1
|
Tan X, Testoni G, Sullivan MA, López-Soldado I, Vilaplana F, Gilbert RG, Guinovart JJ, Schulz BL, Duran J. Glycogenin is dispensable for normal liver glycogen metabolism and body glucose homeostasis. Int J Biol Macromol 2024; 291:139084. [PMID: 39716709 DOI: 10.1016/j.ijbiomac.2024.139084] [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: 11/04/2024] [Revised: 12/08/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
Glycogen is a glucose-storage polysaccharide molecule present in animals, fungi and bacteria. The enzyme glycogenin can self-glycosylate, forming an oligosaccharide chain that primes glycogen synthesis. This priming role of glycogenin was first believed to be essential for glycogen synthesis, but glycogen was then found in the skeletal muscle, heart, liver and brain of glycogenin-knockout mice (Gyg KO), thereby showing that glycogen can be synthesized without glycogenin. Within the liver, glycogen is present in the form of individual glycogen particles, called β particles, and larger composite aggregates of linked β particles, called α particles. Previous studies suggested that liver glycogenin plays a role in linking β particles into α particles and thus participating in glucose homeostasis, which implies that α particles would be absent in Gyg KO mice liver. Here we test this through targeted characterization of glycogen structure and through proteomic and metabolic studies on Gyg KO mice. The results show that, contrary to what had been believed, glycogenin is not necessary for normal liver-glycogen metabolism.
Collapse
Affiliation(s)
- Xinle Tan
- Centre for Animal Science and Centre for Nutrition and Food Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Giorgia Testoni
- Institute for Research in Biomedicine of Barcelona (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Mitchell A Sullivan
- School of Health, University of the Sunshine Coast, Sippy Downs, Queensland 4556, Australia; Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland 4102, Australia
| | - Iliana López-Soldado
- Institute for Research in Biomedicine of Barcelona (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Francisco Vilaplana
- Division of Glycoscience, Department of Chemistry, KTH Royal Institute of Technology, AlbaNova University Centre, 106 91, Stockholm, Sweden
| | - Robert G Gilbert
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Key Laboratory of Plant Functional Genomics of the Ministry of Education, and Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University/Jiangsu Co-Innovation Centre for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, Jiangsu Province 225009, China; Centre for Nutrition and Food Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Joan J Guinovart
- Institute for Research in Biomedicine of Barcelona (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Benjamin L Schulz
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jordi Duran
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona 08017, Spain; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| |
Collapse
|
2
|
Ponte ME, Prom JC, Newcomb MA, Jordan AB, Comfort LL, Hu J, Puchalska P, Geisler CE, Hayes MR, Morris EM. Reduced Liver Mitochondrial Energy Metabolism Impairs Food Intake Regulation Following Gastric Preloads and Fasting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620086. [PMID: 39554188 PMCID: PMC11565831 DOI: 10.1101/2024.10.24.620086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The capacity of the liver to serve as a peripheral sensor in the regulation of food intake has been debated for over half a century. The anatomical position and physiological roles of the liver suggest it is a prime candidate to serve as an interoceptive sensor of peripheral tissue and systemic energy state. Importantly, maintenance of liver ATP levels and within-meal food intake inhibition is impaired in human subjects with obesity and obese pre-clinical models. We demonstrate that decreased hepatic mitochondrial energy metabolism in liver-specific, heterozygous PGC1a mice results in reduced mitochondrial response to changes in ΔGATP and tissue ATP following fasting. These impairments in liver energy state are associated with larger and longer meals during chow feeding, impaired dose-dependent food intake inhibition in response to mixed and individual nutrient oral pre-loads, and greater acute fasting-induced food intake. These data support previous work proposing liver-mediated food intake regulation through modulation of peripheral satiation signals.
Collapse
Affiliation(s)
- Michael E. Ponte
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - John C. Prom
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Mallory A. Newcomb
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Annabelle B. Jordan
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Lucas L. Comfort
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Jiayin Hu
- Dept. of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - Patrycja Puchalska
- Division of Molecular Medicine, University of Minnesota, Minneapolis, MN
| | - Caroline E. Geisler
- Dept. of Psychiatry, University of Pennsylvania, Philadelphia, PA
- Dept. of Pharmaceutical Sciences, University of Kentucky, Lexington, KY
| | - Matthew R. Hayes
- Dept. of Psychiatry, University of Pennsylvania, Philadelphia, PA
| | - E. Matthew Morris
- Dept. of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Center for Children’s Healthy Lifestyle and Nutrition, Children’s Mercy Hospital, Kansas City, Missouri
- University of Kansas Diabetes Institute, Kansas City, Kansas
| |
Collapse
|
3
|
Reis-Costa A, Belew GD, Viegas I, Tavares LC, Meneses MJ, Patrício B, Gastaldelli A, Macedo MP, Jones JG. The Effects of Long-Term High Fat and/or High Sugar Feeding on Sources of Postprandial Hepatic Glycogen and Triglyceride Synthesis in Mice. Nutrients 2024; 16:2186. [PMID: 39064628 PMCID: PMC11279633 DOI: 10.3390/nu16142186] [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: 06/18/2024] [Revised: 07/03/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND In MASLD (formerly called NAFLD) mouse models, oversupply of dietary fat and sugar is more lipogenic than either nutrient alone. Fatty acids suppress de novo lipogenesis (DNL) from sugars, while DNL inhibits fatty acid oxidation. How such factors interact to impact hepatic triglyceride levels are incompletely understood. METHODS Using deuterated water, we measured DNL in mice fed 18-weeks with standard chow (SC), SC supplemented with 55/45-fructose/glucose in the drinking water at 30% (w/v) (HS), high-fat chow (HF), and HF with HS supplementation (HFHS). Liver glycogen levels and its sources were also measured. For HS and HFHS mice, pentose phosphate (PP) fluxes and fructose contributions to DNL and glycogen were measured using [U-13C]fructose. RESULTS The lipogenic diets caused significantly higher liver triglyceride levels compared to SC. DNL rates were suppressed in HF compared to SC and were partially restored in HFHS but supplied a minority of the additional triglyceride in HFHS compared to HF. Fructose contributed a significantly greater fraction of newly synthesized saturated fatty acids compared to oleic acid in both HS and HFHS. Glycogen levels were not different between diets, but significant differences in Direct and Indirect pathway contributions to glycogen synthesis were found. PP fluxes were similar in HS and HFHS mice and were insufficient to account for DNL reducing equivalents. CONCLUSIONS Despite amplifying the lipogenic effects of fat, the fact that sugar-activated DNL per se barely contributes suggests that its role is likely more relevant in the inhibition of fatty acid oxidation. Fructose promotes lipogenesis of saturated over unsaturated fatty acids and contributes to maintenance of glycogen levels. PP fluxes associated with sugar conversion to fat account for a minor fraction of DNL reducing equivalents.
Collapse
Affiliation(s)
- Ana Reis-Costa
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (A.R.-C.); (G.D.B.)
- Center for Neuroscience and Cell Biology (CNC-UC), Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Grupo de Estudos de Investigação Fundamental e Translacional (GIFT) da Sociedade Portuguesa de Diabetologia, 1250-198 Lisboa, Portugal
| | - Getachew D. Belew
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; (A.R.-C.); (G.D.B.)
- Center for Neuroscience and Cell Biology (CNC-UC), Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Ivan Viegas
- Centre for Functional Ecology (CFE), TERRA Associate Laboratory, Department of Life Sciences, University of Coimbra, 3030-790 Coimbra, Portugal;
| | - Ludgero C. Tavares
- Vasco da Gama Research Center (CIVG), University School Vasco da Gama, 3020-210 Coimbra, Portugal;
| | - Maria João Meneses
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (M.J.M.); (B.P.); (M.P.M.)
| | - Bárbara Patrício
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (M.J.M.); (B.P.); (M.P.M.)
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 56124 Pisa, Italy;
- Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Amalia Gastaldelli
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 56124 Pisa, Italy;
- Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Maria Paula Macedo
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal; (M.J.M.); (B.P.); (M.P.M.)
- APDP-Diabetes Portugal Education and Research Center (APDP-ERC), 1250-203 Lisboa, Portugal
| | - John G. Jones
- Center for Neuroscience and Cell Biology (CNC-UC), Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
- Grupo de Estudos de Investigação Fundamental e Translacional (GIFT) da Sociedade Portuguesa de Diabetologia, 1250-198 Lisboa, Portugal
| |
Collapse
|
4
|
Berthoud HR, Münzberg H, Morrison CD, Neuhuber WL. Hepatic interoception in health and disease. Auton Neurosci 2024; 253:103174. [PMID: 38579493 PMCID: PMC11129274 DOI: 10.1016/j.autneu.2024.103174] [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: 12/19/2023] [Revised: 03/14/2024] [Accepted: 03/28/2024] [Indexed: 04/07/2024]
Abstract
The liver is a large organ with crucial functions in metabolism and immune defense, as well as blood homeostasis and detoxification, and it is clearly in bidirectional communication with the brain and rest of the body via both neural and humoral pathways. A host of neural sensory mechanisms have been proposed, but in contrast to the gut-brain axis, details for both the exact site and molecular signaling steps of their peripheral transduction mechanisms are generally lacking. Similarly, knowledge about function-specific sensory and motor components of both vagal and spinal access pathways to the hepatic parenchyma is missing. Lack of progress largely owes to controversies regarding selectivity of vagal access pathways and extent of hepatocyte innervation. In contrast, there is considerable evidence for glucose sensors in the wall of the hepatic portal vein and their importance for glucose handling by the liver and the brain and the systemic response to hypoglycemia. As liver diseases are on the rise globally, and there are intriguing associations between liver diseases and mental illnesses, it will be important to further dissect and identify both neural and humoral pathways that mediate hepatocyte-specific signals to relevant brain areas. The question of whether and how sensations from the liver contribute to interoceptive self-awareness has not yet been explored.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA.
| | - Heike Münzberg
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Christopher D Morrison
- Neurobiology of Nutrition & Metabolism Department, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Winfried L Neuhuber
- Institute for Anatomy and Cell Biology, Friedrich-Alexander University, Erlangen, Germany.
| |
Collapse
|
5
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [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: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
6
|
Sun M, Wan Y, Shi M, Meng ZX, Zeng W. Neural innervation in adipose tissue, gut, pancreas, and liver. LIFE METABOLISM 2023; 2:load022. [PMID: 39872245 PMCID: PMC11749697 DOI: 10.1093/lifemeta/load022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/22/2023] [Accepted: 06/05/2023] [Indexed: 01/30/2025]
Abstract
Efficient communication between the brain and peripheral organs is indispensable for regulating physiological function and maintaining energy homeostasis. The peripheral nervous system (PNS) in vertebrates, consisting of the autonomic and somatic nervous systems, bridges the peripheral organs and the central nervous system (CNS). Metabolic signals are processed by both vagal sensory nerves and somatosensory nerves. The CNS receives sensory inputs via ascending nerves, serves as the coordination and integration center, and subsequently controls internal organs and glands via descending nerves. The autonomic nervous system consists of sympathetic and parasympathetic branches that project peripheral nerves into various anatomical locations to regulate the energy balance. Sympathetic and parasympathetic nerves typically control the reflexive and involuntary functions in organs. In this review article, we outline the innervation of adipose tissue, gut, pancreas, and liver, to illustrate the neurobiological basis of central-peripheral interactions. We emphasize the importance of understanding the functional atlas of neural control of energy metabolism, and more importantly, provide potential avenues for further research in this area.
Collapse
Affiliation(s)
- Mengxue Sun
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Yongwen Wan
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Mengjie Shi
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wenwen Zeng
- Institute for Immunology and School of Medicine, Tsinghua University, and Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| |
Collapse
|
7
|
Hengist A, Davies RG, Rogers PJ, Brunstrom JM, van Loon LJC, Walhin JP, Thompson D, Koumanov F, Betts JA, Gonzalez JT. Restricting sugar or carbohydrate intake does not impact physical activity level or energy intake over 24 h despite changes in substrate use: a randomised crossover study in healthy men and women. Eur J Nutr 2023; 62:921-940. [PMID: 36326863 PMCID: PMC9941259 DOI: 10.1007/s00394-022-03048-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
PURPOSE To determine the effects of dietary sugar or carbohydrate restriction on physical activity energy expenditure, energy intake, and physiological outcomes across 24 h. METHODS In a randomized, open-label crossover design, twenty-five healthy men (n = 10) and women (n = 15) consumed three diets over a 24-h period: moderate carbohydrate and sugar content (MODSUG = 50% carbohydrate [20% sugars], 15% protein, 35% fat); low sugar content (LOWSUG = 50% carbohydrate [< 5% sugars], 15% protein, 35% fat); and low carbohydrate content (LOWCHO = 8% carbohydrate [< 5% sugars], 15% protein, 77% fat). Postprandial metabolic responses to a prescribed breakfast (20% EI) were monitored under laboratory conditions before an ad libitum test lunch, with subsequent diet and physical activity monitoring under free-living conditions until blood sample collection the following morning. RESULTS The MODSUG, LOWSUG and LOWCHO diets resulted in similar mean [95%CI] rates of both physical activity energy expenditure (771 [624, 919] vs. 677 [565, 789] vs. 802 [614, 991] kcal·d-1; p = 0.29] and energy intake (2071 [1794, 2347] vs. 2195 [1918, 2473] vs. 2194 [1890, 2498] kcal·d-1; P = 0.34), respectively. The LOWCHO condition elicited the lowest glycaemic and insulinaemic responses to breakfast (P < 0.01) but the highest 24-h increase in LDL-cholesterol concentrations (P < 0.001), with no differences between the MODSUG and LOWSUG treatments. Leptin concentrations decreased over 24-h of consuming LOWCHO relative to LOWSUG (p < 0.01). CONCLUSION When energy density is controlled for, restricting either sugar or total dietary carbohydrate does not modulate physical activity level or energy intake over a 24-h period (~ 19-h free-living) despite substantial metabolic changes. CLINICAL TRIALS REGISTRATION ID NCT03509610, https://clinicaltrials.gov/show/NCT03509610.
Collapse
Affiliation(s)
- Aaron Hengist
- Department for Health, University of Bath, Bath, BA2 7AY, UK
- Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, UK
| | - Russell G Davies
- Department for Health, University of Bath, Bath, BA2 7AY, UK
- Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, UK
| | - Peter J Rogers
- School of Psychological Sciences, University of Bristol, Bristol, UK
| | - Jeff M Brunstrom
- School of Psychological Sciences, University of Bristol, Bristol, UK
| | - Luc J C van Loon
- Department of Human Biology, Maastricht University, Maastricht, The Netherlands
| | - Jean-Philippe Walhin
- Department for Health, University of Bath, Bath, BA2 7AY, UK
- Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, UK
| | - Dylan Thompson
- Department for Health, University of Bath, Bath, BA2 7AY, UK
- Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, UK
| | - Françoise Koumanov
- Department for Health, University of Bath, Bath, BA2 7AY, UK
- Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, UK
| | - James A Betts
- Department for Health, University of Bath, Bath, BA2 7AY, UK
- Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, UK
| | - Javier T Gonzalez
- Department for Health, University of Bath, Bath, BA2 7AY, UK.
- Centre for Nutrition, Exercise and Metabolism, University of Bath, Bath, UK.
| |
Collapse
|
8
|
Active Glycogen Synthase in the Liver Prevents High-Fat Diet-Induced Glucose Intolerance, Decreases Food Intake, and Lowers Body Weight. Int J Mol Sci 2023; 24:ijms24032574. [PMID: 36768897 PMCID: PMC9917303 DOI: 10.3390/ijms24032574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
Many lines of evidence demonstrate a correlation between liver glycogen content and food intake. We previously demonstrated that mice overexpressing protein targeting to glycogen (PTG) specifically in the liver-which have increased glycogen content in this organ-are protected from high-fat diet (HFD)-induced obesity by reduced food intake. However, the use of PTG to increase liver glycogen implies certain limitations. PTG stimulates glycogen synthesis but also inhibits the enzyme responsible for glycogen degradation. Furthermore, as PTG is a regulatory subunit of protein phosphatase 1 (PP1), which regulates many cellular functions, its overexpression could have side effects beyond the regulation of glycogen metabolism. Therefore, it is necessary to determine whether the direct activation of glycogen synthesis, without affecting its degradation or other cellular functions, has the same effects. To this end, we generated mice overexpressing a non-inactivatable form of glycogen synthase (GS) specifically in the liver (9A-MGSAlb mice). Control and 9a-MGSAlb mice were fed a standard diet (SD) or HFD for 16 weeks. Glucose tolerance and feeding behavior were analyzed. 9A-MGSAlb mice showed an increase in hepatic glycogen in fed and fasting conditions. When fed an HFD, these animals preserved their hepatic energy state, had a reduced food intake, and presented a lower body weight and fat mass than control animals, without changes in energy expenditure. Furthermore, 9A-MGSAlb animals showed improved glucose tolerance when fed an SD or HFD. Moreover, liver triacylglycerol levels that were increased after HFD feeding were lower in these mice. These results confirm that increased liver glycogen stores contribute to decreased appetite and improve glucose tolerance in mice fed an HFD. On the basis of our findings, strategies to preserve hepatic glycogen stores emerge as potential treatments for obesity and hyperglycemia.
Collapse
|
9
|
Yan C, Dai C, Liu N, Qian W, Yang P, Hou X. Effects of Simo decoction on gastric motility of diabetic rats. Neurogastroenterol Motil 2022; 34:e14450. [PMID: 36111645 DOI: 10.1111/nmo.14450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 06/27/2022] [Accepted: 07/27/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND To investigate the effects of simo decoction (SMD) on the gastric motility of diabetic rats. METHODS Diabetic rats were gavaged with various doses of SMD (0.15, 1.5, and 3.0 ml/kg/d) or saline, and their blood glucose levels and body weight were monitored. Gastric emptying and antral motility were assessed by phenol red retention and contractions of antral strips, respectively. The levels of substance P (SP), vasoactive intestinal peptide (VIP), and neurogenic nitric oxide synthase (nNOS) in the gastric antrum were determined by real-time polymerase chain reaction and Western blot. RESULTS Gastric emptying was delayed in diabetic rats (p < 0.01 vs. non-diabetic controls) but accelerated after SMD administration (p < 0.01). The contractions of antral strips were reduced in diabetic rats (p < 0.01 vs. non-diabetic controls) but improved after SMD intervention (p < 0.05). The mRNA expressions of SP, VIP, and nNOS in diabetic rats were downregulated compared with non-diabetic controls (all p < 0.01). Simo decoction treatment did not affect the expression of these factors in diabetic rats. The protein levels of SP, VIP, and nNOS in diabetic rats were decreased (p < 0.01), increased (p < 0.01), and comparable (p > 0.05), respectively, in comparison with non-diabetic controls. Simo decoction administration increased SP protein expression (p < 0.01) and decreased the levels of VIP (p < 0.01) and nNOS (p < 0.01) in diabetic rats. CONCLUSIONS Simo decoction improved gastric dysmotility of diabetic rats possibly by upregulating SP and downregulating VIP and nNOS.
Collapse
Affiliation(s)
- Caihua Yan
- Department of Gastroenterology, Renhe Hospital Affiliated to China Three Gorges University, Yichang, China
| | - Chibing Dai
- Department of Gastroenterology, Renhe Hospital Affiliated to China Three Gorges University, Yichang, China
| | - Na Liu
- Department of Gastroenterology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Qian
- Department of Gastroenterology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengchun Yang
- Department of Gastroenterology, Renhe Hospital Affiliated to China Three Gorges University, Yichang, China
| | - Xiaohua Hou
- Department of Gastroenterology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
D'Alessandro VF, Takeshita A, Yasuma T, Toda M, D'Alessandro-Gabazza CN, Okano Y, Tharavecharak S, Inoue C, Nishihama K, Fujimoto H, Kobayashi T, Yano Y, Gabazza EC. Transforming Growth Factorβ1 Overexpression Is Associated with Insulin Resistance and Rapidly Progressive Kidney Fibrosis under Diabetic Conditions. Int J Mol Sci 2022; 23:ijms232214265. [PMID: 36430743 PMCID: PMC9693927 DOI: 10.3390/ijms232214265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Diabetes mellitus is a global health problem. Diabetic nephropathy is a common complication of diabetes mellitus and the leading cause of end-stage renal disease. The clinical course, response to therapy, and prognosis of nephropathy are worse in diabetic than in non-diabetic patients. The role of transforming growth factorβ1 in kidney fibrosis is undebatable. This study assessed whether the overexpression of transforming growth factorβ1 is associated with insulin resistance and the rapid progression of transforming growth factorβ1-mediated nephropathy under diabetic conditions. Diabetes mellitus was induced with streptozotocin in wild-type mice and transgenic mice with the kidney-specific overexpression of human transforming growth factorβ1. Mice treated with saline were the controls. Glucose tolerance and kidney fibrosis were evaluated. The blood glucose levels, the values of the homeostasis model assessment for insulin resistance, and the area of kidney fibrosis were significantly increased, and the renal function was significantly impaired in the diabetic transforming growth factorβ1 transgenic mice compared to the non-diabetic transgenic mice, diabetic wild-type mice, and non-diabetic mice. Transforming growth factorβ1 impaired the regulatory effect of insulin on glucose in the hepatocyte and skeletal muscle cell lines. This study shows that transforming growth factorβ1 overexpression is associated with insulin resistance and rapidly progressive kidney fibrosis under diabetic conditions in mice.
Collapse
Affiliation(s)
- Valeria Fridman D'Alessandro
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Atsuro Takeshita
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Taro Yasuma
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Masaaki Toda
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Corina N D'Alessandro-Gabazza
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Yuko Okano
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Suphachai Tharavecharak
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Chisa Inoue
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Kota Nishihama
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Hajime Fujimoto
- Department of Pulmonary and Critical care Medicine, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Tetsu Kobayashi
- Department of Pulmonary and Critical care Medicine, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Yutaka Yano
- Department of Diabetes and Endocrinology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| | - Esteban C Gabazza
- Department of Immunology, Mie University Faculty and Graduate School of Medicine, Edobashi 2-174, Tsu 514-8507, Japan
| |
Collapse
|
11
|
Addition of Fructose to a Carbohydrate-Rich Breakfast Improves Cycling Endurance Capacity in Trained Cyclists. Int J Sport Nutr Exerc Metab 2022; 32:439-445. [PMID: 36041732 DOI: 10.1123/ijsnem.2022-0067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 12/26/2022]
Abstract
It was previously demonstrated that postexercise ingestion of fructose-glucose mixtures can lead to superior liver and equal muscle glycogen synthesis as compared with glucose-based carbohydrates (CHOs) only. After an overnight fast, liver glycogen stores are reduced, and based on this we hypothesized that addition of fructose to a glucose-based breakfast would lead to improved subsequent endurance exercise capacity. In this double-blind cross-over randomized study (eight males, peak oxygen uptake: 62.2 ± 5.4 ml·kg-1·min-1), participants completed two experimental trials consisting of two exercise bouts. In the afternoon of Day 1, they completed a cycling interval training session to normalize glycogen stores after which a standardized high-CHO diet was provided for 4 hr. On Day 2, in the morning, participants received 2 g/kg of CHOs in the form of glucose and rice or fructose and rice, both in a CHO ratio of 1:2. Two hours later they commenced cycling exercise session at the intensity of the first ventilatory threshold until task failure. Exercise capacity was higher in fructose and rice (137.0 ± 22.7 min) as compared with glucose and rice (130.06 ± 19.87 min; p = .046). Blood glucose and blood lactate did not differ between the trials (p > .05) and neither did CHO and fat oxidation rates (p > .05). However, due to the duration of exercise, total CHO oxidation was higher in fructose and rice (326 ± 60 g vs. 298 ± 61 g, p = .009). Present data demonstrate that addition of fructose to a glucose-based CHO source at breakfast improves endurance exercise capacity. Further studies are required to determine the mechanisms and optimal dose and ratio.
Collapse
|
12
|
Casagrande BP, Bueno AA, Pisani LP, Estadella D. Hepatic glycogen participates in the regulation of hypothalamic pAkt/Akt ratio in high-sugar/high-fat diet-induced obesity. Metab Brain Dis 2022; 37:1423-1434. [PMID: 35316448 DOI: 10.1007/s11011-022-00944-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/22/2022] [Indexed: 01/07/2023]
Abstract
The hypothalamus is a major integrating centre that controls energy homeostasis and plays a major role in hepatic glycogen (HGlyc) turnover. Not only do hypothalamic and hepatic Akt levels influence glucose homeostasis and glycogen synthesis, but exposure to high-sugar/high-fat diets (HSHF) can also lead to hypothalamic inflammation and HGlyc accumulation. HSHF withdrawal overall restores energy and glucose homeostasis, but the actual relationship between hypothalamic inflammation and HGlyc after short-term HSHF withdrawal has not yet been fully elucidated. Here we investigated the short-term effects of HSHF withdrawal preceded by a 30-day HSHF intake on the liver-hypothalamus crosstalk and glucose homeostasis. Sixty-day old male Wistar rats were fed for 30 days a control chow (n = 10) (Ct), or an HSHF diet (n = 20). On the 30th day of dietary intervention, a random HSHF subset (n = 10) had their diets switched to control chow for 48 h (Hw) whilst the remaining HSHF rats remained in the HSHF diet (n = 10) (Hd). All rats were anaesthetized and euthanized at the end of the protocol. We quantified HGlyc, Akt phosphorylation, inflammation and glucose homeostasis biomarkers. We also assessed the effect of propensity to obesity on those biomarkers, as detailed previously. Hd rats showed impaired glucose homeostasis, higher HGlyc and hypothalamic inflammation, and lower pAkt/Akt. Increased HGlyc was significantly associated with HSHF intake on pAkt/Akt lowered levels. We also found that HGlyc breakdown may have prevented a further pAkt/Akt drop after HSHF withdrawal. Propensity to obesity showed no apparent effect on hypothalamic inflammation or glucose homeostasis. Our findings suggest a comprehensive role of HGlyc as a structural and functional modulator of energy metabolism, and such roles may come into play relatively rapidly.
Collapse
Affiliation(s)
- Breno P Casagrande
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Allain A Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Henwick Grove, WR2 6AJ, Worcester, United Kingdom
| | - Luciana P Pisani
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil
| | - Debora Estadella
- Biosciences Department, Institute of Health and Society, Federal University of São Paulo-UNIFESP/BS, 1015-020, Santos, São Paulo, Brazil.
| |
Collapse
|
13
|
Effects of Monascus purpureus-fermented tartary buckwheat extract on the blood lipid profile, glucose tolerance and antioxidant enzyme activities in KM mice. J Cereal Sci 2022. [DOI: 10.1016/j.jcs.2022.103465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
14
|
López-Soldado I, Guinovart JJ, Duran J. Hepatic overexpression of protein targeting to glycogen attenuates obesity and improves hyperglycemia in db/db mice. Front Endocrinol (Lausanne) 2022; 13:969924. [PMID: 36157460 PMCID: PMC9500150 DOI: 10.3389/fendo.2022.969924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Increased liver glycogen content has been shown to reduce food intake, attenuate obesity, and improve glucose tolerance in a mouse model of high-fat diet (HFD)-induced obesity. Here we studied the contribution of liver glycogen to the regulation of obesity and glucose metabolism in a model of type 2 diabetes and obesity, namely the db/db mouse. To this end, we crossed db/db mice with animals overexpressing protein targeting to glycogen (PTG) in the liver to generate db/db mice with increased liver glycogen content (db/db-PTG). Hepatic PTG overexpression reduced food intake and fat weight and attenuated obesity and hyperglycemia in db/db mice. Db/db-PTG mice showed similar energy expenditure and physical activity to db/db mice. PTG overexpression reduced liver phosphoenolpyruvate carboxykinase (PEPCK) protein levels and repressed hepatic glucose production in db/db mice. Moreover, increased liver glycogen elevated hepatic ATP content in these animals. However, lipid metabolism was not modified by PTG overexpression. In conclusion, increased liver glycogen content ameliorates the diabetic and obesity phenotype in db/db mice.
Collapse
Affiliation(s)
- Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
- *Correspondence: Iliana López-Soldado,
| | - Joan J. Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
15
|
López-Soldado I, Guinovart JJ, Duran J. Increased liver glycogen levels enhance exercise capacity in mice. J Biol Chem 2021; 297:100976. [PMID: 34284060 PMCID: PMC8350413 DOI: 10.1016/j.jbc.2021.100976] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/22/2022] Open
Abstract
Muscle glycogen depletion has been proposed as one of the main causes of fatigue during exercise. However, few studies have addressed the contribution of liver glycogen to exercise performance. Using a low-intensity running protocol, here, we analyzed exercise capacity in mice overexpressing protein targeting to glycogen (PTG) specifically in the liver (PTGOE mice), which show a high concentration of glycogen in this organ. PTGOE mice showed improved exercise capacity, as determined by the distance covered and time ran in an extenuating endurance exercise, compared with control mice. Moreover, fasting decreased exercise capacity in control mice but not in PTGOE mice. After exercise, liver glycogen stores were totally depleted in control mice, but PTGOE mice maintained significant glycogen levels even in fasting conditions. In addition, PTGOE mice displayed an increased hepatic energy state after exercise compared with control mice. Exercise caused a reduction in the blood glucose concentration in control mice that was less pronounced in PTGOE mice. No changes were found in the levels of blood lactate, plasma free fatty acids, or β-hydroxybutyrate. Plasma glucagon was elevated after exercise in control mice, but not in PTGOE mice. Exercise-induced changes in skeletal muscle were similar in both genotypes. These results identify hepatic glycogen as a key regulator of endurance capacity in mice, an effect that may be exerted through the maintenance of blood glucose levels.
Collapse
Affiliation(s)
- Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain; Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Barcelona, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
16
|
Geisler CE, Ghimire S, Bruggink SM, Miller KE, Weninger SN, Kronenfeld JM, Yoshino J, Klein S, Duca FA, Renquist BJ. A critical role of hepatic GABA in the metabolic dysfunction and hyperphagia of obesity. Cell Rep 2021; 35:109301. [PMID: 34192532 PMCID: PMC8851954 DOI: 10.1016/j.celrep.2021.109301] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 04/17/2021] [Accepted: 06/03/2021] [Indexed: 12/31/2022] Open
Abstract
Hepatic lipid accumulation is a hallmark of type II diabetes (T2D) associated with hyperinsulinemia, insulin resistance, and hyperphagia. Hepatic synthesis of GABA, catalyzed by GABA-transaminase (GABA-T), is upregulated in obese mice. To assess the role of hepatic GABA production in obesity-induced metabolic and energy dysregulation, we treated mice with two pharmacologic GABA-T inhibitors and knocked down hepatic GABA-T expression using an antisense oligonucleotide. Hepatic GABA-T inhibition and knockdown decreased basal hyperinsulinemia and hyperglycemia and improved glucose intolerance. GABA-T knockdown improved insulin sensitivity assessed by hyperinsulinemic-euglycemic clamps in obese mice. Hepatic GABA-T knockdown also decreased food intake and induced weight loss without altering energy expenditure in obese mice. Data from people with obesity support the notion that hepatic GABA production and transport are associated with serum insulin, homeostatic model assessment for insulin resistance (HOMA-IR), T2D, and BMI. These results support a key role for hepatocyte GABA production in the dysfunctional glucoregulation and feeding behavior associated with obesity.
Collapse
Affiliation(s)
- Caroline E Geisler
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susma Ghimire
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Stephanie M Bruggink
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Kendra E Miller
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Savanna N Weninger
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Jason M Kronenfeld
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Jun Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Frank A Duca
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Benjamin J Renquist
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
17
|
Geisler CE, Ghimire S, Hepler C, Miller KE, Bruggink SM, Kentch KP, Higgins MR, Banek CT, Yoshino J, Klein S, Renquist BJ. Hepatocyte membrane potential regulates serum insulin and insulin sensitivity by altering hepatic GABA release. Cell Rep 2021; 35:109298. [PMID: 34192533 PMCID: PMC8341405 DOI: 10.1016/j.celrep.2021.109298] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 04/17/2021] [Accepted: 06/03/2021] [Indexed: 01/10/2023] Open
Abstract
Hepatic lipid accumulation in obesity correlates with the severity of hyperinsulinemia and systemic insulin resistance. Obesity-induced hepatocellular lipid accumulation results in hepatocyte depolarization. We have established that hepatocyte depolarization depresses hepatic afferent vagal nerve firing, increases GABA release from liver slices, and causes hyperinsulinemia. Preventing hepatic GABA release or eliminating the ability of the liver to communicate to the hepatic vagal nerve ameliorates the hyperinsulinemia and insulin resistance associated with diet-induced obesity. In people with obesity, hepatic expression of GABA transporters is associated with glucose infusion and disposal rates during a hyperinsulinemic euglycemic clamp. Single-nucleotide polymorphisms in hepatic GABA re-uptake transporters are associated with an increased incidence of type 2 diabetes mellitus. Herein, we identify GABA as a neuro-hepatokine that is dysregulated in obesity and whose release can be manipulated to mute or exacerbate the glucoregulatory dysfunction common to obesity.
Collapse
Affiliation(s)
- Caroline E Geisler
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susma Ghimire
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Chelsea Hepler
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA; Robert H. Lurie Medical Research Center, Northwestern University, Chicago, IL 60611, USA
| | - Kendra E Miller
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Stephanie M Bruggink
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Kyle P Kentch
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Mark R Higgins
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA
| | | | - Jun Yoshino
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Benjamin J Renquist
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
18
|
Zheng Z, Zhang W, Xin C, Zeng N, Li M, Liu X, Cai J, Meng F, Liu D, Zhang J, Yin J, Zhang J, Zhang Z. Laparoscopic total left-sided surgical approach versus traditional bilateral surgical approach for treating hiatal hernia: a study protocol for a randomized controlled trial. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:951. [PMID: 34350266 PMCID: PMC8263855 DOI: 10.21037/atm-20-8000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
Background In China, guidelines for the treatment of hiatal hernia (HH) are lacking. Furthermore, efficacy and safety assessments of surgical approaches for HH and for the protection of the vagus nerve and organ function are needed. Therefore, the present clinical trial is being conducted to establish the normative treatment for HH. Methods The current trial is an ongoing, single-center, randomized controlled trial of patients with HH. The total sample size required for the trial (July 2020–December 2023) is approximately 114 patients. Patients will be randomly assigned to either an experimental group (total left-sided surgical approach; TLSA) or a control group (traditional bilateral surgical approach; TBSA) at a ratio of 1:1 using the block randomization method. We will use case report forms (CRFs) and electronic data capture (EDC) systems to obtain demographic information, preoperative laboratory tests, auxiliary examination results, operation information, and postoperative condition. The patients will be followed up for 3 years after surgery. The primary endpoint is the gastrointestinal quality-of-life index (GIQLI) at 1 year. The secondary endpoints include an efficacy evaluation index [consisting of the incidence of gallstones and gastric emptying disorders, gastrointestinal function recovery time, visual analog scale (VAS) scores, objective evaluation of postoperative indices, and surgical information] and a safety evaluation index (consisting of the incidence of postoperative complications, the 30-day postoperative mortality rate, and the HH recurrence rate at 1 and 3 years after surgery). Discussion TLSA can protect the normal physiological function of organs to a certain extent by protecting the vagus nerve from injury, and has satisfactory short- and long-term efficacy. There is no significant difference in the incidence of postoperative complications and surgical safety between TLSA and TBSA. Our findings will facilitate clinical decision-making for HH and improve the life quality of patients. Trial registration Chinese Clinical Trial Registry, ChiCTR2000034028 (registration date: June 21, 2020).
Collapse
Affiliation(s)
- Zhi Zheng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Weitao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Chenglin Xin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Na Zeng
- National Clinical Research Center for Digestive Diseases, Beijing, China.,Clinical Epidemiology and Evidence-Based Medicine Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mengyi Li
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Xiaoye Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Jun Cai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Fandong Meng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dong Liu
- Department of Ultrasonography, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jie Zhang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jie Yin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Jun Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China.,Beijing Institute of Clinical Medicine, Beijing, China
| |
Collapse
|
19
|
Wei R, Zhuge X, Yue P, Liu M, Zhu L, Liu J, Xia C. Effect of hepatic sympathetic nerve removal on energy metabolism in an animal model of cognitive impairment and its relationship to Glut2 expression. Open Life Sci 2021; 15:311-317. [PMID: 33817219 PMCID: PMC7874542 DOI: 10.1515/biol-2020-0033] [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: 01/09/2020] [Revised: 02/25/2020] [Accepted: 03/19/2020] [Indexed: 01/14/2023] Open
Abstract
The aims of this study were to investigate the effect of hepatic sympathetic nerve removal on glucose and lipid metabolism in rats with cognitive impairment and to evaluate the relationship between these effects and liver Glut2 expression. Hippocampal injection of Aβ1–42 was used to induce cognitive impairment. Impaired rats were divided into experimental, sham, and control groups. The experimental group was injected with 6-hydroxydopamine to remove the sympathetic nerve. At 4 weeks post injection, body weight, food and water intake, blood sugar, and blood lipids were measured, and periodic acid-Schiff (PAS) staining was used to assess the liver glycogen content. Liver Glut2 mRNA and protein were also detected. The experimental group showed reduced body weight, food intake, and blood glucose levels and elevated insulin levels compared with the control group. PAS staining showed higher glycogen contents in the experimental group than in controls. The expression levels of Glut2 mRNA and protein in the experimental group were significantly lower than in the controls. Metabolism was significantly impacted in rats with cognitive impairment following removal of the hepatic sympathetic nerve. Disruption to Glut2 liver expression via sympathetic nerve disruption represents a possible underlying mechanism.
Collapse
Affiliation(s)
- Riming Wei
- College of Biotechnology, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Xiuhong Zhuge
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Pengpeng Yue
- College of Biotechnology, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Manjun Liu
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Lin Zhu
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Jianxiang Liu
- College of Biotechnology, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Chunbo Xia
- Department of Human Anatomy of Basic Medical College, Guilin Medical University, Guilin, Guangxi, 541004, China
| |
Collapse
|
20
|
Siqueira BS, Ceglarek VM, Gomes ECZ, Vettorazzi JF, Rentz T, Nenevê JZ, Volinski KZ, Moraes SS, Malta A, de Freitas Mathias PC, de Oliveira Emilio HR, Balbo SL, Grassiolli S. Vagotomy and Splenectomy Reduce Insulin Secretion and Interleukin-1β. Pancreas 2021; 50:607-616. [PMID: 33939676 DOI: 10.1097/mpa.0000000000001809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES This study aimed to evaluate the effect of vagotomy, when associated with splenectomy, on adiposity and glucose homeostasis in Wistar rats. METHODS Rats were divided into 4 groups: vagotomized (VAG), splenectomized (SPL), VAG + SPL, and SHAM. Glucose tolerance tests were performed, and physical and biochemical parameters evaluated. Glucose-induced insulin secretion and protein expression (Glut2/glucokinase) were measured in isolated pancreatic islets. Pancreases were submitted to histological and immunohistochemical analyses, and vagus nerve neural activity was recorded. RESULTS The vagotomized group presented with reduced body weight, growth, and adiposity; high food intake; reduced plasma glucose and triglyceride levels; and insulin resistance. The association of SPL with the VAG surgery attenuated, or abolished, the effects of VAG and reduced glucose-induced insulin secretion and interleukin-1β area in β cells, in addition to lowering vagal activity. CONCLUSIONS The absence of the spleen attenuated or blocked the effects of VAG on adiposity, triglycerides and glucose homeostasis, suggesting a synergistic effect of both on metabolism. The vagus nerve and spleen modulate the presence of interleukin-1β in β cells, possibly because of the reduction of glucose-induced insulin secretion, indicating a bidirectional flow between autonomous neural firing and the spleen, with repercussions for the endocrine pancreas.
Collapse
Affiliation(s)
- Bruna Schumaker Siqueira
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Vanessa Marieli Ceglarek
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | | | | | - Thiago Rentz
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas
| | - Juliane Zanon Nenevê
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Karoline Zanella Volinski
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Sandra Schmidt Moraes
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá (UEM), Maringá
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá (UEM), Maringá
| | | | - Sandra Lucinei Balbo
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| | - Sabrina Grassiolli
- From the Laboratory of Endocrine Physiology and Metabolism (LAFEM), Western Paraná State University (UNIOESTE)
| |
Collapse
|
21
|
Huang SSY, Makhlouf M, AbouMoussa EH, Ruiz Tejada Segura ML, Mathew LS, Wang K, Leung MC, Chaussabel D, Logan DW, Scialdone A, Garand M, Saraiva LR. Differential regulation of the immune system in a brain-liver-fats organ network during short-term fasting. Mol Metab 2020; 40:101038. [PMID: 32526449 PMCID: PMC7339127 DOI: 10.1016/j.molmet.2020.101038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/24/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Fasting regimens can promote health, mitigate chronic immunological disorders, and improve age-related pathophysiological parameters in animals and humans. Several ongoing clinical trials are using fasting as a potential therapy for various conditions. Fasting alters metabolism by acting as a reset for energy homeostasis, but the molecular mechanisms underlying the beneficial effects of short-term fasting (STF) are not well understood, particularly at the systems or multiorgan level. METHODS We performed RNA-sequencing in nine organs from mice fed ad libitum (0 h) or subjected to fasting five times (2-22 h). We applied a combination of multivariate analysis, differential expression analysis, gene ontology, and network analysis for an in-depth understanding of the multiorgan transcriptome. We used literature mining solutions, LitLab™ and Gene Retriever™, to identify the biological and biochemical terms significantly associated with our experimental gene set, which provided additional support and meaning to the experimentally derived gene and inferred protein data. RESULTS We cataloged the transcriptional dynamics within and between organs during STF and discovered differential temporal effects of STF among organs. Using gene ontology enrichment analysis, we identified an organ network sharing 37 common biological pathways perturbed by STF. This network incorporates the brain, liver, interscapular brown adipose tissue, and posterior-subcutaneous white adipose tissue; hence, we named it the brain-liver-fats organ network. Using Reactome pathways analysis, we identified the immune system, dominated by T cell regulation processes, as a central and prominent target of systemic modulations during STF in this organ network. The changes we identified in specific immune components point to the priming of adaptive immunity and parallel the fine-tuning of innate immune signaling. CONCLUSIONS Our study provides a comprehensive multiorgan transcriptomic profiling of mice subjected to multiple periods of STF and provides new insights into the molecular modulators involved in the systemic immunotranscriptomic changes that occur during short-term energy loss.
Collapse
Affiliation(s)
| | | | | | - Mayra L Ruiz Tejada Segura
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Marchioninistraße 25, 81377, München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| | | | - Kun Wang
- Sidra Medicine, PO Box 26999, Doha, Qatar.
| | | | | | - Darren W Logan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, Marchioninistraße 25, 81377, München, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany; Institute of Computational Biology, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
| | | | - Luis R Saraiva
- Sidra Medicine, PO Box 26999, Doha, Qatar; Monell Chemical Senses Center, 3500 Market Street, Philadelphia, PA, 19104, USA.
| |
Collapse
|
22
|
Huang KP, Goodson ML, Vang W, Li H, Page AJ, Raybould HE. Leptin signaling in vagal afferent neurons supports the absorption and storage of nutrients from high-fat diet. Int J Obes (Lond) 2020; 45:348-357. [PMID: 32917985 PMCID: PMC7854885 DOI: 10.1038/s41366-020-00678-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/30/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022]
Abstract
Objective: Activation of vagal afferent neurons (VAN) by postprandial gastrointestinal signals terminates feeding and facilitates nutrient digestion and absorption. Leptin modulates responsiveness of VAN to meal-related gastrointestinal signals. Rodents with high-fat diet (HF) feeding develop leptin resistance that impairs responsiveness of VAN. We hypothesized that lack of leptin signaling in VAN reduces responses to meal-related signals, which in turn decreases absorption of nutrients and energy storage from high-fat, calorically dense food. Methods: Mice with conditional deletion of the leptin receptor from VAN (Nav1.8-Cre/LepRfl/fl; KO) were used in this study. Six-week-old male mice were fed a 45% HF for 4 weeks; metabolic phenotype, food intake, and energy expenditure were measured. Absorption and storage of nutrients were investigated in the refed state. Results: After 4 weeks of HF feeding, KO mice gained less body weight and fat mass that WT controls, but this was not due to differences in food intake or energy expenditure. KO mice had reduced expression of carbohydrate transporters and absorption of carbohydrate in the jejunum. KO mice had fewer hepatic lipid droplets and decreased expression of de novo lipogenesis-associated enzymes and lipoproteins for endogenous lipoprotein pathway in liver, suggesting decreased long-term storage of carbohydrate in KO mice. Conclusions: Impairment of leptin signaling in VAN reduces responsiveness to gastrointestinal signals, which reduces intestinal absorption of carbohydrates and de novo lipogenesis resulting in reduced long-term energy storage. This study reveals a novel role of vagal afferents to support digestion and energy storage that may contribute to the effectiveness of vagal blockade to induce weight loss.
Collapse
Affiliation(s)
- Kuei-Pin Huang
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Michael L Goodson
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Wendie Vang
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Hui Li
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Amanda J Page
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Helen E Raybould
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA.
| |
Collapse
|
23
|
Determining the contribution of a high-fructose corn syrup formulation to hepatic glycogen synthesis during ad-libitum feeding in mice. Sci Rep 2020; 10:12852. [PMID: 32733017 PMCID: PMC7393509 DOI: 10.1038/s41598-020-69820-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/06/2020] [Indexed: 11/28/2022] Open
Abstract
Excessive sugar intake including high-fructose corn syrup (HFCS) is implicated in the rise of obesity, insulin resistance and non-alcoholic fatty liver disease. Liver glycogen synthesis is influenced by both fructose and insulin signaling. Therefore, the effect of HFCS on hepatic glycogenesis was evaluated in mice feeding ad-libitum. Using deuterated water: the fraction of glycogen derived from triose-P sources, Krebs cycle substrates, and direct pathway + cycling, was measured in 9 normal-chow fed mice (NC) and 12 mice fed normal chow plus a 55% fructose/45% glucose mix in the drinking water at 30% w/v (HFCS-55). This was enriched with [U-13C]fructose or [U-13C]glucose to determine the contribution of each to glycogenesis. For NC, direct pathway + cycling, Krebs cycle, and triose-P sources accounted for 66 ± 0.7%, 23 ± 0.8% and 11 ± 0.4% of glycogen synthesis, respectively. HFCS-55 mice had similar direct pathway + cycling (64 ± 1%) but lower Krebs cycle (12 ± 1%, p < 0.001) and higher triose-P contributions (24 ± 1%, p < 0.001). HFCS-55-fructose contributed 17 ± 1% via triose-P and 2 ± 0% via Krebs cycle. HFCS-55-glucose contributed 16 ± 3% via direct pathway and 1 ± 0% via Krebs cycle. In conclusion, HFCS-55 supplementation resulted in similar hepatic glycogen deposition rates. Indirect pathway contributions shifted from Krebs cycle to Triose-P sources reflecting HFCS-55-fructose utilization, while HFCS-55-glucose was incorporated almost exclusively by the direct pathway.
Collapse
|
24
|
López-Soldado I, Bertini A, Adrover A, Duran J, Guinovart JJ. Maintenance of liver glycogen during long-term fasting preserves energy state in mice. FEBS Lett 2020; 594:1698-1710. [PMID: 32159852 DOI: 10.1002/1873-3468.13770] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/20/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Abstract
Glycogen shortage during fasting coincides with dramatic changes in hepatic adenine nucleotide levels. The aim of this work was to study the relevance of liver glycogen in the regulation of the hepatic energy state during food deprivation. To this end, we examined the response of mice with sustained increased liver glycogen content to prolonged fasting. In order to increase hepatic glycogen content, we generated mice that overexpress protein targeting to glycogen (PTG) in the liver (PTGOE mice). Control and PTGOE mice were fed ad libitum or fasted for 36 h. Upon fasting, PTGOE mice retained significant hepatic glycogen stores and maintained hepatic energy status. Furthermore, we show that liver glycogen controls insulin sensitivity, gluconeogenesis, lipid metabolism, and ketogenesis upon nutrient deprivation.
Collapse
Affiliation(s)
- Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Angelo Bertini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain
| | - Anna Adrover
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain.,Department of Biochemistry and Molecular Biomedicine, University of Barcelona, Spain
| |
Collapse
|
25
|
Hashimoto N, Wakagi M, Ippoushi K, Takano-Ishikawa Y. Involvement of the hepatic branch of the vagus nerve in the regulation of plasma adipokine levels in rats fed a high-fructose diet. J Nutr Biochem 2019; 71:90-97. [DOI: 10.1016/j.jnutbio.2019.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 05/14/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022]
|
26
|
Edinburgh RM, Hengist A, Smith HA, Travers RL, Betts JA, Thompson D, Walhin JP, Wallis GA, Hamilton DL, Stevenson EJ, Tipton KD, Gonzalez JT. Skipping Breakfast Before Exercise Creates a More Negative 24-hour Energy Balance: A Randomized Controlled Trial in Healthy Physically Active Young Men. J Nutr 2019; 149:1326-1334. [PMID: 31321428 PMCID: PMC6675614 DOI: 10.1093/jn/nxz018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/02/2018] [Accepted: 01/23/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND At rest, omission of breakfast lowers daily energy intake, but also lowers energy expenditure, attenuating any effect on energy balance. The effect of breakfast omission on energy balance when exercise is prescribed is unclear. OBJECTIVES The aim of this study was to assess the effect on 24-h energy balance of omitting compared with consuming breakfast prior to exercise. METHODS Twelve healthy physically active young men (age 23 ± 3 y, body mass index 23.6 ± 2.0 kg/m2) completed 3 trials in a randomized order (separated by >1 week): a breakfast of oats and milk (431 kcal; 65 g carbohydrate, 11 g fat, 19 g protein) followed by rest (BR); breakfast before exercise (BE; 60 min cycling at 50 % peak power output); and overnight fasting before exercise (FE). The 24-h energy intake was calculated based on the food consumed for breakfast, followed by an ad libitum lunch, snacks, and dinner. Indirect calorimetry with heart-rate accelerometry was used to measure substrate utilization and 24-h energy expenditure. A [6,6-2H2]glucose infusion was used to investigate tissue-specific carbohydrate utilization. RESULTS The 24-h energy balance was -400 kcal (normalized 95% CI: -230, -571 kcal) for the FE trial; this was significantly lower than both the BR trial (492 kcal; normalized 95% CI: 332, 652 kcal) and the BE trial (7 kcal; normalized 95% CI: -153, 177 kcal; both P < 0.01 compared with FE). Plasma glucose utilization in FE (mainly representing liver glucose utilization) was positively correlated with energy intake compensation at lunch (r = 0.62, P = 0.03), suggesting liver carbohydrate plays a role in postexercise energy-balance regulation. CONCLUSIONS Neither exercise energy expenditure nor restricted energy intake via breakfast omission were completely compensated for postexercise. In healthy men, pre-exercise breakfast omission creates a more negative daily energy balance and could therefore be a useful strategy to induce a short-term energy deficit. This trial was registered at clinicaltrials.gov as NCT02258399.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gareth A Wallis
- School of Sport, Exercise and Rehabilitation, University of Birmingham, Birmingham, UK
| | - D Lee Hamilton
- Physiology, Exercise and Nutrition Research Group, University of Stirling, Stirling, UK
- School of Exercise and Nutrition Sciences, Faculty of Health, Deakin University, Geelong Waurn Ponds, Australia
| | - Emma J Stevenson
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | - Kevin D Tipton
- Physiology, Exercise and Nutrition Research Group, University of Stirling, Stirling, UK
| | | |
Collapse
|
27
|
McKie GL, Medak KD, Knuth CM, Shamshoum H, Townsend LK, Peppler WT, Wright DC. Housing temperature affects the acute and chronic metabolic adaptations to exercise in mice. J Physiol 2019; 597:4581-4600. [PMID: 31297830 DOI: 10.1113/jp278221] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/14/2019] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Mice are commonly housed at room temperatures below their thermoneutral zone meaning they are exposed to chronic thermal stress. Endurance exercise induces browning and mitochondrial biogenesis in white adipose tissue of rodents, but there are conflicting reports of this phenomenon in humans. We hypothesized that the ambient room temperature at which mice are housed could partially explain these discrepant reports between humans and rodents. We housed mice at room temperature or thermoneutrality and studied their physiological responses to acute and chronic exercise. We found that thermoneutral housing altered running behaviour and glucose homeostasis, and further, that exercise-induced markers of mitochondrial biogenesis and the browning of white adipose tissue were reduced in mice housed at thermoneutrality. ABSTRACT Mice are often housed at temperatures below their thermoneutral zone resulting in compensatory increases in thermogenesis. Despite this, many studies report housing mice at room temperature (RT), likely for the convenience of the researchers studying them. As such, the conflicting reports between humans and rodents regarding the ability of exercise to increase mitochondrial and thermogenic markers in white adipose tissue may be explained by the often-overlooked variable, housing temperature. To test this hypothesis, we housed male C57BL/6 mice at RT (22°C) or thermoneutrality (TN) (29°C) with or without access to a voluntary running wheel for 6 weeks or subjected them to an acute exhaustive bout of treadmill running. We examined the gene expression and protein content of select mitochondrial and thermogenic markers in skeletal muscle, epididymal white adipose tissue (eWAT), inguinal white adipose tissue (iWAT) and brown adipose tissue (BAT). We also assessed adipocyte morphology and indices of glucose homeostasis. Housing temperature influenced glucose tolerance and insulin action in vivo, yet the beneficial effects of exercise, both acute and chronic, remained intact in eWAT, BAT and skeletal muscle irrespective of housing temperature. Housing mice at TN led to an attenuation of some of the effects of exercise on iWAT. Collectively, we present data characterizing the acute and chronic metabolic adaptations to exercise at different housing temperatures and demonstrate, for the first time, that temperature influences the ability of exercise to increase markers of mitochondrial biogenesis and the browning of white adipose tissue.
Collapse
Affiliation(s)
- Greg L McKie
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Kyle D Medak
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Carly M Knuth
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Hesham Shamshoum
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Logan K Townsend
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Willem T Peppler
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - David C Wright
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
28
|
Abstract
The objective of this review paper is to evaluate the impact of undertaking aerobic exercise in the overnight-fasted v. fed-state, in the context of optimising the health benefits of regular physical activity. Conducting a single bout of aerobic exercise in the overnight-fasted v. fed-state can differentially modulate the aspects of metabolism and energy balance behaviours. This includes, but is not limited to, increased utilisation of fat as a fuel source, improved plasma lipid profiles, enhanced activation of molecular signalling pathways related to fuel metabolism in skeletal muscle and adipose tissue, and reductions in energy intake over the course of a day. The impact of a single bout of overnight-fasted v. fed-state exercise on short-term glycaemic control is variable, being affected by the experimental conditions, the time frame of measurement and possibly the subject population studied. The health response to undertaking overnight-fasted v. fed-state exercise for a sustained period of time in the form of exercise training is less clear, due to a limited number of studies. From the extant literature, there is evidence that overnight-fasted exercise in young, healthy men can enhance training-induced adaptations in skeletal muscle metabolic profile, and mitigate against the negative consequences of short-term excess energy intake on glucose tolerance compared with exercising in the fed-state. Nonetheless, further long-term studies are required, particularly in populations at-risk or living with cardio-metabolic disease to elucidate if feeding status prior to exercise modulates metabolism or energy balance behaviours to an extent that could impact upon the health or therapeutic benefits of exercise.
Collapse
Affiliation(s)
- Gareth A Wallis
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | | |
Collapse
|
29
|
Hu Z, Li E, Sullivan MA, Tan X, Deng B, Gilbert RG, Li C. Glycogen structure in type 1 diabetic mice: Towards understanding the origin of diabetic glycogen molecular fragility. Int J Biol Macromol 2019; 128:665-672. [PMID: 30708007 DOI: 10.1016/j.ijbiomac.2019.01.186] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 12/31/2018] [Accepted: 01/28/2019] [Indexed: 12/28/2022]
Abstract
Glycogen is a complex branched glucose polymer. Liver glycogen in db/db mouse, a type-2 diabetic mouse model, has been found to be more molecularly fragile than in healthy mice. Size-exclusion chromatography was employed in this study to investigate the molecular structure of liver glycogen in two types of type 1 diabetic mouse models (NOD and C57BL/6J mice), sacrificed at various times throughout the diurnal cycle, and the fragility of liver glycogen after exposure to a hydrogen-bond disruptor were tested. Type 1 diabetic mice exhibit a similar glycogen fragility with that observed for db/db mice. This eliminates many of the potential causes for glycogen molecular fragility; the most likely explanation is that it is caused by high blood-glucose level and/or insulin deficiency, both phenotypes being common to both type 1 and type 2 diabetic mice. This result suggests ways towards new drug targets for the management of diabetes.
Collapse
Affiliation(s)
- Zhenxia Hu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Enpeng Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu Province, China
| | - Mitchell A Sullivan
- Glycation and Diabetes, Translational Research Institute, Mater Research Institute-The University of Queensland, Brisbane, QLD 4102, Australia
| | - Xinle Tan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Bin Deng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Robert G Gilbert
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu Province, China; The University of Queensland, Centre for Nutrition and Food Science, Queensland Alliance for Agriculture and Food Innovation, Brisbane, QLD 4072, Australia.
| | - Cheng Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, Jiangsu Province, China.
| |
Collapse
|
30
|
The vagus neurometabolic interface and clinical disease. Int J Obes (Lond) 2018; 42:1101-1111. [PMID: 29795463 DOI: 10.1038/s41366-018-0086-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 01/07/2023]
Abstract
The nervous system both monitors and modulates body metabolism to maintain homoeostasis. In disease states such as obesity and diabetes, the neurometabolic interface is dysfunctional and contributes to clinical illness. The vagus nerve, in particular, with both sensory and motor fibres, provides an anatomical substrate for this interface. Its sensory fibres contain receptors for important circulating metabolic mediators, including leptin and cholecystokinin, and provide real-time information about these mediators to the central nervous system. In turn, efferent fibres within the vagus nerve participate in a brain-gut axis to regulate metabolism. In this review, we describe these vagus nerve-mediated metabolic pathways and recent clinical trials of vagus nerve stimulation for the management of obesity. These early studies suggest that neuromodulation approaches that employ electricity to tune neurometabolic circuits may represent a new tool in the clinical armamentarium directed against obesity.
Collapse
|
31
|
Li T, Ni L, Zhao Z, Liu X, Lai Z, Di X, Xie Z, Song X, Wang X, Zhang R, Liu C. Melatonin attenuates smoking-induced hyperglycemia via preserving insulin secretion and hepatic glycogen synthesis in rats. J Pineal Res 2018; 64:e12475. [PMID: 29437243 PMCID: PMC5947659 DOI: 10.1111/jpi.12475] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 02/02/2018] [Indexed: 12/11/2022]
Abstract
Epidemiology survey indicated that cigarette smoking is a risk factor of diabetes. However, the precise mechanisms remain to be clarified. In this study, we found that smoking caused metabolic malfunctions on pancreas and liver in experimental animal model. These were indicated by hyperglycemia, increased serum hemoglobin A1c level and decreased insulin secretion, inhibition of liver glycogen synthase (LGS), and hepatic glycogen synthesis. Mechanistic studies revealed that all these alterations were caused by the inflammatory reaction and reactive oxygen species (ROS) induced by the smoking. Melatonin treatment significantly preserved the functions of both pancreas and liver by reducing β cell apoptosis, CD68-cell infiltration, ROS production, and caspase-3 expression. The siRNA-knockdown model identified that the protective effects of melatonin were mediated by melatonin receptor-2 (MT2). This study uncovered potentially underlying mechanisms related to the association between smoking and diabetes. In addition, it is, for first time, to report that melatonin effectively protects against smoking-induced glucose metabolic alterations and the signal transduction pathway of melatonin is mainly mediated by its MT2 receptor. These observations provide solid evidence for the clinically use of melatonin to reduce smoking-related diabetes, and the therapeutic regimens are absent currently.
Collapse
Affiliation(s)
- Tianjia Li
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Leng Ni
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhewei Zhao
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinnong Liu
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhichao Lai
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiao Di
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhibo Xie
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xitao Song
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xuebin Wang
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Rui Zhang
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changwei Liu
- Department of Vascular SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|