual decline in renal function. At a molecular level, the activation of the renin-angiotensin system is responsible for the development of renal fibrosis, eventually leading to renal failure. The same CKD phenotype was observed in a mouse model with a kidney-specific G6Pase deficiency (K.G6pc-/- mice). Furthermore, GSDI patients and mice develop frequently renal cysts at late stages of the nephropathy, classifying GSDI as a potential polycystic kidney disease (PKD). PKDs are genetic disorders characterized by multiple renal cyst formation, frequently caused by the loss of expression of polycystic kidney genes, such as PKD1/2 and PKHD1. Interestingly, these genes are deregulated in K.G6pc-/- kidneys, suggesting their possible role in GSDI cystogenesis. Finally, renal cysts are known to predispose to renal malignancy development. In addition, HNF1B loss is a malignancy prediction factor. Interestingly, Hnf1b expression was decreased in K.G6pc-/- kidneys. While a single case of renal cancer has been reported in a GSDI patient, a clear cell renal carcinoma was recently observed in one K.G6pc-/- mouse (out of 36 studied mice) at a later stage of the disease. This finding highlights the need to further analyze renal cyst development in GSDI patients in order to evaluate the possible associated risk of carcinogenesis, even if the risk might be limited.
- Published
- 2018
- Full Text
- View/download PDF
49. Dietary exacerbation of metabolic stress leads to accelerated hepatic carcinogenesis in glycogen storage disease type Ia.
- Author
-
Gjorgjieva M, Calderaro J, Monteillet L, Silva M, Raffin M, Brevet M, Romestaing C, Roussel D, Zucman-Rossi J, Mithieux G, and Rajas F
- Subjects
- Animals, Autophagy, Diet, High-Fat, Endoplasmic Reticulum Stress, Epithelial-Mesenchymal Transition, Glucose metabolism, Glucose-6-Phosphatase genetics, Glycogen Storage Disease Type I metabolism, Mice, Mice, Inbred C57BL, Sucrose administration & dosage, Carcinoma, Hepatocellular etiology, Glycogen Storage Disease Type I complications, Liver metabolism, Liver Neoplasms etiology
- Abstract
Background & Aims: Glycogen storage disease type Ia (GSDIa) is a rare genetic disease associated with glycogen accumulation in hepatocytes and steatosis. With age, most adult patients with GSDIa develop hepatocellular adenomas (HCA), which can progress to hepatocellular carcinomas (HCC). In this study, we characterized metabolic reprogramming and cellular defense alterations during tumorigenesis in the liver of hepatocyte-specific G6pc deficient (L.G6pc
-/- ) mice, which develop all the hepatic hallmarks of GSDIa., Methods: Liver metabolism and cellular defenses were assessed at pretumoral (four months) and tumoral (nine months) stages in L.G6pc-/- mice fed a high fat/high sucrose (HF/HS) diet., Results: In response to HF/HS diet, hepatocarcinogenesis was highly accelerated since 85% of L.G6pc-/- mice developed multiple hepatic tumors after nine months, with 70% classified as HCA and 30% as HCC. Tumor development was associated with high expression of malignancy markers of HCC, i.e. alpha-fetoprotein, glypican 3 and β-catenin. In addition, L.G6pc-/- livers exhibited loss of tumor suppressors. Interestingly, L.G6pc-/- steatosis exhibited a low-inflammatory state and was less pronounced than in wild-type livers. This was associated with an absence of epithelial-mesenchymal transition and fibrosis, while HCA/HCC showed a partial epithelial-mesenchymal transition in the absence of TGF-β1 increase. In HCA/HCC, glycolysis was characterized by a marked expression of PK-M2, decreased mitochondrial OXPHOS and a decrease of pyruvate entry in the mitochondria, confirming a "Warburg-like" phenotype. These metabolic alterations led to a decrease in antioxidant defenses and autophagy and chronic endoplasmic reticulum stress in L.G6pc-/- livers and tumors. Interestingly, autophagy was reactivated in HCA/HCC., Conclusion: The metabolic remodeling in L.G6pc-/- liver generates a preneoplastic status and leads to a loss of cellular defenses and tumor suppressors that facilitates tumor development in GSDI., Lay Summary: Glycogen storage disease type Ia (GSD1a) is a rare metabolic disease characterized by hypoglycemia, steatosis, excessive glycogen accumulation and tumor development in the liver. In this study, we have observed that GSDIa livers reprogram their metabolism in a similar way to cancer cells, which facilitates tumor formation and progression, in the absence of hepatic fibrosis. Moreover, hepatic burden due to overload of glycogen and lipids in the cells leads to a decrease in cellular defenses, such as autophagy, which could further promote tumorigenesis in the case of GSDI., (Copyright © 2018 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
50. The role of kidney in the inter-organ coordination of endogenous glucose production during fasting.
- Author
-
Kaneko K, Soty M, Zitoun C, Duchampt A, Silva M, Philippe E, Gautier-Stein A, Rajas F, and Mithieux G
- Subjects
- Animals, Blood Glucose metabolism, Fasting physiology, Glucose metabolism, Glucose-6-Phosphatase metabolism, Glycogen metabolism, Glycogen Storage Disease Type I, Hypoglycemia metabolism, Insulin metabolism, Kidney metabolism, Liver metabolism, Liver Glycogen metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Vitamin D metabolism, Gluconeogenesis physiology, Glucose biosynthesis, Kidney physiology
- Abstract
Objective: The respective contributions to endogenous glucose production (EGP) of the liver, kidney and intestine vary during fasting. We previously reported that the deficiency in either hepatic or intestinal gluconeogenesis modulates the repartition of EGP via glucagon secretion (humoral factor) and gut-brain-liver axis (neural factor), respectively. Considering renal gluconeogenesis reportedly accounted for approximately 50% of EGP during fasting, we examined whether a reduction in renal gluconeogenesis could promote alterations in the repartition of EGP in this situation., Methods: We studied mice whose glucose-6-phosphatase (G6Pase) catalytic subunit (G6PC) is specifically knocked down in the kidneys (K-G6pc
-/- mice) during fasting. We also examined the additional effects of intestinal G6pc deletion, renal denervation and vitamin D administration on the altered glucose metabolism in K-G6pc-/- mice., Results: Compared with WT mice, K-G6pc-/- mice exhibited (1) lower glycemia, (2) enhanced intestinal but not hepatic G6Pase activity, (3) enhanced hepatic glucokinase (GK encoded by Gck) activity, (4) increased hepatic glucose-6-phosphate and (5) hepatic glycogen spared from exhaustion during fasting. Increased hepatic Gck expression in the post-absorptive state could be dependent on the enhancement of insulin signal (AKT phosphorylation) in K-G6pc-/- mice. In contrast, the increase in hepatic GK activity was not observed in mice with both kidney- and intestine-knockout (KI-G6pc-/- mice). Hepatic Gck gene expression and hepatic AKT phosphorylation were reduced in KI-G6pc-/- mice. Renal denervation by capsaicin did not induce any effect on glucose metabolism in K-G6pc-/- mice. Plasma level of 1,25 (OH)2 D3 , an active form of vitamin D, was decreased in K-G6pc-/- mice. Interestingly, the administration of 1,25 (OH)2 D3 prevented the enhancement of intestinal gluconeogenesis and hepatic GK activity and blocked the accumulation of hepatic glycogen otherwise observed in K-G6pc-/- mice during fasting., Conclusions: A diminution in renal gluconeogenesis that is accompanied by a decrease in blood vitamin D promotes a novel repartition of EGP among glucose producing organs during fasting, featured by increased intestinal gluconeogenesis that leads to sparing glycogen stores in the liver. Our data suggest a possible involvement of a crosstalk between the kidneys and intestine (via the vitamin D system) and the intestine and liver (via a neural gut-brain axis), which might take place in the situations of deficient renal glucose production, such as chronic kidney disease., (Copyright © 2018 The Authors. Published by Elsevier GmbH.. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.