1
|
Mack KL, Landino NP, Tertyshnaia M, Longo TC, Vera SA, Crew LA, McDonald K, Phifer-Rixey M. Gene-by-environment Interactions and Adaptive Body Size Variation in Mice From the Americas. Mol Biol Evol 2025; 42:msaf078. [PMID: 40172935 PMCID: PMC12015161 DOI: 10.1093/molbev/msaf078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/14/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025] Open
Abstract
The relationship between genotype and phenotype is often mediated by the environment. Moreover, gene-by-environment (GxE) interactions can contribute to variation in phenotypes and fitness. In the last 500 yr, house mice have invaded the Americas. Despite their short residence time, there is evidence of rapid climate adaptation, including shifts in body size and aspects of metabolism with latitude. Previous selection scans have identified candidate genes for metabolic adaptation. However, environmental variation in diet as well as GxE interactions likely impact body mass variation in wild populations. Here, we investigated the role of the environment and GxE interactions in shaping adaptive phenotypic variation. Using new locally adapted inbred strains from North and South America, we evaluated response to a high-fat diet, finding that sex, strain, diet, and the interaction between strain and diet contributed significantly to variation in body size. We also found that the transcriptional response to diet is largely strain-specific, indicating that GxE interactions affecting gene expression are pervasive. Next, we used crosses between strains from contrasting climates to characterize gene expression regulatory divergence on a standard diet and on a high-fat diet. We found that gene regulatory divergence is often condition-specific, particularly for trans-acting changes. Finally, we found evidence for lineage-specific selection on cis-regulatory variation involved in diverse processes, including lipid metabolism. Overlap with scans for selection identified candidate genes for environmental adaptation with diet-specific effects. Together, our results underscore the importance of environmental variation and GxE interactions in shaping adaptive variation in complex traits.
Collapse
Affiliation(s)
- Katya L Mack
- Department of Ecology and Evolutionary Biology, University of Kansas, Lawrence, KS, USA
| | - Nico P Landino
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | | | - Tiffany C Longo
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Sebastian A Vera
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Lilia A Crew
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Kristi McDonald
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
| | - Megan Phifer-Rixey
- Department of Biology, Monmouth University, West Long Branch, NJ, USA
- Department of Biology, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Vily-Petit J, Gautier-Stein A, Mithieux G. [Intestinal gluconeogenesis : When the intestine produces glucose to prevent obesity and hepatic steatosis]. Med Sci (Paris) 2025; 41:246-252. [PMID: 40117549 DOI: 10.1051/medsci/2025023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025] Open
Abstract
Intestinal gluconeogenesis refers to the ability of the gut to produce glucose outside of meals. By initiating a gut-brain neural axis, its activation by dietary fiber or protein improves the regulation of energy balance. Recently, the creation of a genetic activation model of intestinal gluconeogenesis has demonstrated its anti-obesity, anti-diabetes and anti-hepatic steatosis effects. Interestingly, it increases thermogenesis in brown adipose tissue, thereby promoting energy expenditure and contributing to the fight against obesity. Therefore, targeting intestinal gluconeogenesis could be an innovative strategy to address metabolic diseases such as hepatic steatosis and diabetes, paving the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Justine Vily-Petit
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| | - Amandine Gautier-Stein
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| | - Gilles Mithieux
- Inserm U1213 Nutrition, Diabète et Cerveau, université Claude Bernard Lyon 1, Villeurbanne, France
| |
Collapse
|
3
|
Estrada-Meza J, Videlo J, Bron C, Duchampt A, Saint-Béat C, Zergane M, Silva M, Rajas F, Bouret SG, Mithieux G, Gautier-Stein A. Intestinal gluconeogenesis controls the neonatal development of hypothalamic feeding circuits. Mol Metab 2024; 89:102036. [PMID: 39304064 PMCID: PMC11470480 DOI: 10.1016/j.molmet.2024.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
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.
Collapse
Affiliation(s)
| | - Jasmine Videlo
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Clara Bron
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Adeline Duchampt
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Mickael Zergane
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Marine Silva
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Fabienne Rajas
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Sebastien G Bouret
- University Lille, Inserm, CHU Lille, Laboratory of development and plasticity of the Neuroendocrine brain, Lille Neuroscience & Cognition, Inserm UMR-S1172, Lille, France
| | - Gilles Mithieux
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | | |
Collapse
|
4
|
Vily-Petit J, Taki A, Sinet F, Soty M, Guiard B, Zemdegs J, Malleret G, Stefanutti A, Mithieux G, Gautier-Stein A. Absence of the Peptide Transporter 1 Induces a Prediabetic and Depressive-Like Phenotype in Mice. Neuroendocrinology 2024; 115:226-241. [PMID: 38852578 DOI: 10.1159/000539499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024]
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.
Collapse
Affiliation(s)
| | - Amelie Taki
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Flore Sinet
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Maud Soty
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Bruno Guiard
- CRCA - UMR - Université Paul Sabatier, Toulouse, France
| | | | - Gael Malleret
- Forgetting and Cortical Dynamics, Lyon Neuroscience Research Center, Lyon, France
| | - Anne Stefanutti
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France
| | - Gilles Mithieux
- INSERM UMR-S1213, Université Claude Bernard Lyon 1, Lyon, France,
| | | |
Collapse
|
5
|
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, Oosterveer MH. Normalization of hepatic ChREBP activity does not protect against liver disease progression in a mouse model for Glycogen Storage Disease type Ia. Cancer Metab 2023; 11:5. [PMID: 37085901 PMCID: PMC10122297 DOI: 10.1186/s40170-023-00305-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/21/2023] [Indexed: 04/23/2023] Open
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.
Collapse
Affiliation(s)
- Martijn G. S. Rutten
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yu Lei
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Joanne H. Hoogerland
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent W. Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hong Yang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Trijnie Bos
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Kishore A. Krishnamurthy
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Aycha Bleeker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mirjam H. Koster
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rachel E. Thomas
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Justina C. Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hilda van den Bos
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gilles Mithieux
- Institut National de La Santé Et de La Recherche Médicale, U1213 Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| | - Fabienne Rajas
- Institut National de La Santé Et de La Recherche Médicale, U1213 Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Diana C. J. Spierings
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alain de Bruin
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maaike H. Oosterveer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Régnier M, Carbinatti T, Parlati L, Benhamed F, Postic C. The role of ChREBP in carbohydrate sensing and NAFLD development. Nat Rev Endocrinol 2023; 19:336-349. [PMID: 37055547 DOI: 10.1038/s41574-023-00809-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 04/15/2023]
Abstract
Excessive sugar consumption and defective glucose sensing by hepatocytes contribute to the development of metabolic diseases including type 2 diabetes mellitus (T2DM) and nonalcoholic fatty liver disease (NAFLD). Hepatic metabolism of carbohydrates into lipids is largely dependent on the carbohydrate-responsive element binding protein (ChREBP), a transcription factor that senses intracellular carbohydrates and activates many different target genes, through the activation of de novo lipogenesis (DNL). This process is crucial for the storage of energy as triglycerides in hepatocytes. Furthermore, ChREBP and its downstream targets represent promising targets for the development of therapies for the treatment of NAFLD and T2DM. Although lipogenic inhibitors (for example, inhibitors of fatty acid synthase, acetyl-CoA carboxylase or ATP citrate lyase) are currently under investigation, targeting lipogenesis remains a topic of discussion for NAFLD treatment. In this Review, we discuss mechanisms that regulate ChREBP activity in a tissue-specific manner and their respective roles in controlling DNL and beyond. We also provide in-depth discussion of the roles of ChREBP in the onset and progression of NAFLD and consider emerging targets for NAFLD therapeutics.
Collapse
Affiliation(s)
- Marion Régnier
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France.
| | - Thaïs Carbinatti
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
| | - Lucia Parlati
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
| | - Fadila Benhamed
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France
| | - Catherine Postic
- Université Paris Cité, Institut Cochin, CNRS, INSERM, Paris, France.
| |
Collapse
|
7
|
Gautier-Stein A, Chilloux J, Soty M, Thorens B, Place C, Zitoun C, Duchampt A, Da Costa L, Rajas F, Lamaze C, Mithieux G. A caveolin-1 dependent glucose-6-phosphatase trafficking contributes to hepatic glucose production. Mol Metab 2023; 70:101700. [PMID: 36870604 PMCID: PMC10023957 DOI: 10.1016/j.molmet.2023.101700] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/15/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
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.
Collapse
Affiliation(s)
- Amandine Gautier-Stein
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France.
| | - Julien Chilloux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France
| | - Maud Soty
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015, Lausanne, Switzerland
| | - Christophe Place
- Laboratoire de Physique (UMR CNRS 5672), ENS de Lyon, Université de Lyon, F-69364, Lyon cedex 07, France
| | - Carine Zitoun
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France
| | - Adeline Duchampt
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France
| | - Lorine Da Costa
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France
| | - Christophe Lamaze
- Institut Curie, PSL Research University, INSERM U1143, CNRS UMR 3666, Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, 75005, Paris, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, F-69374, Lyon, France
| |
Collapse
|
8
|
Jamshed F, Dashti F, Ouyang X, Mehal WZ, Banini BA. New uses for an old remedy: Digoxin as a potential treatment for steatohepatitis and other disorders. World J Gastroenterol 2023; 29:1824-1837. [PMID: 37032732 PMCID: PMC10080697 DOI: 10.3748/wjg.v29.i12.1824] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/12/2023] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
Repurposing of the widely available and relatively cheap generic cardiac gly-coside digoxin for non-cardiac indications could have a wide-ranging impact on the global burden of several diseases. Over the past several years, there have been significant advances in the study of digoxin pharmacology and its potential non-cardiac clinical applications, including anti-inflammatory, antineoplastic, metabolic, and antimicrobial use. Digoxin holds promise in the treatment of gastrointestinal disease, including nonalcoholic steatohepatitis and alcohol-associated steatohepatitis as well as in obesity, cancer, and treatment of viral infections, among other conditions. In this review, we provide a summary of the clinical uses of digoxin to date and discuss recent research on its emerging applications.
Collapse
Affiliation(s)
- Fatima Jamshed
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
- Griffin Hospital-Yale University, Derby, CT 06418, United States
| | - Farzaneh Dashti
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| | - Xinshou Ouyang
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
- West Haven Veterans Medical Center, West Haven, CT 06516, United States
| | - Bubu A Banini
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06510, United States
| |
Collapse
|
9
|
Petrova IO, Smirnikhina SA. Studies on glycogen storage disease type 1a animal models: a brief perspective. Transgenic Res 2022; 31:593-606. [PMID: 36006546 DOI: 10.1007/s11248-022-00325-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/09/2022] [Indexed: 01/20/2023]
Abstract
Glycogen storage disease type 1 (GSD1) is a rare hereditary monogenic disease characterized by the disturbed glucose metabolism. The most widespread variant of GSD1 is GSD1a, which is a deficiency of glucose-6-phosphatase-ɑ. Glucose-6-phosphatase-ɑ is expressed only in liver, kidney, and intestine, and these organs are primarily affected by its deficiency, and long-term complications of GSD1a include hepatic tumors and chronic liver disease. This article is a brief overview of existing animal models for GSD1a, from the first mouse model of 1996 to modern CRISPR/Cas9-generated ones. First whole-body murine models demonstrated exact metabolic symptoms of GSD1a, but the animals did not survive weaning. The protocol for glucose treatment allowed prolonged survival of affected animals, but long-term complications, such as hepatic tumorigenesis, could not be investigated. Next, organ-specific knockout models were developed, and most of the metabolic research was performed on liver glucose-6-phosphate-deficient mice. Naturally occuring mutation was also discovered in dogs. All these models are widely used to study GSD1a from metabolic and physiological standpoints and to develop possible treatments involving gene therapy. Research performed using these models helped elucidate the role of glycogen and lipid accumulation, hypoxia, mitochondrial dysfunction, and autophagy impairment in long-term complications of GSD1a, including hepatic tumorigenesis. Recently, gene replacement therapy and genome editing were tested on described models, and some of the developed approaches have reached clinical trials.
Collapse
Affiliation(s)
- Irina O Petrova
- Laboratory of Genome Editing, Research Center for Medical Genetics, Moskvorechye 1, Moscow, Russia, 115478.
| | - Svetlana A Smirnikhina
- Laboratory of Genome Editing, Research Center for Medical Genetics, Moskvorechye 1, Moscow, Russia, 115478
| |
Collapse
|
10
|
Gut microbiome and type 2 diabetes. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 191:175-185. [DOI: 10.1016/bs.pmbts.2022.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
11
|
Kang M, Han SK, Kim S, Park S, Jo Y, Kang H, Ko J. Role of small leucine zipper protein in hepatic gluconeogenesis and metabolic disorder. J Mol Cell Biol 2021; 13:361-373. [PMID: 33355643 PMCID: PMC8373270 DOI: 10.1093/jmcb/mjaa069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/03/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Hepatic gluconeogenesis is the central pathway for glucose generation in the body. The imbalance between glucose synthesis and uptake leads to metabolic diseases such as obesity, diabetes, and cardiovascular diseases. Small leucine zipper protein (sLZIP) is an isoform of LZIP and it mainly functions as a transcription factor. Although sLZIP is known to regulate the transcription of genes involved in various cellular processes, the role of sLZIP in hepatic glucose metabolism is not known. In this study, we investigated the regulatory role of sLZIP in hepatic gluconeogenesis and its involvement in metabolic disorder. We found that sLZIP expression was elevated during glucose starvation, leading to the promotion of phosphoenolpyruvate carboxylase and glucose-6-phosphatase expression in hepatocytes. However, sLZIP knockdown suppressed the expression of the gluconeogenic enzymes under low glucose conditions. sLZIP also enhanced glucose production in the human liver cells and mouse primary hepatic cells. Fasting-induced cyclic adenosine monophosphate impeded sLZIP degradation. Results of glucose and pyruvate tolerance tests showed that sLZIP transgenic mice exhibited abnormal blood glucose metabolism. These findings suggest that sLZIP is a novel regulator of gluconeogenic enzyme expression and plays a role in blood glucose homeostasis during starvation.
Collapse
Affiliation(s)
- Minsoo Kang
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Sun Kyoung Han
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Suhyun Kim
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Sungyeon Park
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Yerin Jo
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Hyeryung Kang
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Jesang Ko
- Division of Life Sciences, Korea University, Seoul 02841, South Korea
| |
Collapse
|
12
|
Estrada-Meza J, Videlo J, Bron C, Saint-Béat C, Silva M, Duboeuf F, Peyruchaud O, Rajas F, Mithieux G, Gautier-Stein A. Tamoxifen Treatment in the Neonatal Period Affects Glucose Homeostasis in Adult Mice in a Sex-Dependent Manner. Endocrinology 2021; 162:6277101. [PMID: 33999998 DOI: 10.1210/endocr/bqab098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Indexed: 12/11/2022]
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.
Collapse
Affiliation(s)
- Judith Estrada-Meza
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Jasmine Videlo
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Clara Bron
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Cécile Saint-Béat
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - François Duboeuf
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Olivier Peyruchaud
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1033, Lyon, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-S1213, Lyon, France
| | | |
Collapse
|
13
|
Cai Z, Yang Y, Zhang J. Hepatokine levels during the first or early second trimester of pregnancy and the subsequent risk of gestational diabetes mellitus: a systematic review and meta-analysis. Biomarkers 2021; 26:517-531. [PMID: 34082623 DOI: 10.1080/1354750x.2021.1928754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE The relationship between hepatokine levels during the first or early second trimester of pregnancy and the subsequent risk of gestational diabetes mellitus (GDM) have been studied extensively. However, conclusions remain debateable whether hepatokines are potential markers of GDM. We conducted a meta-analysis of published articles to understand the association between circulating levels of selected hepatokines (including FGF21, fetuin-A, afamin, adropin, ficolin-3, selenoprotein P, ANGPTL4 and AGF) and the risk of GDM. MATERIALS AND METHODS We searched the PubMed, Embase, Cochrane Library and Web of Science databases for studies published before January 2021 that examined the association between hepatokines and GDM (Prospero Registration# CRD42020191408). The quality was assessed by the Newcastle-Ottawa Scale (NOS). Pooled standard mean differences (SMDs) and weighted mean differences (WMDs) with 95% confidence intervals (CIs) were used to compare the levels of hepatokines in different groups using fixed effects or random effects models. Meta-regression analysis and publication bias were conducted in accordance with standard methods. The trim-fill adjustment method was used to further assess the possible effect of publication bias. Sensitivity analysis was performed by omitting each study one at a time. RESULTS The meta-analysis included 31 observational studies relating hepatokine levels to GDM in 4729 participants (1908 GDM, 2821 non-GDM). Serum FGF21 levels in patients with GDM were higher than those in healthy pregnant women during the second trimester and after delivery (SMD 0.89, [95% CI] 0.01-1.78 for the second trimester; SMD 1.42, [95% CI] 0.86-1.98 for after delivery). The serum levels of afamin in patients with GDM were significantly higher than those in healthy pregnant women during the first trimester and before pregnancy (SMD 0.51, [95% CI] 0.15-0.86 for first trimester; SMD 0.97, [95% CI] 0.45-1.50 for before pregnancy). Serum adropin levels in patients with GDM were higher than those in healthy pregnant women during the first and third trimesters of pregnancy (SMD 4.26, [95% CI] 3.30-5.23 for the first trimester; SMD 4.02, [95% CI] 3.09-4.94 for the third trimester). The serum levels of ficolin-3 in GDM patients were higher than those in healthy pregnant women during the second and third trimesters of pregnancy (WMD 1.43, [95% CI] 0.91-1.96 for the second trimester; SMD 1.28, [95% CI] 0.72-1.84 for the third trimester). The serum AGF level of patients with GDM was higher than that of healthy pregnant women in the control group in the third trimester (WMD 61 [95% CI] 37.04-81.96). The serum levels of selenoprotein P in patients with GDM were higher than those in healthy pregnant women in the control group during the first trimester (WMD 7.09 [95% CI] 4.6-9.57). CONCLUSIONS Measurement of circulating hepatokines in the first or second trimester of pregnancy may improve the identification of women at risk of developing GDM later. Prospective evaluation of the combination of hepatokines and maternal characteristics for early identification of those who do and do not require OGTT is warranted. Additional well-designed prospective studies with longitudinal assessment of hepatokines during pregnancy are needed to understand the trajectories and dynamic associations of hepatokines with GDM risk.
Collapse
Affiliation(s)
- Zixin Cai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yan Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
14
|
Fan N, Ding X, Zhen Q, Gu L, Zhang A, Shen T, Wang Y, Peng Y. Association of the Non-Alcoholic Fatty Liver Disease Fibrosis Score with subclinical myocardial remodeling in patients with type 2 diabetes: A cross-sectional study in China. J Diabetes Investig 2021; 12:1035-1041. [PMID: 33030804 PMCID: PMC8169360 DOI: 10.1111/jdi.13430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/11/2020] [Accepted: 10/04/2020] [Indexed: 12/15/2022] Open
Abstract
AIMS/INTRODUCTION Non-alcoholic fatty liver disease, especially with liver fibrosis, is associated with cardiovascular diseases. The Non-Alcoholic Fatty Liver Disease Fibrosis Score (NFS), a non-invasive marker of advanced fibrosis, was found to be associated with cardiovascular diseases in different populations. The aim of the present study was to determine whether the NFS is associated with subclinical myocardial remodeling in type 2 diabetes patients. MATERIALS AND METHODS A cross-sectional study was carried out in type 2 diabetes patients. The NFS derived from available parameters was calculated, and the participants were divided according to the quartiles of the NFS and grades of the NFS (low, intermediate and high). Fibrosis-4 and Aspartate Aminotransferase to Platelet Ratio Index, another two liver fibrosis scores, were also calculated. Subclinical myocardial remodeling was examined by echocardiography, and its associations with NFS, Fibrosis-4 and Aspartate Aminotransferase to Platelet Ratio Index were analyzed. RESULTS A total of 1,878 type 2 diabetes patients were enrolled in the present study. The NFS was independently associated with sex, age, body mass index, low-density lipoprotein cholesterol and glycated hemoglobin in type 2 diabetes patients. Parameters of subclinical myocardial remodeling including left atrial dimension, interventricular septum thickness, left ventricular end-diastolic diameter, left ventricular end-systolic diameter, left ventricular posterior wall thickness (LVPWT) and left ventricular mass index were all gradually increased with the increment of the NFS. Linear regression analysis further showed that the NFS was positively associated with left atrial dimension, interventricular septum thickness, left ventricular end-diastolic diameter, left ventricular end-systolic diameter, LVPWT and left ventricular mass index after adjustment for the confounding factors. Similarly, Fibrosis-4 was associated with left atrial dimension, interventricular septum thickness, LVPWT and left ventricular mass index. In contrast, the Aspartate Aminotransferase to Platelet Ratio Index was only associated with LVPWT. CONCLUSIONS Non-invasive liver fibrosis scores, especially the NFS, are independently associated with subclinical myocardial remodeling in type 2 diabetes patients.
Collapse
Affiliation(s)
- Nengguang Fan
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoying Ding
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Qin Zhen
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Liping Gu
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Aifang Zhang
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Tingting Shen
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Yufan Wang
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Yongde Peng
- Department of Endocrinology and MetabolismShanghai General HospitalShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
15
|
Kowalczuk A, Bourebaba N, Kornicka-Garbowska K, Turlej E, Marycz K, Bourebaba L. Hyoscyamus albus nortropane alkaloids reduce hyperglycemia and hyperinsulinemia induced in HepG2 cells through the regulation of SIRT1/NF-kB/JNK pathway. Cell Commun Signal 2021; 19:61. [PMID: 34034759 PMCID: PMC8152357 DOI: 10.1186/s12964-021-00735-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/24/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Chronic superphysiological glucose and insulin concentrations are known to trigger several tissue and organ failures, including insulin resistance, oxidative stress and chronic low-grade inflammation. Hence, the screening for molecules that may counteract such conditions is essential in current existing therapeutic strategies, thereby the use of medicinal plant derivatives represents a promising axis in this regard. METHODS In this study, the effect of a selected traditional medicinal plant, Hyoscyamus albus from which, calystegines have been isolated, was investigated in an experimental model of hyperinsulinemia and hyperglycemia induced on HepG2 cells. The mRNA and protein expression levels of different insulin signaling, gluconeogenic and inflammatory pathway- related molecules were examined. Additionally, cell viability and apoptosis, oxidative stress extent and mitochondrial dysfunctions were assayed using flow cytometric and qRT-PCR techniques. RESULTS Treatment of IR HepG2 cells with calystegines strongly protected the injured cells from apoptosis, oxidative stress and mitochondrial integrity loss. Interestingly, nortropane alkaloids efficiently regulated the impaired glucose metabolism in IR HepG2 cells, through the stimulation of glucose uptake and the modulation of SIRT1/Foxo1/G6PC/mTOR pathway, which is governing the hepatic gluconeogenesis. Furthermore, the alkaloidal extract restored the defective insulin signaling pathway, mainly by promoting the expression of Insr at the mRNA and protein levels. What is more, treated cells exhibited significant mitigated inflammatory response, as evidenced by the modulation and the regulation of the NF- κB/JNK/TLR4 axis and the downstream proinflammatory cytokines recruitment. CONCLUSION Overall, the present investigation demonstrates that calystegines from Hyoscyamus albus provide cytoprotection to the HepG2 cells against insulin/glucose induced insulin resistance and apoptosis due to the regulation of SIRT1/Foxo1/G6PC/mTOR and NF-κB/JNK/TLR4 signaling pathways. Video Abstract.
Collapse
Affiliation(s)
- Anna Kowalczuk
- grid.419694.70000 0004 0622 0266National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
| | - Nabila Bourebaba
- International Institute of Translational Medicine, Jesionowa 11, 55-114 Malin, Wisznia Mała, Poland ,grid.411200.60000 0001 0694 6014Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland
| | | | - Eliza Turlej
- grid.411200.60000 0001 0694 6014Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland
| | - Krzysztof Marycz
- International Institute of Translational Medicine, Jesionowa 11, 55-114 Malin, Wisznia Mała, Poland ,Collegium Medicum, Institute of Medical Science, Cardinal Stefan Wyszyński University (UKSW), Dewajtis 5, 01-815 Warsaw, Poland
| | - Lynda Bourebaba
- International Institute of Translational Medicine, Jesionowa 11, 55-114 Malin, Wisznia Mała, Poland ,grid.411200.60000 0001 0694 6014Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375 Wrocław, Poland
| |
Collapse
|
16
|
Angiopoietin-Like Growth Factor Involved in Leptin Signaling in the Hypothalamus. Int J Mol Sci 2021; 22:ijms22073443. [PMID: 33810547 PMCID: PMC8037945 DOI: 10.3390/ijms22073443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 11/26/2022] Open
Abstract
The hypothalamic regulation of appetite governs whole-body energy balance. Satiety is regulated by endocrine factors including leptin, and impaired leptin signaling is associated with obesity. Despite the anorectic effect of leptin through the regulation of the hypothalamic feeding circuit, a distinct downstream mediator of leptin signaling in neuron remains unclear. Angiopoietin-like growth factor (AGF) is a peripheral activator of energy expenditure and antagonizes obesity. However, the regulation of AGF expression in brain and localization to mediate anorectic signaling is unknown. Here, we demonstrated that AGF is expressed in proopiomelanocortin (POMC)-expressing neurons located in the arcuate nucleus (ARC) of the hypothalamus. Unlike other brain regions, hypothalamic AGF expression is stimulated by leptin-induced signal transducers and activators of transcription 3 (STAT3) phosphorylation. In addition, leptin treatment to hypothalamic N1 cells significantly enhanced the promoter activity of AGF. This induction was abolished by the pretreatment of ruxolitinib, a leptin signaling inhibitor. These results indicate that hypothalamic AGF expression is induced by leptin and colocalized to POMC neurons.
Collapse
|
17
|
Mishra SP, Jain S, Taraphder S, Yadav H. New Horizons in Microbiota and Metabolic Health Research. J Clin Endocrinol Metab 2021; 106:e1052-e1059. [PMID: 33128374 PMCID: PMC7823252 DOI: 10.1210/clinem/dgaa769] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Indexed: 01/02/2023]
Abstract
Decade-old studies have demonstrated that microbes living in our gut (microbiota) contribute to both maintaining normal metabolic function and to the pathology of metabolic diseases, such as obesity and diabetes. Emerging evidence suggests that gut microbiota influences the personalized effects of diets and drugs and impact the gut-brain axis and leaky gut inflammation to control metabolic function/diseases. Gut microbiota can be an ideal source of prognostic markers and therapies for metabolic diseases. Here we discuss the emerging concepts in the area of microbiota and metabolic interactions in personalized nutrition, drug response, and disease prognosis.
Collapse
Affiliation(s)
- Sidharth P Mishra
- Department of Internal Medicine–Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Animal Genetics and Breeding, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Shalini Jain
- Metabolic Phenotyping Shared Resource, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Internal Medicine-Endocrinology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Subhash Taraphder
- Department of Animal Genetics and Breeding, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Hariom Yadav
- Department of Internal Medicine–Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
18
|
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.
Collapse
|
19
|
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, Oosterveer MH. Hepatic Carbohydrate Response Element Binding Protein Activation Limits Nonalcoholic Fatty Liver Disease Development in a Mouse Model for Glycogen Storage Disease Type 1a. Hepatology 2020; 72:1638-1653. [PMID: 32083759 PMCID: PMC7702155 DOI: 10.1002/hep.31198] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 01/05/2023]
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.
Collapse
Affiliation(s)
- Yu Lei
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Joanne A. Hoogerland
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Vincent W. Bloks
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Trijnie Bos
- Department of Laboratory MedicineUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Aycha Bleeker
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Henk Wolters
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Justina C. Wolters
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Brenda S. Hijmans
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Theo H. van Dijk
- Department of Laboratory MedicineUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Rachel Thomas
- Dutch Molecular Pathology CenterFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic DiseasesAmsterdam Gastroenterology and MetabolismAmsterdam Cardiovascular SciencesAmsterdamthe Netherlands
- Core Facility of MetabolomicsAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamthe Netherlands
| | - Gilles Mithieux
- National Institute of Health and Medical Research, U1213LyonFrance
- University of LyonLyonFrance
- University of Lyon 1VilleurbanneFrance
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic DiseasesAmsterdam Gastroenterology and MetabolismAmsterdam Cardiovascular SciencesAmsterdamthe Netherlands
| | - Alain de Bruin
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Dutch Molecular Pathology CenterFaculty of Veterinary MedicineUtrecht UniversityUtrechtthe Netherlands
| | - Fabienne Rajas
- National Institute of Health and Medical Research, U1213LyonFrance
- University of LyonLyonFrance
- University of Lyon 1VilleurbanneFrance
| | - Folkert Kuipers
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Department of Laboratory MedicineUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Maaike H. Oosterveer
- Department of PediatricsUniversity Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| |
Collapse
|
20
|
Rajas F, Dentin R, Cannella Miliano A, Silva M, Raffin M, Levavasseur F, Gautier-Stein A, Postic C, Mithieux G. The absence of hepatic glucose-6 phosphatase/ChREBP couple is incompatible with survival in mice. Mol Metab 2020; 43:101108. [PMID: 33137488 PMCID: PMC7691719 DOI: 10.1016/j.molmet.2020.101108] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
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. Hepatic deletion of both ChREBP and glucose-6 phosphatase collapses liver lipids. Double deletion leads to excessive glycogen storage and a liver swollen with water. Hepatic deletion of both ChREBP and glucose-6 phosphatase leads to death. Glucose-6 phosphate homeostasis in hepatocytes is a vital function.
Collapse
Affiliation(s)
- Fabienne Rajas
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-1213, Lyon, France.
| | - Renaud Dentin
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | | | - Marine Silva
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-1213, Lyon, France
| | - Margaux Raffin
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-1213, Lyon, France
| | | | | | - Catherine Postic
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, Université de Lyon, INSERM UMR-1213, Lyon, France
| |
Collapse
|
21
|
Mokou M, Yang S, Zhan B, Geng S, Li K, Yang M, Yang G, Deng W, Liu H, Liu D, Zhu Z, Li L. Elevated Circulating Fetuin-B Levels Are Associated with Insulin Resistance and Reduced by GLP-1RA in Newly Diagnosed PCOS Women. Mediators Inflamm 2020; 2020:2483435. [PMID: 33061822 PMCID: PMC7545451 DOI: 10.1155/2020/2483435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Previous studies have suggested that Fetuin-B seems to be a secreted adipokine related to metabolic diseases. However, the results have been inconsistent. Here, our objective is to investigate the changes in circulating Fetuin-B levels in women with polycystic ovary syndrome (PCOS) and analyze the association of Fetuin-B and insulin resistance (IR). METHODS The current study is comprised of a cross-sectional study and a series of interventional studies. Oral glucose tolerance test (OGTT) and euglycemic-hyperinsulinemic clamp (EHC) were engaged to assess glucose tolerance and insulin sensitivity. Serum Fetuin-B levels were determined by ELISA. RESULTS Serum Fetuin-B and TNF-α levels were markedly increased in women with PCOS compared to healthy women. Circulating Fetuin-B was positively associated with body mass index, waist-to-hip ratio, the percentage of body fat (FAT%), systolic blood pressure, triglyceride, low-density lipoprotein cholesterol, fasting blood glucose, 2 h blood glucose after glucose overload, fasting insulin, 2 h insulin after glucose overload, HOMA-insulin resistance index (HOMA-IR), the area under the curve for insulin (AUCi), AUCg, and TNF-α, while negatively associated with M value and follicular stimulating hormone (FSH). During the EHC, Fetuin-B levels were found to be significantly increased in PCOS women. After a glucose challenge, serum Fetuin-B levels in healthy women were significantly increased. Lipid infusion reduced serum Fetuin-B levels in 30 healthy subjects. After six months of glucagon-like peptide-1 receptor agonist (GLP-1RA) intervention, serum Fetuin-B concentrations in PCOS women markedly decreased following ameliorated IR. CONCLUSION Our results indicate that Fetuin-B may be a biomarker of IR in individuals with PCOS. This trial is registered with ChiCTR-IIR-16007901.
Collapse
Affiliation(s)
- Mani Mokou
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, 400016 Chongqing, China
| | - Shan Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Bin Zhan
- The Thirteenth People's Hospital of Chongqing, Chongqing 400016, China
| | - Shan Geng
- The Thirteenth People's Hospital of Chongqing, Chongqing 400016, China
| | - Kejia Li
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Mengliu Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Gangyi Yang
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Wuquan Deng
- Department of Endocrinology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing 400014, China
| | - Hua Liu
- Department of Pediatrics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216-4505, USA
| | - Dongfang Liu
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing 400010, China
| | - Ling Li
- Key Laboratory of Diagnostic Medicine (Ministry of Education) and Department of Clinical Biochemistry, College of Laboratory Medicine, Chongqing Medical University, 400016 Chongqing, China
| |
Collapse
|
22
|
Fan KC, Wu HT, Wei JN, Chuang LM, Hsu CY, Yen IW, Lin CH, Lin MS, Shih SR, Wang SH, Chang TJ, Li HY. Serum Angiopoietin-like Protein 6, Risk of Type 2 Diabetes, and Response to Hyperglycemia: A Prospective Cohort Study. J Clin Endocrinol Metab 2020; 105:5775438. [PMID: 32123920 DOI: 10.1210/clinem/dgaa103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 02/27/2020] [Indexed: 02/06/2023]
Abstract
CONTEXT Angiopoietin-like protein 6 (ANGPTL6) is a hepatokine that improves insulin sensitivity in animals. However, serum ANGPTL6 concentration was found to be higher in human participants with diabetes or metabolic syndrome in cross-sectional studies, implying that ANGPTL6 may be induced to counteract hyperglycemia. OBJECTIVE To investigate whether serum ANGPTL6 can predict incident diabetes and explore whether glucose or insulin can regulate ANGPTL6 expression and secretion. DESIGN This cohort study included adults without diabetes at baseline who were followed every 2 years for incident diabetes. Serum ANGPTL6 concentrations were measured at baseline and during oral glucose tolerance tests (OGTTs). A hepatic cell line, HepG2, and diet-induced obesity mouse model were used to evaluate the response of ANGPTL6 expression and secretion to hyperglycemia and the metabolic syndrome. RESULTS We recruited 1103 participants without diabetes at baseline. During the 4.22-year follow-up, 113 (10.2%) participants developed incident diabetes. Serum ANGPTL6 was negatively associated with the incidence of diabetes (adjusted hazard ratio, 0.77; P = 0.042). However, serum ANGPTL6 level was higher in participants with prediabetes (P = 0.018) and was elevated during OGTT. In HepG2 cells, treatment with glucose, but not insulin, induced ANGPTL6 expression. Hepatic ANGPTL6 expression and serum ANGPTL6 concentrations were significantly higher in mice fed with a high-fat diet than in those fed with a standard chow (both P < 0.05). CONCLUSION A high serum ANGPTL6 level is associated with a low incidence of diabetes in humans. ANGPTL6 is expressed and secreted in response to hyperglycemia to maintain glucose homeostasis.
Collapse
Affiliation(s)
- Kang-Chih Fan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hung-Tsung Wu
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Jung-Nan Wei
- Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Yao Hsu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - I-Weng Yen
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Chia-Hung Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Mao-Shin Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shyang-Rong Shih
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shu-Huei Wang
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tien-Jyun Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Yuan Li
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
23
|
Qaddoumi MG, Alanbaei M, Hammad MM, Al Khairi I, Cherian P, Channanath A, Thanaraj TA, Al-Mulla F, Abu-Farha M, Abubaker J. Investigating the Role of Myeloperoxidase and Angiopoietin-like Protein 6 in Obesity and Diabetes. Sci Rep 2020; 10:6170. [PMID: 32277104 PMCID: PMC7148302 DOI: 10.1038/s41598-020-63149-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/02/2020] [Indexed: 02/07/2023] Open
Abstract
Myeloperoxidase (MPO) is positively associated with obesity and diet-induced insulin resistance. Angiopoietin-like protein 6 (ANGPTL6) regulates metabolic processes and counteract obesity through increased energy expenditure. This study aims to evaluate the plasma MPO and ANGPTL6 levels in obese and diabetic individuals as well as MPO association with biochemical markers of obesity. A total of 238 participants were enrolled, including 137 control and 101 type 2 diabetes (T2D) patients. ANGPTL6 and MPO levels and other biomarkers were measured via ELISA. ANGPTL6 levels were significantly higher in the diabetic population and obese individuals. When the group was stratified based on T2D, ANGPTL6 levels were significantly higher in obese-diabetic participants compared with non-obese-diabetics, but obese-non-diabetic individuals had similar ANGPTL6 levels to their controls. MPO levels were higher in obese compared with non-obese participants but did not differ between T2D and control participants. MPO levels were upregulated in obese compared with non-obese in both diabetics and non-diabetics. MPO was positively associated with ANGPTL6, triglyceride, BMI, TNF-alpha, high-sensitivity C-reactive protein, interleukin-6, and plasminogen activator inhibitor-1. Taken together, our findings suggest that both MPO and ANGPTL6 may regulate obesity, although MPO exerts this effect independent of diabetes while ANGPTL6 may have a modulatory role in diabetes.
Collapse
Affiliation(s)
- Mohammad G Qaddoumi
- Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
- Pharmacology and Therapeutics Department, Faculty of Pharmacy, Kuwait University, Kuwait City, Kuwait
| | - Muath Alanbaei
- Department of Medicine, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Maha M Hammad
- Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Irina Al Khairi
- Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Preethi Cherian
- Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Arshad Channanath
- Functional Genomic Unit, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | | | - Fahd Al-Mulla
- Functional Genomic Unit, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | - Mohamed Abu-Farha
- Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait.
| | - Jehad Abubaker
- Biochemistry and Molecular Biology, Dasman Diabetes Institute, Kuwait City, Kuwait.
| |
Collapse
|
24
|
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, Oosterveer MH. Glucose-6-Phosphate Regulates Hepatic Bile Acid Synthesis in Mice. Hepatology 2019; 70:2171-2184. [PMID: 31102537 PMCID: PMC6859192 DOI: 10.1002/hep.30778] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 05/15/2019] [Indexed: 12/22/2022]
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.
Collapse
Affiliation(s)
- Joanne A. Hoogerland
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Yu Lei
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Justina C. Wolters
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Jan Freark de Boer
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
- Laboratory MedicineUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Trijnie Bos
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Aycha Bleeker
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Niels L. Mulder
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Theo H. van Dijk
- Laboratory MedicineUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Jan A. Kuivenhoven
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213Université Claude Bernard LyonVilleurbanneFrance
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213Université Claude Bernard LyonVilleurbanneFrance
| | - Rebecca A. Haeusler
- Department of Pathology and Cell BiologyColumbia University College of Physicians and SurgeonsNew YorkNY
| | - Henkjan J. Verkade
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Vincent W. Bloks
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| | - Folkert Kuipers
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
- Laboratory MedicineUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Maaike H. Oosterveer
- Department of PediatricsUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
25
|
Rajas F, Gautier-Stein A, Mithieux G. Glucose-6 Phosphate, A Central Hub for Liver Carbohydrate Metabolism. Metabolites 2019; 9:metabo9120282. [PMID: 31756997 PMCID: PMC6950410 DOI: 10.3390/metabo9120282] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 12/23/2022] Open
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.
Collapse
Affiliation(s)
- Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, F-69008 Lyon, France; (A.G.-S.); (G.M.)
- Université de Lyon, F-69008 Lyon, France
- Université Lyon 1, F-69622 Villeurbanne, France
- Correspondence:
| | - Amandine Gautier-Stein
- Institut National de la Santé et de la Recherche Médicale, U1213, F-69008 Lyon, France; (A.G.-S.); (G.M.)
- Université de Lyon, F-69008 Lyon, France
- Université Lyon 1, F-69622 Villeurbanne, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, F-69008 Lyon, France; (A.G.-S.); (G.M.)
- Université de Lyon, F-69008 Lyon, France
- Université Lyon 1, F-69622 Villeurbanne, France
| |
Collapse
|
26
|
Batra T, Malik I, Kumar V. Illuminated night alters behaviour and negatively affects physiology and metabolism in diurnal zebra finches. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 254:112916. [PMID: 31376605 DOI: 10.1016/j.envpol.2019.07.084] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/01/2019] [Accepted: 07/16/2019] [Indexed: 05/29/2023]
Abstract
Light at night (LAN) negatively impacts the behaviour and physiology; however, very little is known about molecular correlates of LAN-induced effects in diurnal animals. Here, we assessed LAN-induced effects on behaviour and physiology, and examined molecular changes in the liver of diurnal zebra finches (Taeniopygia guttata). Birds were exposed to dim LAN (dLAN: 12L = 150 lux: 12D = 5 lux), with controls on 12L (150 lux): 12D (0 lux). dLAN altered daily activity-rest and eating patterns, induced nocturnal eating and caused body fattening and weight gain, and reduced nocturnal melatonin levels. Concomitant increased nighttime glucose levels, decreased daytime thyroxine and triglycerides levels, and hepatic lipid accumulation suggested the impairment of metabolism under dLAN. Transcriptional assays evidenced dLAN-induced negative effects on metabolism in the liver, the site of metabolic homeostasis. Particularly, increased g6pc and foxo1 mRNA expressions suggested an enhanced gluconeogenesis, while increased egr1 and star expressions suggested enhanced cholesterol biosynthesis and lipid metabolism, respectively. Similarly, overexpressed sirt1 indicated protection from the metabolic damage due to elevated gluconeogenesis and cholesterol biosynthesis under dLAN. However, no effect on genes involved in lipogenesis (fasn) and insulin signalling pathway (socs3 and insig1) might indicate for the post transcriptional/post translational modification effects or the involvement of other genetic pathways in LAN-induced effects. We also found daily rhythm in the hepatic expression of selected clock and clock-controlled genes (per2, bmal1 and reverb-beta), with an elevated mesor and amplitude of per2 oscillation, suggesting a role of per2 in the liver metabolism. These results demonstrate dLAN-induced negative effects on the behaviour and physiology, and provide molecular insights into metabolic risks of the exposure to illuminated nights to diurnal animals including humans in an urban setting.
Collapse
Affiliation(s)
- Twinkle Batra
- Department of Zoology, University of Delhi, Delhi, 110 007, India
| | - Indu Malik
- Department of Zoology, University of Delhi, Delhi, 110 007, India
| | - Vinod Kumar
- Department of Zoology, University of Delhi, Delhi, 110 007, India.
| |
Collapse
|
27
|
Gjorgjieva M, Mithieux G, Rajas F. Hepatic stress associated with pathologies characterized by disturbed glucose production. Cell Stress 2019; 3:86-99. [PMID: 31225503 PMCID: PMC6551742 DOI: 10.15698/cst2019.03.179] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
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.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France.,Université de Lyon, Lyon, F-69008 France.,Université Lyon I, Villeurbanne, F-69622 France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France.,Université de Lyon, Lyon, F-69008 France.,Université Lyon I, Villeurbanne, F-69622 France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France.,Université de Lyon, Lyon, F-69008 France.,Université Lyon I, Villeurbanne, F-69622 France
| |
Collapse
|
28
|
|
29
|
Goyal N, Tiwary S, Kesharwani D, Datta M. Long non-coding RNA H19 inhibition promotes hyperglycemia in mice by upregulating hepatic FoxO1 levels and promoting gluconeogenesis. J Mol Med (Berl) 2018; 97:115-126. [PMID: 30465059 DOI: 10.1007/s00109-018-1718-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 10/24/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022]
Abstract
In a previous report from our laboratory, it was reported that hepatic levels of the long non-coding RNA (lncRNA), H19 are decreased in diabetic mice which elevates hepatic gluconeogenesis and glucose output. But, the mechanisms of H19 inhibition in elevating gluconeogenic genes' transcription and promoting hepatic glucose output were not known. In this study, we aimed to decipher this regulatory role of H19 on glucose metabolism and on FoxO1, an important transcriptional regulator of gluconeogenesis. While H19 inhibition in HepG2 cells increased the levels of FoxO1, its overexpression led to significant inhibition in FoxO1 levels, thereby identifying H19 as an important regulator of FoxO1. Our data also demonstrates that in the absence of H19, there is increased occupancy of p53 on the FoxO1 promoter that possibly is responsible for increased FoxO1 transcription. In vivo silencing of H19 in normal mice caused hyperglycemia, hyperinsulinemia and impaired glucose, insulin, and pyruvate tolerance. Serum triglyceride and cholesterol levels, however, did not show any change. H19 inhibition significantly elevated the hepatic levels of FoxO1 and all the gluconeogenic genes. While fasting increased gluconeogenic genes' transcription, the levels of H19 were decreased and these patterns reversed upon refeeding the mice. Thus, gluconeogenic genes and H19 levels show inverse patterns of expression, and these results indicate towards an important regulatory role of the lncRNA, H19. It acts as an upstream regulator of gluconeogenesis by regulating the transcription of FoxO1, an important transcription factor of gluconeogenic genes, and hence, regulates hepatic glucose metabolism. KEY MESSAGES: H19 regulates FoxO1 transcript and protein levels. H19 inhibition increases p53 occupancy on the FoxO1 promoter that promotes FoxO1 transcription. H19 inhibition in vivo induces hyperglycemia and impairs glucose, insulin, and pyruvate tolerance. In vivo H19 inhibition increases the hepatic transcript levels of gluconeogenic genes and FoxO1. Transcript levels of H19 and gluconeogenic genes are inversely regulated during fed and fasted states.
Collapse
Affiliation(s)
- Neha Goyal
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India.,CSIR-HRDC, Academy of Scientific and Innovative Research, Kamala Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - Shweta Tiwary
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India.,CSIR-HRDC, Academy of Scientific and Innovative Research, Kamala Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - Devesh Kesharwani
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India.,CSIR-HRDC, Academy of Scientific and Innovative Research, Kamala Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India
| | - Malabika Datta
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, 110007, India. .,CSIR-HRDC, Academy of Scientific and Innovative Research, Kamala Nehru Nagar, Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
30
|
ŠIMJÁK P, CINKAJZLOVÁ A, ANDERLOVÁ K, KLOUČKOVÁ J, KRATOCHVÍLOVÁ H, LACINOVÁ Z, KAVÁLKOVÁ P, KREJČÍ H, MRÁZ M, PAŘÍZEK A, KRŠEK M, HALUZÍK M. Changes in Plasma Concentrations and mRNA Expression of Hepatokines Fetuin A, Fetuin B and FGF21 in Physiological Pregnancy and Gestational Diabetes Mellitus. Physiol Res 2018; 67:S531-S542. [DOI: 10.33549/physiolres.934017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We measured plasma concentrations, adipose tissue and placental mRNA expression of hepatokines fetuin A, fetuin B and fibroblast growth factor 21 (FGF21) in 12 healthy pregnant women (P group), 12 pregnant women with gestational diabetes (GDM) and 10 healthy non-pregnant women (N group) to explore their potential role in the etiopathogenesis of GDM. GDM and P group had comparable BMI, C-reactive protein (CRP) and glycated hemoglobin levels while IL-10 and TNF-α levels were higher in GDM group. Fetuin A and fetuin B levels were higher in pregnancy as compared to N group and decreased after delivery with no apparent influence of GDM. In contrast, the pattern of changes of circulating FGF21 levels differed between GDM and P group. Fetuin A concentrations positively correlated with CRP, TNF-α mRNA expression in adipose tissue and IL-6 mRNA expression in placenta. Fetuin B positively correlated with CRP. FGF21 levels correlated positively with IFN-γ mRNA in adipose tissue and inversely with IL-8 mRNA in the placenta. Taken together, fetuin A and fetuin B levels were increased during pregnancy regardless of the presence of GDM. In contrast, FGF21 patterns differed between healthy pregnant women and GDM patients suggesting a possible role of this hepatokine in the etiopathogenesis of GDM.
Collapse
|
31
|
Gjorgjieva M, Calderaro J, Monteillet L, Silva M, Raffin M, Brevet M, Romestaing C, Roussel D, Zucman-Rossi J, Mithieux G, Rajas F. Dietary exacerbation of metabolic stress leads to accelerated hepatic carcinogenesis in glycogen storage disease type Ia. J Hepatol 2018; 69:1074-1087. [PMID: 30193922 DOI: 10.1016/j.jhep.2018.07.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 06/11/2018] [Accepted: 07/08/2018] [Indexed: 12/18/2022]
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.
Collapse
Affiliation(s)
- Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon F-69008, France; Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France
| | - Julien Calderaro
- Inserm UMR-1162, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France; Université Paris Est Créteil, Créteil, France; APHP, Assistance-Publique Hôpitaux-de-Paris, Département de Pathologie, Hôpital Henri Mondor, Créteil F-94010, France
| | - Laure Monteillet
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon F-69008, France; Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France
| | - Marine Silva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon F-69008, France; Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France
| | - Margaux Raffin
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon F-69008, France; Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France
| | - Marie Brevet
- Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France; Service de Pathologie Lyon Est, Centre hospitalier universitaire de Lyon, Lyon F-69437, France
| | - Caroline Romestaing
- Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France; Centre National de la Recherche Scientifique, UMR 5023, Villeurbanne F-69622 France
| | - Damien Roussel
- Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France; Centre National de la Recherche Scientifique, UMR 5023, Villeurbanne F-69622 France
| | - Jessica Zucman-Rossi
- Inserm UMR-1162, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, Paris, France; Hôpital Européen Georges Pompidou, AP-HP, Assistance Publique-Hôpitaux de Paris, Paris F-75015, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon F-69008, France; Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon F-69008, France; Université de Lyon, Lyon F-69008 France; Université Lyon I, Villeurbanne F-69622 France.
| |
Collapse
|
32
|
Monteillet L, Gjorgjieva M, Silva M, Verzieux V, Imikirene L, Duchampt A, Guillou H, Mithieux G, Rajas F. Intracellular lipids are an independent cause of liver injury and chronic kidney disease in non alcoholic fatty liver disease-like context. Mol Metab 2018; 16:100-115. [PMID: 30100243 PMCID: PMC6157648 DOI: 10.1016/j.molmet.2018.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022] Open
Abstract
Objective Ectopic lipid accumulation in the liver and kidneys is a hallmark of metabolic diseases leading to non-alcoholic fatty liver disease (NAFLD) and chronic kidney disease (CKD). Moreover, recent data have highlighted a strong correlation between NAFLD and CKD incidences. In this study, we use two mouse models of hepatic steatosis or CKD, each initiated independently of the other upon the suppression of glucose production specifically in the liver or kidneys, to elucidate the mechanisms underlying the development of CKD in the context of NAFLD-like pathology. Methods Mice with a deletion of G6pc, encoding glucose-6 phosphatase catalytic subunit, specifically in the liver (L.G6pc−/− mice) or the kidneys (K.G6pc−/− mice), were fed with either a standard diet or a high fat/high sucrose (HF/HS) diet during 9 months. These mice represent two original models of a rare metabolic disease named Glycogen Storage Disease Type Ia (GSDIa) that is characterized by both NAFLD-like pathology and CKD. Two other groups of L.G6pc−/− and K.G6pc−/− mice were fed a standard diet for 6 months and then treated with fenofibrate for 3 months. Lipid and glucose metabolisms were characterized, and NAFLD-like and CKD damages were evaluated. Results Lipid depot exacerbation upon high-calorie diet strongly accelerated hepatic and renal pathologies induced by the G6pc-deficiency. In L.G6pc−/− mice, HF/HS diet increased liver injuries, characterized by higher levels of plasmatic transaminases and increased hepatic tumor incidence. In K.G6pc−/− mice, HF/HS diet increased urinary albumin and lipocalin 2 excretion and aggravated renal fibrosis. In both cases, the worsening of NAFLD-like injuries and CKD was independent of glycogen content. Furthermore, fenofibrate, via the activation of lipid oxidation significantly decreased the hepatic or renal lipid accumulations and prevented liver or kidney damages in L.G6pc−/− and K.G6pc−/− mice, respectively. Finally, we show that L.G6pc−/− mice and K.G6pc−/− mice developed NAFLD-like pathology and CKD independently. Conclusions This study highlights the crucial role that lipids play in the independent development of both NAFLD and CKD and demonstrates the importance of lipid-lowering treatments in various metabolic diseases featured by lipid load, from the “rare” GSDIa to the “epidemic” morbid obesity or type 2 diabetes. Exacerbating lipid accumulation aggravates liver/kidney injury in GSDI. Fenofibrate-mediated PPARα activation induces hepatic and renal lipid turnover. Increased lipid turnover prevents glycogen synthesis and accumulation. PPARα–mediated metabolic reprograming prevents hepatic and renal GSDI complications. NAFLD and CKD develop independently.
Collapse
Affiliation(s)
- Laure Monteillet
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Marine Silva
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Vincent Verzieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Linda Imikirene
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Hervé Guillou
- Toxalim, Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, 31027, France.
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon, F-69008, France; Université de Lyon, Lyon, F-69008, France; Université Lyon1, Villeurbanne, F-69622, France.
| |
Collapse
|
33
|
Prebiotic effects of white button mushroom (Agaricus bisporus) feeding on succinate and intestinal gluconeogenesis in C57BL/6 mice. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
34
|
Disruptions in gut microbial-host co-metabolism and the development of metabolic disorders. Clin Sci (Lond) 2018; 132:791-811. [PMID: 29661926 DOI: 10.1042/cs20171328] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/20/2018] [Accepted: 02/23/2018] [Indexed: 12/14/2022]
Abstract
The microbial-mammalian metabolic axis has become recognized as an important component governing the overall homeostatic balance of the mammalian host. Disruption of the state of homeostasis among the gut microbiota has been shown to be causally linked to the development of host metabolic diseases including obesity, cardiovascular, diabetes, and fatty liver disease. This disruption is often referred to as gut dysbiosis. Gut dysbiosis leads to altered metabolic products derived from the microbiota and these in turn, typically shift the homeostatic metabolic balance of the host towards a low-grade chronic inflammation, a hallmark of metabolic syndrome. The primary objective of this review is to examine and discuss some very current research that has been done to study the effect of bacterial metabolites on host metabolism, sometimes referred to as microbiota-host co-metabolism. The metabolic conditions reviewed here include obesity, a known risk factor for all of the other metabolic conditions, as well as, cardiovascular disease, diabetes and nonalcoholic fatty liver disease. Only by further understanding the cause and result of gut dysbiosis will an adequate solution be found for metabolic disease, a viewpoint shared by many.
Collapse
|
35
|
Liu Y, Wang HY, Zhou L, Su Y, Shen WC. Biodistribution, activation, and retention of proinsulin-transferrin fusion protein in the liver: Mechanism of liver-targeting as an insulin prodrug. J Control Release 2018; 275:186-191. [DOI: 10.1016/j.jconrel.2018.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/08/2018] [Accepted: 02/19/2018] [Indexed: 12/18/2022]
|
36
|
Kinyua AW, Ko CM, Doan KV, Yang DJ, Huynh MKQ, Moh SH, Choi YH, Kim KW. 4-hydroxy-3-methoxycinnamic acid regulates orexigenic peptides and hepatic glucose homeostasis through phosphorylation of FoxO1. Exp Mol Med 2018; 50:e437. [PMID: 29391540 PMCID: PMC5903816 DOI: 10.1038/emm.2017.253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/05/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
4-hydroxy-3-methoxycinnamic acid (ferulic acid, FA) is known to have numerous beneficial health effects, including anti-obesity and anti-hyperglycemic properties. However, the molecular networks that modulate the beneficial FA-induced metabolic effects have not been well elucidated. In this study, we explored the molecular mechanisms mediating the beneficial metabolic effects of FA. In mice, FA protected against high-fat diet-induced weight gain, reduced food intake and exhibited an overall improved metabolic phenotype. The food intake suppression by FA was accompanied by a specific reduction in hypothalamic orexigenic neuropeptides, including agouti-related protein and neuropeptide Y, with no significant changes in the anorexigenic peptides pro-opiomelanocortin and cocaine and amphetamine-regulated transcript. FA treatment also inhibited fat accumulation in the liver and white adipose tissue and suppressed the expression of gluconeogenic genes, including phosphoenolpyruvate carboxylase and glucose-6-phosphatase. Furthermore, we show that FA phosphorylated and inactivated the transcription factor FoxO1, which positively regulates the expression of gluconeogenic and orexigenic genes, providing evidence that FA might exert its beneficial metabolic effects through inhibition of FoxO1 function in the periphery and the hypothalamus.
Collapse
Affiliation(s)
- Ann W Kinyua
- Departments of Pharmacology and Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Chang Mann Ko
- Departments of Pharmacology and Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Khanh V Doan
- Departments of Pharmacology and Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Dong Joo Yang
- Departments of Pharmacology and Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - My Khanh Q Huynh
- Departments of Pharmacology and Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Sang Hyun Moh
- Anti-aging Research Institute of BIO-FD&C Co. Ltd., Incheon, Republic of Korea
| | - Yun-Hee Choi
- Anti-aging Research Institute of BIO-FD&C Co. Ltd., Incheon, Republic of Korea
| | - Ki Woo Kim
- Departments of Pharmacology and Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| |
Collapse
|
37
|
Litwak SA, Pang L, Galic S, Igoillo-Esteve M, Stanley WJ, Turatsinze JV, Loh K, Thomas HE, Sharma A, Trepo E, Moreno C, Gough DJ, Eizirik DL, de Haan JB, Gurzov EN. JNK Activation of BIM Promotes Hepatic Oxidative Stress, Steatosis, and Insulin Resistance in Obesity. Diabetes 2017; 66:2973-2986. [PMID: 28928277 DOI: 10.2337/db17-0348] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 09/13/2017] [Indexed: 11/13/2022]
Abstract
The members of the BCL-2 family are crucial regulators of the mitochondrial pathway of apoptosis in normal physiology and disease. Besides their role in cell death, BCL-2 proteins have been implicated in the regulation of mitochondrial oxidative phosphorylation and cellular metabolism. It remains unclear, however, whether these proteins have a physiological role in glucose homeostasis and metabolism in vivo. In this study, we report that fat accumulation in the liver increases c-Jun N-terminal kinase-dependent BCL-2 interacting mediator of cell death (BIM) expression in hepatocytes. To determine the consequences of hepatic BIM deficiency in diet-induced obesity, we generated liver-specific BIM-knockout (BLKO) mice. BLKO mice had lower hepatic lipid content, increased insulin signaling, and improved global glucose metabolism. Consistent with these findings, lipogenic and lipid uptake genes were downregulated and lipid oxidation enhanced in obese BLKO mice. Mechanistically, BIM deficiency improved mitochondrial function and decreased oxidative stress and oxidation of protein tyrosine phosphatases, and ameliorated activation of peroxisome proliferator-activated receptor γ/sterol regulatory element-binding protein 1/CD36 in hepatocytes from high fat-fed mice. Importantly, short-term knockdown of BIM rescued obese mice from insulin resistance, evidenced by reduced fat accumulation and improved insulin sensitivity. Our data indicate that BIM is an important regulator of liver dysfunction in obesity and a novel therapeutic target for restoring hepatocyte function.
Collapse
Affiliation(s)
- Sara A Litwak
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Lokman Pang
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Sandra Galic
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | | | - William J Stanley
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - Kim Loh
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Arpeeta Sharma
- Oxidative Stress Laboratory, Basic Science Division, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Eric Trepo
- Clinique Universitaire de Bruxelles, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Christophe Moreno
- Clinique Universitaire de Bruxelles, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Daniel J Gough
- Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Judy B de Haan
- Oxidative Stress Laboratory, Basic Science Division, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Esteban N Gurzov
- St. Vincent's Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Abdul-Wahed A, Guilmeau S, Postic C. Sweet Sixteenth for ChREBP: Established Roles and Future Goals. Cell Metab 2017; 26:324-341. [PMID: 28768172 DOI: 10.1016/j.cmet.2017.07.004] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/01/2017] [Accepted: 07/12/2017] [Indexed: 12/25/2022]
Abstract
With the identification of ChREBP in 2001, our interest in understanding the molecular control of carbohydrate sensing has surged. While ChREBP was initially studied as a master regulator of lipogenesis in liver and fat tissue, it is now clear that ChREBP functions as a central metabolic coordinator in a variety of cell types in response to environmental and hormonal signals, with wide implications in health and disease. Celebrating its sweet sixteenth birthday, we review here the current knowledge about the function and regulation of ChREBP throughout usual and less explored tissues, to recapitulate ChREBP's role as a whole-body glucose sensor.
Collapse
Affiliation(s)
- Aya Abdul-Wahed
- Inserm, U1016, Institut Cochin, 75014 Paris, France; CNRS UMR 8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Sandra Guilmeau
- Inserm, U1016, Institut Cochin, 75014 Paris, France; CNRS UMR 8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France
| | - Catherine Postic
- Inserm, U1016, Institut Cochin, 75014 Paris, France; CNRS UMR 8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75006 Paris, France.
| |
Collapse
|
39
|
Lee J, Choi J, Selen Alpergin ES, Zhao L, Hartung T, Scafidi S, Riddle RC, Wolfgang MJ. Loss of Hepatic Mitochondrial Long-Chain Fatty Acid Oxidation Confers Resistance to Diet-Induced Obesity and Glucose Intolerance. Cell Rep 2017; 20:655-667. [PMID: 28723568 PMCID: PMC5546239 DOI: 10.1016/j.celrep.2017.06.080] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/01/2017] [Accepted: 06/27/2017] [Indexed: 11/23/2022] Open
Abstract
The liver has a large capacity for mitochondrial fatty acid β-oxidation, which is critical for systemic metabolic adaptations such as gluconeogenesis and ketogenesis. To understand the role of hepatic fatty acid oxidation in response to a chronic high-fat diet (HFD), we generated mice with a liver-specific deficiency of mitochondrial long-chain fatty acid β-oxidation (Cpt2L-/- mice). Paradoxically, Cpt2L-/- mice were resistant to HFD-induced obesity and glucose intolerance with an absence of liver damage, although they exhibited serum dyslipidemia, hepatic oxidative stress, and systemic carnitine deficiency. Feeding an HFD induced hepatokines in mice, with a loss of hepatic fatty acid oxidation that enhanced systemic energy expenditure and suppressed adiposity. Additionally, the suppression in hepatic gluconeogenesis was sufficient to improve HFD-induced glucose intolerance. These data show that inhibiting hepatic fatty acid oxidation results in a systemic hormetic response that protects mice from HFD-induced obesity and glucose intolerance.
Collapse
Affiliation(s)
- Jieun Lee
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joseph Choi
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ebru S Selen Alpergin
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Liang Zhao
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Thomas Hartung
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biology, University of Konstanz, 78464 Konstanz, Germany
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Baltimore Veterans Administration Medical Center, Baltimore, MD 21201, USA
| | - Michael J Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
40
|
Abstract
Intestinal gluconeogenesis is a recently identified function influencing energy homeostasis. Intestinal gluconeogenesis induced by specific nutrients releases glucose, which is sensed by the nervous system surrounding the portal vein. This initiates a signal positively influencing parameters involved in glucose control and energy management controlled by the brain. This knowledge has extended our vision of the gut-brain axis, classically ascribed to gastrointestinal hormones. Our work raises several questions relating to the conditions under which intestinal gluconeogenesis proceeds and may provide its metabolic benefits. It also leads to questions on the advantage conferred by its conservation through a process of natural selection.
Collapse
Affiliation(s)
- Maud Soty
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon I, Villeurbanne 69622, France
| | - Amandine Gautier-Stein
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon I, Villeurbanne 69622, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon I, Villeurbanne 69622, France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon I, Villeurbanne 69622, France.
| |
Collapse
|
41
|
Casteras S, Abdul-Wahed A, Soty M, Vulin F, Guillou H, Campana M, Le Stunff H, Pirola L, Rajas F, Mithieux G, Gautier-Stein A. The suppression of hepatic glucose production improves metabolism and insulin sensitivity in subcutaneous adipose tissue in mice. Diabetologia 2016; 59:2645-2653. [PMID: 27631137 DOI: 10.1007/s00125-016-4097-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/05/2016] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Despite the strong correlation between non-alcoholic fatty liver disease and insulin resistance, hepatic steatosis is associated with greater whole-body insulin sensitivity in several models. We previously reported that the inhibition of hepatic glucose production (HGP) protects against the development of obesity and diabetes despite severe steatosis, thanks to the secretion of specific hepatokines such as fibroblast growth factor 21 (FGF21) and angiopoietin-related growth factor. In this work, we focused on adipose tissue to assess whether liver metabolic fluxes might, by interorgan communication, control insulin signalling in lean animals. METHODS Insulin signalling was studied in the adipose tissue of mice lacking the catalytic subunit of glucose 6-phosphatase, the key enzyme in endogenous glucose production, in the liver (L-G6pc -/- mice). Morphological and metabolic changes in the adipose tissues were characterised by histological analyses, gene expression and protein content. RESULTS Mice lacking HGP exhibited improved insulin sensitivity of the phosphoinositide 3-kinase/Akt pathway in the subcutaneous adipose tissue associated with a browning of adipocytes. The suppression of HGP increased FGF21 levels in lean animals, and increased FGF21 was responsible for the metabolic changes in the subcutaneous adipose tissue but not for its greater insulin sensitivity. The latter might be linked to an increase in the ratio of monounsaturated to saturated fatty acids released by the liver. CONCLUSIONS Our work provides evidence that HGP controls subcutaneous adipose tissue browning and insulin sensitivity through two pathways: the release of beneficial hepatokines and changes in hepatic fatty acids profile.
Collapse
Affiliation(s)
- Sylvie Casteras
- Inserm U1213, Faculté Laennec, 7 rue Guillaume Paradin, 69372, Lyon cedex 08, France
- Université de Lyon, Lyon, France
- Université Lyon1, Villeurbanne, France
| | - Aya Abdul-Wahed
- Inserm U1213, Faculté Laennec, 7 rue Guillaume Paradin, 69372, Lyon cedex 08, France
- Université de Lyon, Lyon, France
- Université Lyon1, Villeurbanne, France
| | - Maud Soty
- Inserm U1213, Faculté Laennec, 7 rue Guillaume Paradin, 69372, Lyon cedex 08, France
- Université de Lyon, Lyon, France
- Université Lyon1, Villeurbanne, France
| | - Fanny Vulin
- Inserm U1213, Faculté Laennec, 7 rue Guillaume Paradin, 69372, Lyon cedex 08, France
- Université de Lyon, Lyon, France
- Université Lyon1, Villeurbanne, France
| | - Hervé Guillou
- INRA, ToxAlim UMR1331 (Research Center in Food Toxicology), Toulouse, France
| | - Mélanie Campana
- Unité Biologie Fonctionnelle et Adaptative -UMR CNRS 8251, Université Paris- Diderot (7), Paris, France
- I2BC - UMR 9198 Université Paris Sud, Gif sur Yvette, France
| | - Hervé Le Stunff
- Unité Biologie Fonctionnelle et Adaptative -UMR CNRS 8251, Université Paris- Diderot (7), Paris, France
- I2BC - UMR 9198 Université Paris Sud, Gif sur Yvette, France
| | - Luciano Pirola
- Université de Lyon, Lyon, France
- Université Lyon1, Villeurbanne, France
- Laboratoire de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition, CarMeN, Oullins, France
| | - Fabienne Rajas
- Inserm U1213, Faculté Laennec, 7 rue Guillaume Paradin, 69372, Lyon cedex 08, France
- Université de Lyon, Lyon, France
- Université Lyon1, Villeurbanne, France
| | - Gilles Mithieux
- Inserm U1213, Faculté Laennec, 7 rue Guillaume Paradin, 69372, Lyon cedex 08, France
- Université de Lyon, Lyon, France
- Université Lyon1, Villeurbanne, France
| | - Amandine Gautier-Stein
- Inserm U1213, Faculté Laennec, 7 rue Guillaume Paradin, 69372, Lyon cedex 08, France.
- Université de Lyon, Lyon, France.
- Université Lyon1, Villeurbanne, France.
| |
Collapse
|
42
|
Gjorgjieva M, Oosterveer MH, Mithieux G, Rajas F. Mechanisms by Which Metabolic Reprogramming in GSD1 Liver Generates a Favorable Tumorigenic Environment. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2016. [DOI: 10.1177/2326409816679429] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Monika Gjorgjieva
- Institut National de la Santé et de la Recherche Médicale, U1213 “Nutrition, Diabetes and the Brain”, Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| | - Maaike H. Oosterveer
- Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213 “Nutrition, Diabetes and the Brain”, Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| | - Fabienne Rajas
- Institut National de la Santé et de la Recherche Médicale, U1213 “Nutrition, Diabetes and the Brain”, Lyon, France
- Université de Lyon, Lyon, France
- Université Lyon 1, Villeurbanne, France
| |
Collapse
|
43
|
Shao J, Zeng S, Zhou B, Xu H, Bian Y, Xu Y. Angiogenic factor with G patch and FHA domains 1 (Aggf1) promotes hepatic steatosis in mice. Biochem Biophys Res Commun 2016; 482:134-140. [PMID: 27865839 DOI: 10.1016/j.bbrc.2016.10.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 11/18/2022]
Abstract
Increased uptake of nutrients coupled with reduced activity leads to the development of a host of metabolic disorders in humans. In the present study we examined the role of angiogenic factor with G patch and FHA domains 1 (Aggf1) in the pathogenesis of steatosis, characterized by accumulation of lipids in the liver and consequently hepatic insulin resistance. We report here that Aggf1 expression was up-regulated in the liver in both genetically predisposed and diet-induced mouse model of steatosis. Aggf1 expression was also stimulated by free fatty acids in primary hepatocytes. Over-expression of Aggf1 in mice promoted steatosis. On the contrary, Aggf1 depletion ameliorated steatosis in mice. Mechanistically, Aggf1 activated the expression of gluconeogenesis gene and skewed the insulin signaling pathway to induce insulin resistance. Taken together, our data suggest that Aggf1 plays a role in steatosis in vivo and as such may be a new target in the development of therapeutics solutions against steatosis.
Collapse
Affiliation(s)
- Jing Shao
- College of Basic Medical Sciences, Nanjing University of Chinese Medicine, Nanjing, China
| | - Sheng Zeng
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Bisheng Zhou
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China.
| | - Huihui Xu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China
| | - Yaoyao Bian
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yong Xu
- Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
44
|
Rines AK, Sharabi K, Tavares CDJ, Puigserver P. Targeting hepatic glucose metabolism in the treatment of type 2 diabetes. Nat Rev Drug Discov 2016; 15:786-804. [PMID: 27516169 DOI: 10.1038/nrd.2016.151] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus is characterized by the dysregulation of glucose homeostasis, resulting in hyperglycaemia. Although current diabetes treatments have exhibited some success in lowering blood glucose levels, their effect is not always sustained and their use may be associated with undesirable side effects, such as hypoglycaemia. Novel antidiabetic drugs, which may be used in combination with existing therapies, are therefore needed. The potential of specifically targeting the liver to normalize blood glucose levels has not been fully exploited. Here, we review the molecular mechanisms controlling hepatic gluconeogenesis and glycogen storage, and assess the prospect of therapeutically targeting associated pathways to treat type 2 diabetes.
Collapse
Affiliation(s)
- Amy K Rines
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Clint D J Tavares
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, and Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
45
|
De Vadder F, Kovatcheva-Datchary P, Zitoun C, Duchampt A, Bäckhed F, Mithieux G. Microbiota-Produced Succinate Improves Glucose Homeostasis via Intestinal Gluconeogenesis. Cell Metab 2016; 24:151-7. [PMID: 27411015 DOI: 10.1016/j.cmet.2016.06.013] [Citation(s) in RCA: 495] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 05/12/2016] [Accepted: 06/21/2016] [Indexed: 12/16/2022]
Abstract
Beneficial effects of dietary fiber on glucose and energy homeostasis have long been described, focusing mostly on the production of short-chain fatty acids by the gut commensal bacteria. However, bacterial fermentation of dietary fiber also produces large amounts of succinate and, to date, no study has focused on the role of succinate on host metabolism. Here, we fed mice a fiber-rich diet and found that succinate was the most abundant carboxylic acid in the cecum. Dietary succinate was identified as a substrate for intestinal gluconeogenesis (IGN), a process that improves glucose homeostasis. Accordingly, dietary succinate improved glucose and insulin tolerance in wild-type mice, but those effects were absent in mice deficient in IGN. Conventional mice colonized with the succinate producer Prevotella copri exhibited metabolic benefits, which could be related to succinate-activated IGN. Thus, microbiota-produced succinate is a previously unsuspected bacterial metabolite improving glycemic control through activation of IGN.
Collapse
Affiliation(s)
- Filipe De Vadder
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon 1, Villeurbanne 69622, France; Wallenberg Laboratory and Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Petia Kovatcheva-Datchary
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Carine Zitoun
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon 1, Villeurbanne 69622, France
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon 1, Villeurbanne 69622, France
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, University of Gothenburg, 41345 Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark.
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale, U1213, Lyon 69372, France; Université de Lyon, Lyon 69008, France; Université Lyon 1, Villeurbanne 69622, France.
| |
Collapse
|
46
|
Shao J, Zaro JL, Shen WC. Proinsulin-Transferrin Fusion Protein Exhibits a Prolonged and Selective Effect on the Control of Hepatic Glucose Production in an Experimental Model of Type 1 Diabetes. Mol Pharm 2016; 13:2641-6. [PMID: 27280852 DOI: 10.1021/acs.molpharmaceut.6b00168] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An ideal basal insulin (INS) replacement therapy requires the distribution or action of exogenous INS to more closely mimic physiological INS in terms of its preferential hepatic action. In this paper, we introduce a novel strategy to exert liver-specific INS action by hepatic activation of INS's precursor, proinsulin (ProINS). We demonstrated the conversion of human ProINS-transferrin (Tf) fusion protein, ProINS-Tf, into an active and immuno-reactive form of INS-Tf in the liver via the slow Tf receptor mediated recycling pathway. ProINS-Tf displayed prolonged basal blood glucose lowering effects for up to 40 h in streptozotocin-induced type 1 diabetic mice following a single subcutaneous injection. The effect of ProINS-Tf on blood glucose levels was observed predominantly under fasting conditions, with little effect under free-feeding conditions. In addition, both the pyruvate tolerance assay in normal mice and the Akt-phosphorylation assay in H-4-II-E hepatoma cells indicated that the hepatic-activated ProINS-Tf possessed a much longer effect on the control of hepatic glucose production than INS. These results indicated that ProINS-Tf may serve as an effective and safe hepatoselective INS analog to reduce the frequency of INS injections as well as avert severe hypoglycemia episodes and other side effects frequently encountered with long-acting INS therapeutics due to their peripheral action.
Collapse
Affiliation(s)
- Juntang Shao
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , 1985 Zonal Avenue, Los Angeles, California 90033, United States
| | - Jennica L Zaro
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , 1985 Zonal Avenue, Los Angeles, California 90033, United States
| | - Wei-Chiang Shen
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , 1985 Zonal Avenue, Los Angeles, California 90033, United States
| |
Collapse
|
47
|
Soty M, Chilloux J, Delalande F, Zitoun C, Bertile F, Mithieux G, Gautier-Stein A. Post-Translational Regulation of the Glucose-6-Phosphatase Complex by Cyclic Adenosine Monophosphate Is a Crucial Determinant of Endogenous Glucose Production and Is Controlled by the Glucose-6-Phosphate Transporter. J Proteome Res 2016; 15:1342-9. [PMID: 26958868 DOI: 10.1021/acs.jproteome.6b00110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The excessive endogenous glucose production (EGP) induced by glucagon participates in the development of type 2 diabetes. To further understand this hormonal control, we studied the short-term regulation by cyclic adenosine monophosphate (cAMP) of the glucose-6-phosphatase (G6Pase) enzyme, which catalyzes the last reaction of EGP. In gluconeogenic cell models, a 1-h treatment by the adenylate cyclase activator forskolin increased G6Pase activity and glucose production independently of any change in enzyme protein amount or G6P content. Using specific inhibitors or protein overexpression, we showed that the stimulation of G6Pase activity involved the protein kinase A (PKA). Results of site-directed mutagenesis, mass spectrometry analyses, and in vitro phosphorylation experiments suggested that the PKA stimulation of G6Pase activity did not depend on a direct phosphorylation of the enzyme. However, the temperature-dependent induction of both G6Pase activity and glucose release suggested a membrane-based mechanism. G6Pase is composed of a G6P transporter (G6PT) and a catalytic unit (G6PC). Surprisingly, we demonstrated that the increase in G6PT activity was required for the stimulation of G6Pase activity by forskolin. Our data demonstrate the existence of a post-translational mechanism that regulates G6Pase activity and reveal the key role of G6PT in the hormonal regulation of G6Pase activity and of EGP.
Collapse
Affiliation(s)
- Maud Soty
- INSERM U1213, 7-11 rue Paradin, F-69008 Lyon, France.,Université de Lyon, 7-11 rue Paradin, F-69008 Lyon, France.,Université Lyon1, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Julien Chilloux
- INSERM U1213, 7-11 rue Paradin, F-69008 Lyon, France.,Université de Lyon, 7-11 rue Paradin, F-69008 Lyon, France.,Université Lyon1, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - François Delalande
- Institut Pluridisciplinaire Hubert Curien, Département Sciences Analytiques, CNRS UMR7178 , 25 rue Becquerel, 67087 Strasbourg Cedex 2, France.,Université de Strasbourg, 4 rue Blaise Pascal, F-67081 Strasbourg Cedex, France
| | - Carine Zitoun
- INSERM U1213, 7-11 rue Paradin, F-69008 Lyon, France.,Université de Lyon, 7-11 rue Paradin, F-69008 Lyon, France.,Université Lyon1, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Fabrice Bertile
- Institut Pluridisciplinaire Hubert Curien, Département Sciences Analytiques, CNRS UMR7178 , 25 rue Becquerel, 67087 Strasbourg Cedex 2, France.,Université de Strasbourg, 4 rue Blaise Pascal, F-67081 Strasbourg Cedex, France
| | - Gilles Mithieux
- INSERM U1213, 7-11 rue Paradin, F-69008 Lyon, France.,Université de Lyon, 7-11 rue Paradin, F-69008 Lyon, France.,Université Lyon1, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne, France
| | - Amandine Gautier-Stein
- INSERM U1213, 7-11 rue Paradin, F-69008 Lyon, France.,Université de Lyon, 7-11 rue Paradin, F-69008 Lyon, France.,Université Lyon1, 43 Boulevard du 11 Novembre 1918, F-69622 Villeurbanne, France
| |
Collapse
|
48
|
Reprogramming of hepatic fat accumulation and 'browning' of adipose tissue by the short-chain fatty acid acetate. Int J Obes (Lond) 2016; 40:955-63. [PMID: 26975441 DOI: 10.1038/ijo.2016.23] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/30/2015] [Accepted: 12/28/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES Short-chain fatty acids, produced by microbiome fermentation of carbohydrates, have been linked to a reduction in appetite, body weight and adiposity. However, determining the contribution of central and peripheral mechanisms to these effects has not been possible. SUBJECTS/METHODS C57BL/6 mice fed with either normal or high-fat diet were treated with nanoparticle-delivered acetate, and the effects on metabolism were investigated. RESULTS In the liver, acetate decreased lipid accumulation and improved hepatic function, as well as increasing mitochondrial efficiency. In white adipose tissue, it inhibited lipolysis and induced 'browning', increasing thermogenic capacity that led to a reduction in body adiposity. CONCLUSIONS This study provides novel insights into the peripheral mechanism of action of acetate, independent of central action, including 'browning' and enhancement of hepatic mitochondrial function.
Collapse
|
49
|
Iroz A, Couty JP, Postic C. Hepatokines: unlocking the multi-organ network in metabolic diseases. Diabetologia 2015; 58:1699-703. [PMID: 26032022 DOI: 10.1007/s00125-015-3634-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 04/27/2015] [Indexed: 01/16/2023]
Abstract
In the face of urbanisation, surplus energy intake, sedentary habits and obesity, type 2 diabetes has developed into a major health concern worldwide. Commonly overlooked in contemporary obesity research, the liver is emerging as a central regulator of whole body energy homeostasis. Liver-derived proteins known as hepatokines are now considered attractive targets for the development of novel type 2 diabetes treatments. This commentary presents examples of three leading hepatokines: fetuin-A, the first to be described and correlated with increased inflammation and insulin resistance; angiopoietin-like protein (ANGPTL)8/betatrophin, initially proposed for its action on beta cell proliferation, although this effect has recently been brought into question; and fibroblast growth factor 21 (FGF21), an insulin-sensitising hormone that is an appealing drug target because of its beneficial metabolic actions. Novel discoveries in hepatokine research may lead to promising biomarkers and treatments for metabolic disorders and type 2 diabetes. This is one of a series of commentaries under the banner '50 years forward', giving personal opinions on future perspectives in diabetes, to celebrate the 50th anniversary of Diabetologia (1965-2015).
Collapse
Affiliation(s)
- Alison Iroz
- Inserm U1016 Institut Cochin, 24 rue du Faubourg Saint Jacques, 75014, Paris, France
| | | | | |
Collapse
|
50
|
Kim GY, Lee YM, Cho JH, Pan CJ, Jun HS, Springer DA, Mansfield BC, Chou JY. Mice expressing reduced levels of hepatic glucose-6-phosphatase-α activity do not develop age-related insulin resistance or obesity. Hum Mol Genet 2015; 24:5115-25. [PMID: 26089201 DOI: 10.1093/hmg/ddv230] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/15/2015] [Indexed: 12/26/2022] Open
Abstract
Glycogen storage disease type-Ia (GSD-Ia) is caused by a lack of glucose-6-phosphatase-α (G6Pase-α or G6PC) activity. We have shown that gene therapy mediated by a recombinant adeno-associated virus (rAAV) vector expressing human G6Pase-α normalizes blood glucose homeostasis in the global G6pc knockout (G6pc(-/-)) mice for 70-90 weeks. The treated G6pc(-/-) mice expressing 3-63% of normal hepatic G6Pase-α activity (AAV mice) produce endogenous hepatic glucose levels 61-68% of wild-type littermates, have a leaner phenotype and exhibit fasting blood insulin levels more typical of young adult mice. We now show that unlike wild-type mice, the lean AAV mice have increased caloric intake and do not develop age-related obesity or insulin resistance. Pathway analysis shows that signaling by hepatic carbohydrate response element binding protein that improves glucose tolerance and insulin signaling is activated in AAV mice. In addition, several longevity factors in the calorie restriction pathway, including the NADH shuttle systems, NAD(+) concentrations and the AMP-activated protein kinase/sirtuin 1/peroxisome proliferator-activated receptor-γ coactivator 1α pathway are upregulated in the livers of AAV mice. The finding that partial restoration of hepatic G6Pase-α activity in GSD-Ia mice not only attenuates the phenotype of hepatic G6Pase-α deficiency but also prevents the development of age-related obesity and insulin resistance seen in wild-type mice may suggest relevance of the G6Pase-α enzyme to obesity and diabetes.
Collapse
Affiliation(s)
- Goo-Young Kim
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development and
| | - Young Mok Lee
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development and
| | - Jun-Ho Cho
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development and
| | - Chi-Jiunn Pan
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development and
| | - Hyun Sik Jun
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development and
| | - Danielle A Springer
- Mouse Phenotyping Core Facility, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA and
| | - Brian C Mansfield
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development and Foundation Fighting Blindness, Columbia, MD 21046, USA
| | - Janice Y Chou
- Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development and
| |
Collapse
|