1
|
Neoh GKS, Tan X, Chen S, Roura E, Dong X, Gilbert RG. Glycogen metabolism and structure: A review. Carbohydr Polym 2024; 346:122631. [PMID: 39245499 DOI: 10.1016/j.carbpol.2024.122631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024]
Abstract
Glycogen is a glucose polymer that plays a crucial role in glucose homeostasis by functioning as a short-term energy storage reservoir in animals and bacteria. Abnormalities in its metabolism and structure can cause several problems, including diabetes, glycogen storage diseases (GSDs) and muscular disorders. Defects in the enzymes involved in glycogen synthesis or breakdown, resulting in either excessive accumulation or insufficient availability of glycogen in cells seem to account for the most common pathogenesis. This review discusses glycogen metabolism and structure, including molecular architecture, branching dynamics, and the role of associated components within the granules. The review also discusses GSD type XV and Lafora disease, illustrating the broader implications of aberrant glycogen metabolism and structure. These conditions also impart information on important regulatory mechanisms of glycogen, which hint at potential therapeutic targets. Knowledge gaps and potential future research directions are identified.
Collapse
Affiliation(s)
- Galex K S Neoh
- School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Xinle Tan
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia; Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Si Chen
- School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Eugeni Roura
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Xin Dong
- School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Robert G Gilbert
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia; Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Key Laboratory of Plant Functional Genomics of the Ministry of Education/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
2
|
Colpaert M, Singh PK, Donohue KJ, Pires NT, Fuller DD, Corti M, Byrne BJ, Sun RC, Vander Kooi CW, Gentry MS. Neurological glycogen storage diseases and emerging therapeutics. Neurotherapeutics 2024; 21:e00446. [PMID: 39277505 DOI: 10.1016/j.neurot.2024.e00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
Glycogen storage diseases (GSDs) comprise a group of inherited metabolic disorders characterized by defects in glycogen metabolism, leading to abnormal glycogen accumulation in multiple tissues, most notably affecting the liver, skeletal muscle, and heart. Recent findings have uncovered the importance of glycogen metabolism in the brain, sustaining a myriad of physiological functions and linking its perturbation to central nervous system (CNS) pathology. This link resulted in classification of neurological-GSDs (n-GSDs), a group of diseases with shared deficits in neurological glycogen metabolism. The n-GSD patients exhibit a spectrum of clinical presentations with common etiology while requiring tailored therapeutic approaches from the traditional GSDs. Recent research has elucidated the genetic and biochemical mechanisms and pathophysiological basis underlying different n-GSDs. Further, the last decade has witnessed some promising developments in novel therapeutic approaches, including enzyme replacement therapy (ERT), substrate reduction therapy (SRT), small molecule drugs, and gene therapy targeting key aspects of glycogen metabolism in specific n-GSDs. This preclinical progress has generated noticeable success in potentially modifying disease course and improving clinical outcomes in patients. Herein, we provide an overview of current perspectives on n-GSDs, emphasizing recent advances in understanding their molecular basis, therapeutic developments, underscore key challenges and the need to deepen our understanding of n-GSDs pathogenesis to develop better therapeutic strategies that could offer improved treatment and sustainable benefits to the patients.
Collapse
Affiliation(s)
- Matthieu Colpaert
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | | | | | - David D Fuller
- Department of Physical Therapy and Breathing Research and Therapeutics Center, University of Florida, Gainesville, FL, USA
| | - Manuela Corti
- Department of Pediatrics, Powell Gene Therapy Center, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Barry J Byrne
- Department of Pediatrics, Powell Gene Therapy Center, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ramon C Sun
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research (CASBR), University of Florida, Gainesville, FL, USA
| | - Craig W Vander Kooi
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research (CASBR), University of Florida, Gainesville, FL, USA
| | - Matthew S Gentry
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research (CASBR), University of Florida, Gainesville, FL, USA.
| |
Collapse
|
3
|
Liu X, Gilbert RG. Normal and abnormal glycogen structure - A review. Carbohydr Polym 2024; 338:122195. [PMID: 38763710 DOI: 10.1016/j.carbpol.2024.122195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/21/2024]
Abstract
Glycogen, a complex branched glucose polymer, is found in animals and bacteria, where it serves as an energy storage molecule. It has linear (1 → 4)-α glycosidic bonds between anhydroglucose monomer units, with branch points connected by (1 → 6)-α bonds. Individual glycogen molecules are referred to as β particles. In organs like the liver and heart, these β particles can bind into larger aggregate α particles, which exhibit a rosette-like morphology. The mechanisms and bonding underlying the aggregation process are not fully understood. For example, mammalian liver glycogen has been observed to be molecularly fragile under certain conditions, such as glycogen from diabetic livers fragmenting when exposed to dimethyl sulfoxide (DMSO), while glycogen from healthy livers is much less fragile; this indicates some difference, as yet unknown, in the bonding between β particles in healthy and diabetic glycogen. This fragility may have implications for blood sugar regulation, especially in pathological conditions such as diabetes.
Collapse
Affiliation(s)
- Xin Liu
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Zhongshan Biological Breeding Laboratory, and Jiangsu Key Laboratory of Crop Genetics and Physiology, College of Agriculture, Yangzhou University, Yangzhou 225009, China; Centre for Nutrition & Food Sciences, Queensland Alliance for Agriculture & Food Innovations (QAAFI), The University of Queensland, QLD 4072, Australia
| | - Robert G Gilbert
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Zhongshan Biological Breeding Laboratory, and Jiangsu Key Laboratory of Crop Genetics and Physiology, College of Agriculture, Yangzhou University, Yangzhou 225009, China; Centre for Nutrition & Food Sciences, Queensland Alliance for Agriculture & Food Innovations (QAAFI), The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
4
|
Mishra K, Sweetat S, Baraghithy S, Sprecher U, Marisat M, Bastu S, Glickstein H, Tam J, Rosenmann H, Weil M, Malfatti E, Kakhlon O. The Autophagic Activator GHF-201 Can Alleviate Pathology in a Mouse Model and in Patient Fibroblasts of Type III Glycogenosis. Biomolecules 2024; 14:893. [PMID: 39199279 PMCID: PMC11352067 DOI: 10.3390/biom14080893] [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: 06/05/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Glycogen storage disease type III (GSDIII) is a hereditary glycogenosis caused by deficiency of the glycogen debranching enzyme (GDE), an enzyme, encoded by Agl, enabling glycogen degradation by catalyzing alpha-1,4-oligosaccharide side chain transfer and alpha-1,6-glucose cleavage. GDE deficiency causes accumulation of phosphorylase-limited dextrin, leading to liver disorder followed by fatal myopathy. Here, we tested the capacity of the new autophagosomal activator GHF-201 to alleviate disease burden by clearing pathogenic glycogen surcharge in the GSDIII mouse model Agl-/-. We used open field, grip strength, and rotarod tests for evaluating GHF-201's effects on locomotion, a biochemistry panel to quantify hematological biomarkers, indirect calorimetry to quantify in vivo metabolism, transmission electron microscopy to quantify glycogen in muscle, and fibroblast image analysis to determine cellular features affected by GHF-201. GHF-201 was able to improve all locomotion parameters and partially reversed hypoglycemia, hyperlipidemia and liver and muscle malfunction in Agl-/- mice. Treated mice burnt carbohydrates more efficiently and showed significant improvement of aberrant ultrastructural muscle features. In GSDIII patient fibroblasts, GHF-201 restored mitochondrial membrane polarization and corrected lysosomal swelling. In conclusion, GHF-201 is a viable candidate for treating GSDIII as it recovered a wide range of its pathologies in vivo, in vitro, and ex vivo.
Collapse
Affiliation(s)
- Kumudesh Mishra
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (K.M.); (S.S.); (H.R.)
| | - Sahar Sweetat
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (K.M.); (S.S.); (H.R.)
| | - Saja Baraghithy
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (J.T.)
| | - Uri Sprecher
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 6997801, Israel; (U.S.); (M.M.); (M.W.)
| | - Monzer Marisat
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 6997801, Israel; (U.S.); (M.M.); (M.W.)
| | - Sultan Bastu
- Centre de Reference de Maladies Neuromusculaires, UPEC—Paris Est University, IMRB INSERM U955, Team Biology of the Neuromuscular System, Faculty of Medicine, APHP Hopital Henri Mondor, 1 Rue Gustave Eiffel, 94010 Creteil, France; (S.B.); (E.M.)
| | - Hava Glickstein
- Electron Microscopy Unit, The Hebrew University-Hadassah Medical School, Ein Kerem, Jerusalem 9112001, Israel;
| | - Joseph Tam
- Obesity and Metabolism Laboratory, Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (S.B.); (J.T.)
| | - Hanna Rosenmann
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (K.M.); (S.S.); (H.R.)
| | - Miguel Weil
- The Shmunis School of Biomedicine and Cancer Research, The George S. Wise Faculty for Life Sciences, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 6997801, Israel; (U.S.); (M.M.); (M.W.)
| | - Edoardo Malfatti
- Centre de Reference de Maladies Neuromusculaires, UPEC—Paris Est University, IMRB INSERM U955, Team Biology of the Neuromuscular System, Faculty of Medicine, APHP Hopital Henri Mondor, 1 Rue Gustave Eiffel, 94010 Creteil, France; (S.B.); (E.M.)
| | - Or Kakhlon
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (K.M.); (S.S.); (H.R.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| |
Collapse
|
5
|
Zhong C, Nidetzky B. Bottom-Up Synthesized Glucan Materials: Opportunities from Applied Biocatalysis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400436. [PMID: 38514194 DOI: 10.1002/adma.202400436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/05/2024] [Indexed: 03/23/2024]
Abstract
Linear d-glucans are natural polysaccharides of simple chemical structure. They are comprised of d-glucosyl units linked by a single type of glycosidic bond. Noncovalent interactions within, and between, the d-glucan chains give rise to a broad variety of macromolecular nanostructures that can assemble into crystalline-organized materials of tunable morphology. Structure design and functionalization of d-glucans for diverse material applications largely relies on top-down processing and chemical derivatization of naturally derived starting materials. The top-down approach encounters critical limitations in efficiency, selectivity, and flexibility. Bottom-up approaches of d-glucan synthesis offer different, and often more precise, ways of polymer structure control and provide means of functional diversification widely inaccessible to top-down routes of polysaccharide material processing. Here the natural and engineered enzymes (glycosyltransferases, glycoside hydrolases and phosphorylases, glycosynthases) for d-glucan polymerization are described and the use of applied biocatalysis for the bottom-up assembly of specific d-glucan structures is shown. Advanced material applications of the resulting polymeric products are further shown and their important role in the development of sustainable macromolecular materials in a bio-based circular economy is discussed.
Collapse
Affiliation(s)
- Chao Zhong
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, Graz, 8010, Austria
| | - Bernd Nidetzky
- Institute of Biotechnology and Biochemical Engineering, Graz University of Technology, NAWI Graz, Petersgasse 12, Graz, 8010, Austria
- Austrian Centre of Industrial Biotechnology (acib), Krenngasse 37, Graz, 8010, Austria
| |
Collapse
|
6
|
Isoda T, Takeda E, Hosokawa S, Hotta-Ren S, Ohsumi Y. Atg45 is an autophagy receptor for glycogen, a non-preferred cargo of bulk autophagy in yeast. iScience 2024; 27:109810. [PMID: 38832010 PMCID: PMC11145338 DOI: 10.1016/j.isci.2024.109810] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/03/2023] [Accepted: 04/22/2024] [Indexed: 06/05/2024] Open
Abstract
The mechanisms governing autophagy of proteins and organelles have been well studied, but how other cytoplasmic components such as RNA and polysaccharides are degraded remains largely unknown. In this study, we examine autophagy of glycogen, a storage form of glucose. We find that cells accumulate glycogen in the cytoplasm during nitrogen starvation and that this carbohydrate is rarely observed within autophagosomes and autophagic bodies. However, sequestration of glycogen by autophagy is observed following prolonged nitrogen starvation. We identify a yet-uncharacterized open reading frame, Yil024c (herein Atg45), as encoding a cytosolic receptor protein that mediates autophagy of glycogen (glycophagy). Furthermore, we show that, during sporulation, Atg45 is highly expressed and is associated with an increase in glycophagy. Our results suggest that cells regulate glycophagic activity by controlling the expression level of Atg45.
Collapse
Affiliation(s)
- Takahiro Isoda
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
- School and Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama 226-8503, Japan
- Frontier Research Center, POLA Chemical Industries, Inc, Yokohama 244-0812, Japan
| | - Eigo Takeda
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Sachiko Hosokawa
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Shukun Hotta-Ren
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Yoshinori Ohsumi
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| |
Collapse
|
7
|
Visuttijai K, Hedberg-Oldfors C, Costello DJ, Bermingham N, Oldfors A. Proteomic profiling of polyglucosan bodies associated with glycogenin-1 deficiency in skeletal muscle. Neuropathol Appl Neurobiol 2024; 50:e12995. [PMID: 38923610 DOI: 10.1111/nan.12995] [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: 01/29/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
AIMS Polyglucosan storage disorders represent an emerging field within neurodegenerative and neuromuscular conditions, including Lafora disease (EPM2A, EPM2B), adult polyglucosan body disease (APBD, GBE1), polyglucosan body myopathies associated with RBCK1 deficiency (PGBM1, RBCK1) or glycogenin-1 deficiency (PGBM2, GYG1). While the storage material primarily comprises glycans, this study aimed to gain deeper insights into the protein components by proteomic profiling of the storage material in glycogenin-1 deficiency. METHODS We employed molecular genetic analyses, quantitative mass spectrometry of laser micro-dissected polyglucosan bodies and muscle homogenate, immunohistochemistry and western blot analyses in muscle tissue from a 45-year-old patient with proximal muscle weakness from late teenage years due to polyglucosan storage myopathy. RESULTS The muscle tissue exhibited a complete absence of glycogenin-1 due to a novel homozygous deep intronic variant in GYG1 (c.7+992T>G), introducing a pseudo-exon causing frameshift and a premature stop codon. Accumulated proteins in the polyglucosan bodies constituted components of glycogen metabolism, protein quality control pathways and desmin. Muscle fibres containing polyglucosan bodies frequently exhibited depletion of normal glycogen. CONCLUSIONS The absence of glycogenin-1, a protein important for glycogen synthesis initiation, causes storage of polyglucosan that displays accumulation of several proteins, including those essential for glycogen synthesis, sequestosome 1/p62 and desmin, mirroring findings in RBCK1 deficiency. These results suggest shared pathogenic pathways across different diseases exhibiting polyglucosan storage. Such insights have implications for therapy in these rare yet devastating and presently untreatable disorders.
Collapse
Affiliation(s)
- Kittichate Visuttijai
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Carola Hedberg-Oldfors
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Daniel J Costello
- Department of Neurology, Cork University Hospital and College of Medicine and Health, University College Cork, Cork, Ireland
| | - Niamh Bermingham
- Department of Neuropathology, Cork University Hospital, Cork, Ireland
| | - Anders Oldfors
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Zhang N, Liu F, Zhao Y, Sun X, Wen B, Lu JQ, Yan C, Li D. Defect in degradation of glycogenin-exposed residual glycogen in lysosomes is the fundamental pathomechanism of Pompe disease. J Pathol 2024; 263:8-21. [PMID: 38332735 DOI: 10.1002/path.6255] [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: 05/13/2023] [Revised: 11/27/2023] [Accepted: 12/18/2023] [Indexed: 02/10/2024]
Abstract
Pompe disease is a lysosomal storage disorder that preferentially affects muscles, and it is caused by GAA mutation coding acid alpha-glucosidase in lysosome and glycophagy deficiency. While the initial pathology of Pompe disease is glycogen accumulation in lysosomes, the special role of the lysosomal pathway in glycogen degradation is not fully understood. Hence, we investigated the characteristics of accumulated glycogen and the mechanism underlying glycophagy disturbance in Pompe disease. Skeletal muscle specimens were obtained from the affected sites of patients and mouse models with Pompe disease. Histological analysis, immunoblot analysis, immunofluorescence assay, and lysosome isolation were utilized to analyze the characteristics of accumulated glycogen. Cell culture, lentiviral infection, and the CRISPR/Cas9 approach were utilized to investigate the regulation of glycophagy accumulation. We demonstrated residual glycogen, which was distinguishable from mature glycogen by exposed glycogenin and more α-amylase resistance, accumulated in the skeletal muscle of Pompe disease. Lysosome isolation revealed glycogen-free glycogenin in wild type mouse lysosomes and variously sized glycogenin in Gaa-/- mouse lysosomes. Our study identified that a defect in the degradation of glycogenin-exposed residual glycogen in lysosomes was the fundamental pathological mechanism of Pompe disease. Meanwhile, glycogenin-exposed residual glycogen was absent in other glycogen storage diseases caused by cytoplasmic glycogenolysis deficiencies. In vitro, the generation of residual glycogen resulted from cytoplasmic glycogenolysis. Notably, the inhibition of glycogen phosphorylase led to a reduction in glycogenin-exposed residual glycogen and glycophagy accumulations in cellular models of Pompe disease. Therefore, the lysosomal hydrolysis pathway played a crucial role in the degradation of residual glycogen into glycogenin, which took place in tandem with cytoplasmic glycogenolysis. These findings may offer a novel substrate reduction therapeutic strategy for Pompe disease. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Na Zhang
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, PR China
| | - Fuchen Liu
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, PR China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Yuying Zhao
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, PR China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Xiaohan Sun
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, PR China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Bing Wen
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, PR China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Jian-Qiang Lu
- Department of Pathology and Molecular Medicine, Division of Neuropathology, McMaster University, Hamilton, Ontario, Canada
| | - Chuanzhu Yan
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, PR China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, PR China
- Qingdao Key Laboratory of Rare Diseases, Qilu Hospital (Qingdao) of Shandong University, Qingdao, PR China
| | - Duoling Li
- Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital, Shandong University, Jinan, PR China
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, PR China
| |
Collapse
|
9
|
Liu X, Zhang P, Gilbert RG. Formation mechanism of α particles in glycogen: Testing the budding hypothesis by Monte-Carlo simulation. Int J Biol Macromol 2024; 263:130332. [PMID: 38401580 DOI: 10.1016/j.ijbiomac.2024.130332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
Glycogen, a complex branched glucose polymer and a blood-sugar reservoir in animals, comprises small β particles joined together into composite α particles. In diabetic animals, α particles fragment more easily than those in healthy animals. Finding evidence for or against postulated mechanisms for α-particle formation is thus important for diabetes research. Insight into this is obtained here using Monte-Carlo simulations, including addition and loss of glucose monomer, branching and debranching, based on earlier simulations which were in acceptable agreement with experiment [Zhang et al., Int J Biol Macromolecules 2018, 116, 264]. One postulated mechanism for α-particle formation is "budding": occasionally a glucan chain temporarily protrudes from the particle, and if its growing end is sufficiently far from its parent particle, it propagates to a new linked particle. We tested this by simulations in which an "artificial" bud (a chain extending well outside the average particle radius) is added to a glycogen molecule in a dynamic steady state, and the system allowed to evolve. In some simulations, the particle reached a new steady state having an irregular dumbbell shape: a rudimentary α particle. Thus 'budding' is a possible mechanism for α particles to form. If no simulations had shown this behaviour, it would have refuted the postulate.
Collapse
Affiliation(s)
- Xin Liu
- Jiangsu Key Laboratory of Crop Genetics and Physiology, Key Laboratory of Plant Functional Genomics of the Ministry of Education, College of Agriculture, Yangzhou University, Yangzhou 225009, Jiangsu Province, China; Co-Innovation Centre for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou 225009, Jiangsu Province, China; Centre for Nutrition & Food Sciences, Queensland Alliance for Agriculture & Food Innovations (QAAFI), The University of Queensland, QLD 4072, Australia
| | - Peng Zhang
- School of Electronic Engineering, Tongling University, Tongling 244061, PR China
| | - Robert G Gilbert
- Jiangsu Key Laboratory of Crop Genetics and Physiology, Key Laboratory of Plant Functional Genomics of the Ministry of Education, College of Agriculture, Yangzhou University, Yangzhou 225009, Jiangsu Province, China; Co-Innovation Centre for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou 225009, Jiangsu Province, China; Centre for Nutrition & Food Sciences, Queensland Alliance for Agriculture & Food Innovations (QAAFI), The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
10
|
Koeberl DD, Koch RL, Lim JA, Brooks ED, Arnson BD, Sun B, Kishnani PS. Gene therapy for glycogen storage diseases. J Inherit Metab Dis 2024; 47:93-118. [PMID: 37421310 PMCID: PMC10874648 DOI: 10.1002/jimd.12654] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/24/2023] [Accepted: 07/05/2023] [Indexed: 07/10/2023]
Abstract
Glycogen storage disorders (GSDs) are inherited disorders of metabolism resulting from the deficiency of individual enzymes involved in the synthesis, transport, and degradation of glycogen. This literature review summarizes the development of gene therapy for the GSDs. The abnormal accumulation of glycogen and deficiency of glucose production in GSDs lead to unique symptoms based upon the enzyme step and tissues involved, such as liver and kidney involvement associated with severe hypoglycemia during fasting and the risk of long-term complications including hepatic adenoma/carcinoma and end stage kidney disease in GSD Ia from glucose-6-phosphatase deficiency, and cardiac/skeletal/smooth muscle involvement associated with myopathy +/- cardiomyopathy and the risk for cardiorespiratory failure in Pompe disease. These symptoms are present to a variable degree in animal models for the GSDs, which have been utilized to evaluate new therapies including gene therapy and genome editing. Gene therapy for Pompe disease and GSD Ia has progressed to Phase I and Phase III clinical trials, respectively, and are evaluating the safety and bioactivity of adeno-associated virus vectors. Clinical research to understand the natural history and progression of the GSDs provides invaluable outcome measures that serve as endpoints to evaluate benefits in clinical trials. While promising, gene therapy and genome editing face challenges with regard to clinical implementation, including immune responses and toxicities that have been revealed during clinical trials of gene therapy that are underway. Gene therapy for the glycogen storage diseases is under development, addressing an unmet need for specific, stable therapy for these conditions.
Collapse
Affiliation(s)
- Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Rebecca L Koch
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Jeong-A Lim
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Elizabeth D Brooks
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Benjamin D Arnson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Baodong Sun
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
| | - Priya S Kishnani
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical School, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
11
|
Zhao H, Wong RJ, Stevenson DK. The placental vasculature is affected by changes in gene expression and glycogen-rich cells in a diet-induced obesity mouse model. PLoS One 2023; 18:e0294185. [PMID: 37948457 PMCID: PMC10637699 DOI: 10.1371/journal.pone.0294185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
Maternal obesity is a risk factor for pregnancy complications. Obesity caused by a high-fat diet (HFD) may alter maternal glucose/glycogen metabolism. Here, our objective was to investigate whether the placental vasculature is altered via changes in gene expression and glycogen-rich cells using a preclinical mouse model of diet-induced obesity. We subjected female FVB/N mice to one of three feeding regimens: regular chow (RC) given at preconception and during pregnancy (Control); RC given at preconception and then a HFD during pregnancy (HFD-P); or HFD initiated 4 weeks preconception and during pregnancy (HFD-PreCP). Daily food consumption and weekly maternal weights were recorded. Maternal blood glucose levels were measured at preconception and 4 gestational epochs (E6.5-E9.5, E10.5-E12.5, E13.5-E15.5, E16.5-E19.5). At E8.5-E16.5, total RNA in placentas were isolated for gene expression analyses. Placentas were also collected for HE and periodic acid Schiff's (PAS) staining and glycogen content assays. Dams in the HFD-P and HFD-PreCP groups gained significantly more weight than controls. Pre- and antenatal glucose levels were also significantly higher (15%-30%) in HFD-PreCP dams. Expression of several placental genes were also altered in HFD dams compared with controls. Consumption of the HFD also led to phenotypic and morphologic changes in glycogen trophoblasts (GlyTs) and uterine natural killer (uNK) cells. Alterations in vascularity were also observed in the labyrinth of HFD-PreCP placentas, which correlated with decreased placental efficiency. Overall, we observed that a HFD induces gestational obesity in mice, alters expression of placental genes, affects glucose homeostasis, and alters glycogen-positive GlyTs and uNK cells. All these changes may lead to impaired placental vascular development, and thus heighten the risk for pregnancy complications.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Ronald J. Wong
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - David K. Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| |
Collapse
|
12
|
Broadwin M, Harris DD, Sabe SA, Sengun E, Sylvestre AJ, Alexandrov BS, Selke FW, Usheva A. Impaired cardiac glycolysis and glycogen depletion are linked to poor myocardial outcomes in juvenile male swine with metabolic syndrome and ischemia. Physiol Rep 2023; 11:e15742. [PMID: 37537137 PMCID: PMC10400405 DOI: 10.14814/phy2.15742] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 08/05/2023] Open
Abstract
Obesity continues to rise in the juveniles and obese children are more likely to develop metabolic syndrome (MetS) and related cardiovascular disease. Unfortunately, effective prevention and long-term treatment options remain limited. We determined the juvenile cardiac response to MetS in a swine model. Juvenile male swine were fed either an obesogenic diet, to induce MetS, or a lean diet, as a control (LD). Myocardial ischemia was induced with surgically placed ameroid constrictor on the left circumflex artery. Physiological data were recorded and at 22 weeks of age the animals underwent a terminal harvest procedure and myocardial tissue was extracted for total metabolic and proteomic LC/MS-MS, RNA-seq analysis, and data underwent nonnegative matrix factorization for metabolic signatures. Significantly altered in MetS versus. LD were the glycolysis-related metabolites and enzymes. In MetS compared with LD Glycogen synthase 1 (GYS1)-glycogen phosphorylases (PYGM/PYGL) expression disbalance resulted in a loss of myocardial glycogen. Our findings are consistent with the concept that transcriptionally driven myocardial changes in glycogen and glucose metabolism-related enzymes lead to a deficiency of their metabolite products in MetS. This abnormal energy metabolism provides insight into the pathogenesis of the juvenile heart in MetS. This study reveals that MetS and ischemia diminishes ATP availability in the myocardium via altering the glucose-G6P-pyruvate axis at the level of metabolites and gene expression of related enzymes. The observed severe glycogen depletion in MetS coincides with disbalance in expression of GYS1 and both PYGM and PYGL. This altered energy substrate metabolism is a potential target of pharmacological agents for improving juvenile myocardial function in MetS and ischemia.
Collapse
Affiliation(s)
- Mark Broadwin
- Division of Cardiothoracic Surgery, Department of SurgeryWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Dwight D. Harris
- Division of Cardiothoracic Surgery, Department of SurgeryWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Sharif A. Sabe
- Division of Cardiothoracic Surgery, Department of SurgeryWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Elif Sengun
- Division of Cardiology, Department of MedicineWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Amber J. Sylvestre
- Division of Cardiothoracic Surgery, Department of SurgeryWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | | | - Frank W. Selke
- Division of Cardiothoracic Surgery, Department of SurgeryWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| | - Anny Usheva
- Division of Cardiothoracic Surgery, Department of SurgeryWarren Alpert Medical School of Brown UniversityProvidenceRhode IslandUSA
| |
Collapse
|
13
|
Abstract
The biophysical response of the human body to electric current is widely appreciated as a barometer of fluid distribution and cell function. From distinct raw bioelectrical impedance (BIA) variables assessed in the field of body composition, phase angle (PhA) has been repeatedly indicated as a functional marker of the cell's health and mass. Although resistance training (RT) programs have demonstrated to be effective to improve PhA, with varying degrees of change depending on other raw BIA variables, there is still limited research explaining the biological mechanisms behind these changes. Here, we aim to provide the rationale for the responsiveness of PhA determinants to RT, as well as to summarize all available evidence addressing the effect of varied RT programs on PhA of different age groups. Available data led us to conclude that RT modulates the cell volume by increasing the levels of intracellular glycogen and water, thus triggering structural and functional changes in different cell organelles. These alterations lead, respectively, to shifts in the resistive path of the electric current (resistance, R) and capacitive properties of the human body (reactance, Xc), which ultimately impact PhA, considering that it is the angular transformation of the ratio between Xc and R. Evidence drawn from experimental research suggests that RT is highly effective for enhancing PhA, especially when adopting high-intensity, volume, and duration RT programs combining other types of exercise. Still, additional research exploring the effects of RT on whole-body and regional BIA variables of alternative population groups is recommended for further knowledge development.
Collapse
Affiliation(s)
- Luís B Sardinha
- Exercise and Health Laboratory, Faculdade de Motricidade Humana, CIPER, Universidade de Lisboa, , Cruz Quebrada, Portugal.
| | - Gil B Rosa
- Exercise and Health Laboratory, Faculdade de Motricidade Humana, CIPER, Universidade de Lisboa, , Cruz Quebrada, Portugal
| |
Collapse
|
14
|
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
|
15
|
Wu SH, Lu IC, Yang SM, Hsieh CF, Chai CY, Tai MH, Huang SH. Spinal Irisin Gene Delivery Attenuates Burn Injury-Induced Muscle Atrophy by Promoting Axonal Myelination and Innervation of Neuromuscular Junctions. Int J Mol Sci 2022; 23:ijms232415899. [PMID: 36555538 PMCID: PMC9784798 DOI: 10.3390/ijms232415899] [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: 11/23/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Muscle loss and weakness after a burn injury are typically the consequences of neuronal dysregulation and metabolic change. Hypermetabolism has been noted to cause muscle atrophy. However, the mechanism underlying the development of burn-induced motor neuropathy and its contribution to muscle atrophy warrant elucidation. Current therapeutic interventions for burn-induced motor neuropathy demonstrate moderate efficacy and have side effects, which limit their usage. We previously used a third-degree burn injury rodent model and found that irisin-an exercise-induced myokine-exerts a protective effect against burn injury-induced sensory and motor neuropathy by attenuating neuronal damage in the spinal cord. In the current study, spinal irisin gene delivery was noted to attenuate burn injury-induced sciatic nerve demyelination and reduction of neuromuscular junction innervation. Spinal overexpression of irisin leads to myelination rehabilitation and muscular innervation through the modulation of brain-derived neurotrophic factor and glial-cell-line-derived neurotrophic factor expression along the sciatic nerve to the muscle tissues and thereby modulates the Akt/mTOR pathway and metabolic derangement and prevents muscle atrophy.
Collapse
Affiliation(s)
- Sheng-Hua Wu
- Department of Anesthesiology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
- Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - I-Cheng Lu
- Department of Anesthesiology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Anesthesiology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
| | - Shih-Ming Yang
- Institute of Biomedical Sciences, National Sun Yat-Sun University, Kaohsiung 804, Taiwan
| | - Chia-Fang Hsieh
- Department of Anesthesiology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
| | - Chee-Yin Chai
- Institute of Biomedical Sciences, National Sun Yat-Sun University, Kaohsiung 804, Taiwan
- Department of Pathology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-Sun University, Kaohsiung 804, Taiwan
- Correspondence: (M.-H.T.); (S.-H.H.)
| | - Shu-Hung Huang
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Surgery, Division of Plastic Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
- Correspondence: (M.-H.T.); (S.-H.H.)
| |
Collapse
|
16
|
Katz A. The role of glycogen phosphorylase in glycogen biogenesis in skeletal muscle after exercise. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 5:29-33. [PMID: 36994178 PMCID: PMC10040329 DOI: 10.1016/j.smhs.2022.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 11/15/2022] Open
Abstract
Initially it was believed that phosphorylase was responsible for both glycogen breakdown and synthesis in the living cell. The discovery of glycogen synthase and McArdle's disease (lack of phosphorylase activity), together with the high Pi/glucose 1-P ratio in skeletal muscle, demonstrated that glycogen synthesis could not be attributed to reversal of the phosphorylase reaction. Rather, glycogen synthesis was attributable solely to the activity of glycogen synthase, subsequent to the transport of glucose into the cell. However, the well-established observation that phosphorylase was inactivated (i.e., dephosphorylated) during the initial recovery period after prior exercise, when the rate of glycogen accumulation is highest and independent of insulin, suggested that phosphorylase could play an active role in glycogen accumulation. But the quantitative contribution of phosphorylase inactivation was not established until recently, when studying isolated murine muscle preparations during recovery from repeated contractions at temperatures ranging from 25 to 35 °C. Thus, in both slow-twitch, oxidative and fast-twitch, glycolytic muscles, inactivation of phosphorylase accounted for 45%-75% of glycogen accumulation during the initial hours of recovery following repeated contractions. Such data indicate that phosphorylase inactivation may be the most important mechanism for glycogen accumulation under defined conditions. These results support the initial belief that phosphorylase plays a quantitative role in glycogen formation in the living cell. However, the mechanism is not via activation of phosphorylase, but rather via inactivation of the enzyme.
Collapse
|
17
|
Fastman NM, Liu Y, Ramanan V, Merritt H, Ambing E, DePaoli-Roach AA, Roach PJ, Hurley TD, Mellem KT, Ullman JC, Green E, Morgans D, Tzitzilonis C. The structural mechanism of human glycogen synthesis by the GYS1-GYG1 complex. Cell Rep 2022; 40:111041. [PMID: 35793618 DOI: 10.1016/j.celrep.2022.111041] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/15/2022] [Accepted: 06/11/2022] [Indexed: 11/03/2022] Open
Abstract
Glycogen is the primary energy reserve in mammals, and dysregulation of glycogen metabolism can result in glycogen storage diseases (GSDs). In muscle, glycogen synthesis is initiated by the enzymes glycogenin-1 (GYG1), which seeds the molecule by autoglucosylation, and glycogen synthase-1 (GYS1), which extends the glycogen chain. Although both enzymes are required for proper glycogen production, the nature of their interaction has been enigmatic. Here, we present the human GYS1:GYG1 complex in multiple conformations representing different functional states. We observe an asymmetric conformation of GYS1 that exposes an interface for close GYG1 association, and propose this state facilitates handoff of the GYG1-associated glycogen chain to a GYS1 subunit for elongation. Full activation of GYS1 widens the GYG1-binding groove, enabling GYG1 release concomitant with glycogen chain growth. This structural mechanism connecting chain nucleation and extension explains the apparent stepwise nature of glycogen synthesis and suggests distinct states to target for GSD-modifying therapeutics.
Collapse
Affiliation(s)
- Nathan M Fastman
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Yuxi Liu
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Vyas Ramanan
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Hanne Merritt
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Eileen Ambing
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Anna A DePaoli-Roach
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46220, USA
| | - Peter J Roach
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46220, USA
| | - Thomas D Hurley
- Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46220, USA
| | - Kevin T Mellem
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Julie C Ullman
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Eric Green
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - David Morgans
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA
| | - Christos Tzitzilonis
- Maze Therapeutics, 171 Oyster Point Blvd, Suite 300, South San Francisco, CA 94080, USA.
| |
Collapse
|
18
|
Zhang L, Guo Q, Duan Y, Wang W, Yang Y, Yin Y, Gong S, Han M, Li F, Yin Y. Potential nutritional healthy-aging strategy: enhanced protein metabolism by balancing branched-chain amino acids in a finishing pig model. Food Funct 2022; 13:6217-6232. [PMID: 35583212 DOI: 10.1039/d1fo03970a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Branched-chain amino acids (BCAAs) have key physiological roles in the regulation of protein synthesis, metabolism, food intake and aging. This study aimed to investigate the protective effect of balanced BCAAs on healthy aging by increasing skeletal muscle mass and muscle fiber composition in a finishing pig model. A balanced BCAA ratio (Leu : Ile : Val = 2 : 2 : 1) significantly activated the mTOR pathway and upregulated the expression of amino acid transporters, such as ASCT2, SNAT2, LAT1, PAT1, and SLC38A9, simultaneously modulating mitochondrial function and muscle fiber composition, thereby inhibiting inflammatory cytokines, such as IL-6 and TNF-α, regulating amino acid metabolism, and ultimately increasing skeletal muscle mass. Overall, our results suggest that a BCAA ratio around 2 : 2 : 1 may be a promising candidate for healthy aging in humans and animals.
Collapse
Affiliation(s)
- Lingyu Zhang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China. .,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.,National Engineering Laboratory for Rice and By-Product Deep Processing, Central South University of Forestry and Technology, Changsha 410004, China
| | - Qiuping Guo
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| | - Yehui Duan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| | - Wenlong Wang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| | - Yuhuan Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yunju Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Saiming Gong
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Mengmeng Han
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China. .,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengna Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China. .,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Changsha 410125, China.
| |
Collapse
|
19
|
Comparative transcriptome analysis of diurnal alterations of liver glycogen structure: A pilot study. Carbohydr Polym 2022; 295:119710. [DOI: 10.1016/j.carbpol.2022.119710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/21/2022] [Accepted: 06/03/2022] [Indexed: 11/20/2022]
|
20
|
A century of exercise physiology: key concepts in regulation of glycogen metabolism in skeletal muscle. Eur J Appl Physiol 2022; 122:1751-1772. [PMID: 35355125 PMCID: PMC9287217 DOI: 10.1007/s00421-022-04935-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/15/2022] [Indexed: 01/20/2023]
Abstract
Glycogen is a branched, glucose polymer and the storage form of glucose in cells. Glycogen has traditionally been viewed as a key substrate for muscle ATP production during conditions of high energy demand and considered to be limiting for work capacity and force generation under defined conditions. Glycogenolysis is catalyzed by phosphorylase, while glycogenesis is catalyzed by glycogen synthase. For many years, it was believed that a primer was required for de novo glycogen synthesis and the protein considered responsible for this process was ultimately discovered and named glycogenin. However, the subsequent observation of glycogen storage in the absence of functional glycogenin raises questions about the true role of the protein. In resting muscle, phosphorylase is generally considered to be present in two forms: non-phosphorylated and inactive (phosphorylase b) and phosphorylated and constitutively active (phosphorylase a). Initially, it was believed that activation of phosphorylase during intense muscle contraction was primarily accounted for by phosphorylation of phosphorylase b (activated by increases in AMP) to a, and that glycogen synthesis during recovery from exercise occurred solely through mechanisms controlled by glucose transport and glycogen synthase. However, it now appears that these views require modifications. Moreover, the traditional roles of glycogen in muscle function have been extended in recent years and in some instances, the original concepts have undergone revision. Thus, despite the extensive amount of knowledge accrued during the past 100 years, several critical questions remain regarding the regulation of glycogen metabolism and its role in living muscle.
Collapse
|
21
|
Oh M, Ha DI, Son C, Kang JG, Hwang H, Moon SB, Kim M, Nam J, Kim JS, Song SY, Kim YS, Park S, Yoo JS, Ko JH, Park K. Defect in cytosolic Neu2 sialidase abrogates lipid metabolism and impairs muscle function in vivo. Sci Rep 2022; 12:3216. [PMID: 35217678 PMCID: PMC8881595 DOI: 10.1038/s41598-022-07033-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/27/2022] [Indexed: 12/25/2022] Open
Abstract
Sialic acid (SA) is present in glycoconjugates and important in cell-cell recognition, cell adhesion, and cell growth and as a receptor. Among the four mammalian sialidases, cytosolic NEU2 has a pivotal role in muscle and neuronal differentiation in vitro. However, its biological functions in vivo remain unclear due to its very low expression in humans. However, the presence of cytoplasmic glycoproteins, gangliosides, and lectins involved in cellular metabolism and glycan recognition has suggested the functional importance of cytosolic Neu2 sialidases. We generated a Neu2 knockout mouse model via CRISPR/Cas9-mediated genome engineering and analyzed the offspring littermates at different ages to investigate the in vivo function of cytosolic Neu2 sialidase. Surprisingly, knocking out the Neu2 gene in vivo abrogated overall lipid metabolism, impairing motor function and leading to diabetes. Consistent with these results, Neu2 knockout led to alterations in sialylated glycoproteins involved in lipid metabolism and muscle function, as shown by glycoproteomics analysis.
Collapse
Affiliation(s)
- Mijung Oh
- Medical Research Institute, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Dae-In Ha
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141, Daejeon, Republic of Korea
| | - Chaeyeon Son
- Medical Research Institute, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Jeong Gu Kang
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141, Daejeon, Republic of Korea
| | - Heeyoun Hwang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Su Bin Moon
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141, Daejeon, Republic of Korea
| | - Minjeong Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Jihae Nam
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141, Daejeon, Republic of Korea
- Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jung Soo Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141, Daejeon, Republic of Korea
| | - Sang Yong Song
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| | - Yong-Sam Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141, Daejeon, Republic of Korea
| | - Sangwoo Park
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Jong Shin Yoo
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Jeong-Heon Ko
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, 34141, Daejeon, Republic of Korea.
- Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.
| | - Kyoungsook Park
- Medical Research Institute, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea.
| |
Collapse
|
22
|
Wan Y, Hu Z, Liu Q, Wang L, Sullivan MA, Gilbert RG. Liver fibrosis alters the molecular structures of hepatic glycogen. Carbohydr Polym 2022; 278:118991. [PMID: 34973794 DOI: 10.1016/j.carbpol.2021.118991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 01/06/2023]
Abstract
Liver fibrosis (LF) leads to liver failure and short survival. Liver glycogen is a hyperbranched glucose polymer, comprising individual β particles, which can bind together to form aggregated α particles. Glycogen functionality depends on its molecular structure. This study compared the molecular structure of liver glycogen from both LF and healthy rats, and explored underlying mechanisms for observed differences. Glycogen from both groups contained α and β particles; the LF group contained a higher proportion of β particles, with the glycogen containing fewer long chains than seen in the control group. Both glycogen branching enzyme and glycogen phosphorylase showed a significant decrease of activity in the LF group. Transcriptomics and proteomics revealed a functional deficiency of mitochondria in the LF group, which may lead to changes in glycogen structure. These results provide for the first time an understanding of how liver fibrosis affects liver glycogen metabolism and glycogen structure. HYPOTHESIS: We hypothesized that the molecular structure of liver glycogen from a rat model of liver fibrosis would be altered compared to the control group.
Collapse
Affiliation(s)
- Yujun Wan
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Zhenxia Hu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Qinghua Liu
- Jiangsu Provincial Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China; State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Liang Wang
- Jiangsu Provincial Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China; Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, Jiangsu Province 221000, China
| | - Mitchell A Sullivan
- Glycation and Diabetes Group, Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Robert G Gilbert
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Brisbane, Queensland 4072, Australia; Joint International Research Laboratory of Agriculture and Agri-Product Safety, College of Agriculture, Yangzhou University, Yangzhou, Jiangsu 225009, China.
| |
Collapse
|
23
|
Meister J, Bone DBJ, Knudsen JR, Barella LF, Velenosi TJ, Akhmedov D, Lee RJ, Cohen AH, Gavrilova O, Cui Y, Karsenty G, Chen M, Weinstein LS, Kleinert M, Berdeaux R, Jensen TE, Richter EA, Wess J. Clenbuterol exerts antidiabetic activity through metabolic reprogramming of skeletal muscle cells. Nat Commun 2022; 13:22. [PMID: 35013148 PMCID: PMC8748640 DOI: 10.1038/s41467-021-27540-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 11/24/2021] [Indexed: 12/14/2022] Open
Abstract
Activation of the sympathetic nervous system causes pronounced metabolic changes that are mediated by multiple adrenergic receptor subtypes. Systemic treatment with β2-adrenergic receptor agonists results in multiple beneficial metabolic effects, including improved glucose homeostasis. To elucidate the underlying cellular and molecular mechanisms, we chronically treated wild-type mice and several newly developed mutant mouse strains with clenbuterol, a selective β2-adrenergic receptor agonist. Clenbuterol administration caused pronounced improvements in glucose homeostasis and prevented the metabolic deficits in mouse models of β-cell dysfunction and insulin resistance. Studies with skeletal muscle-specific mutant mice demonstrated that these metabolic improvements required activation of skeletal muscle β2-adrenergic receptors and the stimulatory G protein, Gs. Unbiased transcriptomic and metabolomic analyses showed that chronic β2-adrenergic receptor stimulation caused metabolic reprogramming of skeletal muscle characterized by enhanced glucose utilization. These findings strongly suggest that agents targeting skeletal muscle metabolism by modulating β2-adrenergic receptor-dependent signaling pathways may prove beneficial as antidiabetic drugs.
Collapse
Affiliation(s)
- Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA.
| | - Derek B J Bone
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Jonas R Knudsen
- Departments of Nutrition, Exercise and Sports, University of Copenhagen, København, Denmark
| | - Luiz F Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Thomas J Velenosi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Dmitry Akhmedov
- Departments of Integrative Biology and Pharmacology, Houston Medical School, Houston, TX, 77030, USA
| | - Regina J Lee
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Amanda H Cohen
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Gerard Karsenty
- Departments of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| | - Maximilian Kleinert
- Departments of Nutrition, Exercise and Sports, University of Copenhagen, København, Denmark
- Muscle Physiology and Metabolism Group, German Institute of Human Nutrition, Potsdam-Rehbrücke, Nuthetal, Germany
| | - Rebecca Berdeaux
- Departments of Integrative Biology and Pharmacology, Houston Medical School, Houston, TX, 77030, USA
| | - Thomas E Jensen
- Departments of Nutrition, Exercise and Sports, University of Copenhagen, København, Denmark
| | - Erik A Richter
- Departments of Nutrition, Exercise and Sports, University of Copenhagen, København, Denmark
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Hu Y, Li C, Hou Y. Possible regulation of liver glycogen structure through the gut-liver axis by resistant starch: a review. Food Funct 2021; 12:11154-11164. [PMID: 34694313 DOI: 10.1039/d1fo02416g] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Liver glycogen α particles in diabetic patients are fragile relative to those in healthy individuals, and restoring these fragile glycogen particles to a normal state shows potential to contribute to the remission of diabetes. Resistant starch (RS) is beneficial for diabetes management through its interactions with the gut microbiota. However, its effects on glycogen fragility are not fully understood. This review aims to summarize the recent understanding of the interactions between RS and the human gut microbiota and the possible connections to liver glycogen biosynthesis to elucidate its role in the development of glycogen fragility. RS might regulate glycogen fragility in diabetes by modulating the postprandial glycemic response and glycogen biosynthesis pathways. Before RS can be applied to repair fragile glycogen, more work should be done to better understand in vivo RS structures and identify the factor binding glycogen β particles together. This review contains important information on the connections between glycogen fragility and RS-gut microbiota interactions, which could help to better understand the health benefits of RS consumption.
Collapse
Affiliation(s)
- Yiming Hu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200031, China.
| | - Cheng Li
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200031, China.
| |
Collapse
|
25
|
Liu QH, Tang JW, Wen PB, Wang MM, Zhang X, Wang L. From Prokaryotes to Eukaryotes: Insights Into the Molecular Structure of Glycogen Particles. Front Mol Biosci 2021; 8:673315. [PMID: 33996916 PMCID: PMC8116748 DOI: 10.3389/fmolb.2021.673315] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/07/2021] [Indexed: 12/25/2022] Open
Abstract
Glycogen is a highly-branched polysaccharide that is widely distributed across the three life domains. It has versatile functions in physiological activities such as energy reserve, osmotic regulation, blood glucose homeostasis, and pH maintenance. Recent research also confirms that glycogen plays important roles in longevity and cognition. Intrinsically, glycogen function is determined by its structure that has been intensively studied for many years. The recent association of glycogen α-particle fragility with diabetic conditions further strengthens the importance of glycogen structure in its function. By using improved glycogen extraction procedures and a series of advanced analytical techniques, the fine molecular structure of glycogen particles in human beings and several model organisms such as Escherichia coli, Caenorhabditis elegans, Mus musculus, and Rat rattus have been characterized. However, there are still many unknowns about the assembly mechanisms of glycogen particles, the dynamic changes of glycogen structures, and the composition of glycogen associated proteins (glycogen proteome). In this review, we explored the recent progresses in glycogen studies with a focus on the structure of glycogen particles, which may not only provide insights into glycogen functions, but also facilitate the discovery of novel drug targets for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Qing-Hua Liu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China.,Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jia-Wei Tang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Peng-Bo Wen
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Meng-Meng Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Xiao Zhang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China
| | - Liang Wang
- Department of Bioinformatics, School of Medical Informatics and Engineering, Xuzhou Medical University, Xuzhou, China.,Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
26
|
Jensen R, Ørtenblad N, Stausholm MLH, Skjaerbaek MC, Larsen DN, Hansen M, Holmberg HC, Plomgaard P, Nielsen J. Glycogen supercompensation is due to increased number, not size, of glycogen particles in human skeletal muscle. Exp Physiol 2021; 106:1272-1284. [PMID: 33675088 DOI: 10.1113/ep089317] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/01/2021] [Indexed: 01/10/2023]
Abstract
NEW FINDINGS What is the central question of this study? Glycogen supercompensation after glycogen-depleting exercise can be achieved by consuming a carbohydrate-enriched diet, but the associated effects on the size, number and localization of intramuscular glycogen particles are unknown. What is the main finding and its importance? Using transmission electron microscopy to inspect individual glycogen particles visually, we show that glycogen supercompensation is achieved by increasing the number of particles while keeping them at submaximal sizes. This might be a strategy to ensure that glycogen particles can be used fast, because particles that are too large might impair utilization rate. ABSTRACT Glycogen supercompensation after glycogen-depleting exercise can be achieved by consuming a carbohydrate-enriched diet, but the associated effects on the size, number and localization of intramuscular glycogen particles are unknown. We investigated how a glycogen-loading protocol affects fibre type-specific glycogen volume density, particle diameter and numerical density in three subcellular pools: between (intermyofibrillar) or within (intramyofibrillar) the myofibrils or beneath the sarcolemma (subsarcolemmal). Resting muscle biopsies from 11 physically active men were analysed using transmission electron microscopy after mixed (MIX), LOW or HIGH carbohydrate consumption separated by glycogen-lowering cycling at 75% of maximal oxygen consumption until exhaustion. After HIGH, the total volumetric glycogen content was 40% [95% confidence interval 16, 68] higher than after MIX in type I fibres (P < 0.001), with little to no difference in type II fibres (9% [95% confidence interval -9, 27]). Median particle diameter was 22.5 (interquartile range 20.8-24.7) nm across glycogen pools and fibre types, and the numerical density was 61% [25, 107] and 40% [9, 80] higher in the subsarcolemmal (P < 0.001) and intermyofibrillar (P < 0.01) pools of type I fibres, respectively, with little to no difference in the intramyofibrillar pool (3% [-20, 32]). In LOW, total glycogen was in the range of 21-23% lower, relative to MIX, in both fibre types, reflected in a 21-46% lower numerical density across pools. In comparison to MIX, particle diameter was unaffected by other diets ([-1.4, 1.3] nm). In conclusion, glycogen supercompensation after prolonged cycling is exclusive to type I fibres, predominantly in the subsarcolemmal pool, and involves an increase in the numerical density rather than the size of existing glycogen particles.
Collapse
Affiliation(s)
- Rasmus Jensen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Marie-Louise H Stausholm
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Mette C Skjaerbaek
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Daniel N Larsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Mette Hansen
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Hans-Christer Holmberg
- Department of Physiology and Pharmacology, Biomedicum C5, Karolinska Institutet, Stockholm, Sweden
| | - Peter Plomgaard
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Joachim Nielsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
27
|
López-Soldado I, Guinovart JJ, Duran J. Increasing hepatic glycogen moderates the diabetic phenotype in insulin-deficient Akita mice. J Biol Chem 2021; 296:100498. [PMID: 33667544 PMCID: PMC8027280 DOI: 10.1016/j.jbc.2021.100498] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatic glycogen metabolism is impaired in diabetes. We previously demonstrated that strategies to increase liver glycogen content in a high-fat-diet mouse model of obesity and insulin resistance led to a reduction in food intake and ameliorated obesity and glucose tolerance. These effects were accompanied by a decrease in insulin levels, but whether this decrease contributed to the phenotype observed in this animal was unclear. Here we sought to evaluate this aspect directly, by examining the long-term effects of increasing liver glycogen in an animal model of insulin-deficient and monogenic diabetes, namely the Akita mouse, which is characterized by reduced insulin production. We crossed Akita mice with animals overexpressing protein targeting to glycogen (PTG) in the liver to generate Akita mice with increased liver glycogen content (Akita-PTGOE). Akita-PTGOE animals showed lower glycemia, lower food intake, and decreased water consumption and urine output compared with Akita mice. Furthermore, Akita-PTGOE mice showed a restoration of the hepatic energy state and a normalization of gluconeogenesis and glycolysis back to nondiabetic levels. Moreover, hepatic lipogenesis, which is reduced in Akita mice, was reverted in Akita-PTGOE animals. These results demonstrate that strategies to increase liver glycogen content lead to the long-term reduction of the diabetic phenotype, independently of circulating insulin.
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
|
28
|
García-Ponce ÁL, Martínez-Poveda B, Blanco-López Á, Quesada AR, Suárez F, Alonso-Carrión FJ, Medina MÁ. A problem-/case-based learning approach as an useful tool for studying glycogen metabolism and its regulation. BIOCHEMISTRY AND MOLECULAR BIOLOGY EDUCATION : A BIMONTHLY PUBLICATION OF THE INTERNATIONAL UNION OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2021; 49:236-241. [PMID: 32897596 DOI: 10.1002/bmb.21449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/09/2020] [Accepted: 08/21/2020] [Indexed: 06/11/2023]
Abstract
Metabolism and its regulation is one of the most complex and difficult topics for students learning biochemistry. A problem-/case-based learning (PBL) approach can be useful to help biochemistry students to fulfill the goal of acquiring an integrated view of metabolism and its regulation. The present article describes our experience enrolling volunteer students to learn glycogen metabolism making use of a design-based research methodology to develop teaching learning sequences focused on a PBL approach. Enrolled undergraduate students had better final scores than those students that did not participates. Furthermore, enrolled students were satisfied with the experience, finding it interesting, formative, and challenging.
Collapse
Affiliation(s)
- Ángel Luis García-Ponce
- Departamento de Didáctica de la Matemática, de las Ciencias Sociales y de las Ciencias Experimentales, Facultad de Ciencias de la Educación, Universidad de Málaga, Andalucía Tech, Málaga, Spain
| | - Beatriz Martínez-Poveda
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, Málaga, Spain
| | - Ángel Blanco-López
- Departamento de Didáctica de la Matemática, de las Ciencias Sociales y de las Ciencias Experimentales, Facultad de Ciencias de la Educación, Universidad de Málaga, Andalucía Tech, Málaga, Spain
| | - Ana R Quesada
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, Málaga, Spain
| | - Fernanda Suárez
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, Málaga, Spain
| | - Francisco José Alonso-Carrión
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, Málaga, Spain
| | - Miguel Ángel Medina
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, Málaga, Spain
| |
Collapse
|
29
|
Hokken R, Laugesen S, Aagaard P, Suetta C, Frandsen U, Ørtenblad N, Nielsen J. Subcellular localization- and fibre type-dependent utilization of muscle glycogen during heavy resistance exercise in elite power and Olympic weightlifters. Acta Physiol (Oxf) 2021; 231:e13561. [PMID: 32961628 DOI: 10.1111/apha.13561] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/13/2020] [Accepted: 09/14/2020] [Indexed: 12/16/2022]
Abstract
AIM Glycogen particles are found in different subcellular localizations, which are utilized heterogeneously in different fibre types during endurance exercise. Although resistance exercise typically involves only a moderate use of mixed muscle glycogen, the hypothesis of the present study was that high-volume heavy-load resistance exercise would mediate a pattern of substantial glycogen depletion in specific subcellular localizations and fibre types. METHODS 10 male elite weightlifters performed resistance exercise consisting of four sets of five (4 × 5) repetitions at 75% of 1RM back squats, 4 × 5 at 75% of 1RM deadlifts and 4 × 12 at 65% of 1RM rear foot elevated split squats. Muscle biopsies (vastus lateralis) were obtained before and after the exercise session. The volumetric content of intermyofibrillar (between myofibrils), intramyofibrillar (within myofibrils) and subsarcolemmal glycogen was assessed by transmission electron microscopy. RESULTS After exercise, biochemically determined muscle glycogen decreased by 38 (31:45)%. Location-specific glycogen analyses revealed in type 1 fibres a large decrement in intermyofibrillar glycogen, but no or only minor changes in intramyofibrillar or subsarcolemmal glycogen. In type 2 fibres, large decrements in glycogen were observed in all subcellular localizations. Notably, a substantial fraction of the type 2 fibres demonstrated near-depleted levels of intramyofibrillar glycogen after the exercise session. CONCLUSION Heavy resistance exercise mediates a substantial utilization of glycogen from all three subcellular localization in type 2 fibres, while mostly taxing intermyofibrillar glycogen stores in type 1 fibres. Thus, a better understanding of the impact of resistance training on myocellular metabolism and performance requires a focus on compartmentalized glycogen utilization.
Collapse
Affiliation(s)
- Rune Hokken
- Department of Sports Science and Clinical Biomechanics University of Southern Denmark Odense M Denmark
| | - Simon Laugesen
- Department of Sports Science and Clinical Biomechanics University of Southern Denmark Odense M Denmark
| | - Per Aagaard
- Department of Sports Science and Clinical Biomechanics University of Southern Denmark Odense M Denmark
| | - Charlotte Suetta
- Geriatric Research Unit Department of Geriatrics Bispebjerg‐Frederiksberg and Herlev‐Gentofte HospitalsUniversity of Copenhagen Kobenhavn Denmark
| | - Ulrik Frandsen
- Department of Sports Science and Clinical Biomechanics University of Southern Denmark Odense M Denmark
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics University of Southern Denmark Odense M Denmark
| | - Joachim Nielsen
- Department of Sports Science and Clinical Biomechanics University of Southern Denmark Odense M Denmark
| |
Collapse
|
30
|
Laforêt P, Oldfors A, Malfatti E, Vissing J. 251st ENMC international workshop: Polyglucosan storage myopathies 13-15 December 2019, Hoofddorp, the Netherlands. Neuromuscul Disord 2021; 31:466-477. [PMID: 33602551 DOI: 10.1016/j.nmd.2021.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Pascal Laforêt
- Neurology Unit, Raymond Poincaré Hospital, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - Anders Oldfors
- Department of Laboratory Medicine, Sahlgrenska University Hospital, Institute of Biomedicine, University of Gothenburg, Sweden.
| | - Edoardo Malfatti
- Neuromuscular Reference Center, Henri Mondor University Hospital, Université Versailles Saint-Quentin-en-Yvelines, Montigny-le-Bretonneux, France
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Denmark
| | | |
Collapse
|
31
|
Deshmukh AS, Steenberg DE, Hostrup M, Birk JB, Larsen JK, Santos A, Kjøbsted R, Hingst JR, Schéele CC, Murgia M, Kiens B, Richter EA, Mann M, Wojtaszewski JFP. Deep muscle-proteomic analysis of freeze-dried human muscle biopsies reveals fiber type-specific adaptations to exercise training. Nat Commun 2021; 12:304. [PMID: 33436631 PMCID: PMC7803955 DOI: 10.1038/s41467-020-20556-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 12/01/2020] [Indexed: 01/29/2023] Open
Abstract
Skeletal muscle conveys several of the health-promoting effects of exercise; yet the underlying mechanisms are not fully elucidated. Studying skeletal muscle is challenging due to its different fiber types and the presence of non-muscle cells. This can be circumvented by isolation of single muscle fibers. Here, we develop a workflow enabling proteomics analysis of pools of isolated muscle fibers from freeze-dried human muscle biopsies. We identify more than 4000 proteins in slow- and fast-twitch muscle fibers. Exercise training alters expression of 237 and 172 proteins in slow- and fast-twitch muscle fibers, respectively. Interestingly, expression levels of secreted proteins and proteins involved in transcription, mitochondrial metabolism, Ca2+ signaling, and fat and glucose metabolism adapts to training in a fiber type-specific manner. Our data provide a resource to elucidate molecular mechanisms underlying muscle function and health, and our workflow allows fiber type-specific proteomic analyses of snap-frozen non-embedded human muscle biopsies.
Collapse
Affiliation(s)
- A S Deshmukh
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- The Novo Nordisk Foundation Center for Basic Metablic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - D E Steenberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - M Hostrup
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - J B Birk
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - J K Larsen
- The Novo Nordisk Foundation Center for Basic Metablic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - A Santos
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - R Kjøbsted
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - J R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - C C Schéele
- The Novo Nordisk Foundation Center for Basic Metablic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Centre of Inflammation and Metabolism and Centre for Physical Activity Research Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - M Murgia
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - B Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - E A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - M Mann
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | - J F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
32
|
Almodóvar-Payá A, Villarreal-Salazar M, de Luna N, Nogales-Gadea G, Real-Martínez A, Andreu AL, Martín MA, Arenas J, Lucia A, Vissing J, Krag T, Pinós T. Preclinical Research in Glycogen Storage Diseases: A Comprehensive Review of Current Animal Models. Int J Mol Sci 2020; 21:ijms21249621. [PMID: 33348688 PMCID: PMC7766110 DOI: 10.3390/ijms21249621] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
GSD are a group of disorders characterized by a defect in gene expression of specific enzymes involved in glycogen breakdown or synthesis, commonly resulting in the accumulation of glycogen in various tissues (primarily the liver and skeletal muscle). Several different GSD animal models have been found to naturally present spontaneous mutations and others have been developed and characterized in order to further understand the physiopathology of these diseases and as a useful tool to evaluate potential therapeutic strategies. In the present work we have reviewed a total of 42 different animal models of GSD, including 26 genetically modified mouse models, 15 naturally occurring models (encompassing quails, cats, dogs, sheep, cattle and horses), and one genetically modified zebrafish model. To our knowledge, this is the most complete list of GSD animal models ever reviewed. Importantly, when all these animal models are analyzed together, we can observe some common traits, as well as model specific differences, that would be overlooked if each model was only studied in the context of a given GSD.
Collapse
Affiliation(s)
- Aitana Almodóvar-Payá
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
| | - Mónica Villarreal-Salazar
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
| | - Noemí de Luna
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Laboratori de Malalties Neuromusculars, Institut de Recerca Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
| | - Gisela Nogales-Gadea
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Grup de Recerca en Malalties Neuromusculars i Neuropediàtriques, Department of Neurosciences, Institut d’Investigacio en Ciencies de la Salut Germans Trias i Pujol i Campus Can Ruti, Universitat Autònoma de Barcelona, 08916 Badalona, Spain
| | - Alberto Real-Martínez
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
| | - Antoni L. Andreu
- EATRIS, European Infrastructure for Translational Medicine, 1081 HZ Amsterdam, The Netherlands;
| | - Miguel Angel Martín
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Mitochondrial and Neuromuscular Diseases Laboratory, 12 de Octubre Hospital Research Institute (i+12), 28041 Madrid, Spain
| | - Joaquin Arenas
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Mitochondrial and Neuromuscular Diseases Laboratory, 12 de Octubre Hospital Research Institute (i+12), 28041 Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, European University, 28670 Madrid, Spain;
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark; (J.V.); (T.K.)
| | - Thomas Krag
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark; (J.V.); (T.K.)
| | - Tomàs Pinós
- Mitochondrial and Neuromuscular Disorders Unit, Vall d’Hebron Institut de Recerca, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (A.A.-P.); (M.V.-S.); (A.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; (N.d.L.); (G.N.-G.); (M.A.M.); (J.A.)
- Correspondence: ; Tel.: +34-934894057
| |
Collapse
|
33
|
Joseph VM, Nagy MT, Akhtar S, Ng CY. Sequential spontaneous compartment syndrome in multiple limbs in a young adult with GYG1 gene mutation. BMJ Case Rep 2020; 13:13/11/e236226. [PMID: 33257366 DOI: 10.1136/bcr-2020-236226] [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] [Indexed: 11/03/2022] Open
Abstract
Compartment syndrome is a surgical emergency requiring immediate intervention. Majority of compartment syndromes are associated with trauma or surgery. Spontaneous compartment syndrome in multiple limbs is rare and alternative diagnosis should be sought. We report a young adult man who developed compartment syndrome in all four limbs sequentially over 4 years. On further evaluation, he was found to have a gene mutation in exon 3 of GYG1 gene. Spontaneous compartment syndrome in patients with GYG1 gene mutation does not appear to have been previously recognised. Although a direct causality cannot be confidently drawn, this gene is involved in muscle energy utilisation and is known to cause metabolic defect. Acute compartment syndrome, once diagnosed, warrants emergency surgical decompression. The subsequent management of spontaneous compartment syndrome demands a thorough medical assessment to identify any underlying metabolic or genetic predisposition.
Collapse
Affiliation(s)
- Vinay Mathew Joseph
- Department of Trauma and Orthopaedic Surgery, Wrightington Hospital, Wigan, UK
| | - Mathias Thomas Nagy
- Department of Trauma and Orthopaedic Surgery, Wrightington Hospital, Wigan, UK
| | - Sohail Akhtar
- Department of Trauma and Orthopaedic Surgery, Wrightington Hospital, Wigan, UK
| | - Chye Yew Ng
- Department of Trauma and Orthopaedic Surgery, Wrightington Hospital, Wigan, UK
| |
Collapse
|
34
|
Testoni G, Olmeda B, Duran J, López-Rodríguez E, Aguilera M, Hernández-Álvarez MI, Prats N, Pérez-Gil J, Guinovart JJ. Pulmonary glycogen deficiency as a new potential cause of respiratory distress syndrome. Hum Mol Genet 2020; 29:3554-3565. [PMID: 33219378 DOI: 10.1093/hmg/ddaa249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/06/2020] [Accepted: 11/12/2020] [Indexed: 11/14/2022] Open
Abstract
The glycogenin knockout mouse is a model of Glycogen Storage Disease type XV. These animals show high perinatal mortality (90%) due to respiratory failure. The lungs of glycogenin-deficient embryos and P0 mice have a lower glycogen content than that of wild-type counterparts. Embryonic lungs were found to have decreased levels of mature surfactant proteins SP-B and SP-C, together with incomplete processing of precursors. Furthermore, non-surviving pups showed collapsed sacculi, which may be linked to a significantly reduced amount of surfactant proteins. A similar pattern was observed in glycogen synthase1-deficient mice, which are devoid of glycogen in the lungs and are also affected by high perinatal mortality due to atelectasis. These results indicate that glycogen availability is a key factor for the burst of surfactant production required to ensure correct lung expansion at the establishment of air breathing. Our findings confirm that glycogen deficiency in lungs can cause respiratory distress syndrome and suggest that mutations in glycogenin and glycogen synthase 1 genes may underlie cases of idiopathic neonatal death.
Collapse
Affiliation(s)
- Giorgia Testoni
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Bárbara Olmeda
- Department of Biochemistry, Faculty of Biology, and Research Institute of Hospital 12 de Octubre, Complutense University, 28040 Madrid, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Elena López-Rodríguez
- Institute of Functional Anatomy Wilhelm-Waldeyer-Haus, Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Mònica Aguilera
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - María Isabel Hernández-Álvarez
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Neus Prats
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Jesús Pérez-Gil
- Department of Biochemistry, Faculty of Biology, and Research Institute of Hospital 12 de Octubre, Complutense University, 28040 Madrid, Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain.,Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
35
|
Besford QA, Weiss ACG, Schubert J, Ryan TM, Maitz MF, Tomanin PP, Savioli M, Werner C, Fery A, Caruso F, Cavalieri F. Protein Component of Oyster Glycogen Nanoparticles: An Anchor Point for Functionalization. ACS APPLIED MATERIALS & INTERFACES 2020; 12:38976-38988. [PMID: 32805918 DOI: 10.1021/acsami.0c10699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Biosourced nanoparticles have a range of desirable properties for therapeutic applications, including biodegradability and low immunogenicity. Glycogen, a natural polysaccharide nanoparticle, has garnered much interest as a component of advanced therapeutic materials. However, functionalizing glycogen for use as a therapeutic material typically involves synthetic approaches that can negatively affect the intrinsic physiological properties of glycogen. Herein, the protein component of glycogen is examined as an anchor point for the photopolymerization of functional poly(N-isopropylacrylamide) (PNIPAM) polymers. Oyster glycogen (OG) nanoparticles partially degrade to smaller spherical particles in the presence of protease enzymes, reflecting a population of surface-bound proteins on the polysaccharide. The grafting of PNIPAM to the native protein component of OG produces OG-PNIPAM nanoparticles of ∼45 nm in diameter and 6.2 MDa in molecular weight. PNIPAM endows the nanoparticles with temperature-responsive aggregation properties that are controllable and reversible and that can be removed by the biodegradation of the protein. The OG-PNIPAM nanoparticles retain the native biodegradability of glycogen. Whole blood incubation assays revealed that the OG-PNIPAM nanoparticles have a low cell association and inflammatory response similar to that of OG. The reported strategy provides functionalized glycogen nanomaterials that retain their inherent biodegradability and low immune cell association.
Collapse
Affiliation(s)
- Quinn A Besford
- Leibniz Institute for Polymer Research, Hohe Straße 6, 01069 Dresden, Germany
| | - Alessia C G Weiss
- Leibniz Institute for Polymer Research, Hohe Straße 6, 01069 Dresden, Germany
| | - Jonas Schubert
- Leibniz Institute for Polymer Research, Hohe Straße 6, 01069 Dresden, Germany
| | - Timothy M Ryan
- The Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Manfred F Maitz
- Leibniz Institute for Polymer Research, Hohe Straße 6, 01069 Dresden, Germany
| | - Pietro Pacchin Tomanin
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marco Savioli
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Carsten Werner
- Leibniz Institute for Polymer Research, Hohe Straße 6, 01069 Dresden, Germany
| | - Andreas Fery
- Leibniz Institute for Polymer Research, Hohe Straße 6, 01069 Dresden, Germany
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Francesca Cavalieri
- School of Science, RMIT University, Melbourne, Victoria 3000, Australia
- Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma "Tor Vergata", Via Della Ricerca Scientifica 1, 00133 Rome, Italy
| |
Collapse
|
36
|
Nitschke S, Petković S, Ahonen S, Minassian BA, Nitschke F. Sensitive quantification of α-glucans in mouse tissues, cell cultures, and human cerebrospinal fluid. J Biol Chem 2020; 295:14698-14709. [PMID: 32817315 DOI: 10.1074/jbc.ra120.015061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/04/2020] [Indexed: 12/30/2022] Open
Abstract
The soluble α-polyglucan glycogen is a central metabolite enabling transient glucose storage to suit cellular energy needs. Glycogen storage diseases (GSDs) comprise over 15 entities caused by generalized or tissue-specific defects in enzymes of glycogen metabolism. In several, e.g. in Lafora disease caused by the absence of the glycogen phosphatase laforin or its interacting partner malin, degradation-resistant abnormally structured insoluble glycogen accumulates. Sensitive quantification methods for soluble and insoluble glycogen are critical to research, including therapeutic studies, in such diseases. This paper establishes methodological advancements relevant to glycogen metabolism investigations generally, and GSDs. Introducing a pre-extraction incubation method, we measure degradation-resistant glycogen in as little as 30 mg of skeletal muscle or a single hippocampus from Lafora disease mouse models. The digestion-resistant glycogen correlates with the disease-pathogenic insoluble glycogen and can readily be detected in very young mice where glycogen accumulation has just begun. Second, we establish a high-sensitivity glucose assay with detection of ATP depletion, enabling 1) quantification of α-glucans in cell culture using a medium-throughput assay suitable for assessment of candidate glycogen synthesis inhibitors, and 2) discovery of α-glucan material in healthy human cerebrospinal fluid, establishing a novel methodological platform for biomarker analyses in Lafora disease and other GSDs.
Collapse
Affiliation(s)
- Silvia Nitschke
- Departments of Pediatrics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sara Petković
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Saija Ahonen
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Berge A Minassian
- Departments of Pediatrics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Felix Nitschke
- Departments of Pediatrics and Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
37
|
Metastasis of Uveal Melanoma with Monosomy-3 Is Associated with a Less Glycogenetic Gene Expression Profile and the Dysregulation of Glycogen Storage. Cancers (Basel) 2020; 12:cancers12082101. [PMID: 32751097 PMCID: PMC7463985 DOI: 10.3390/cancers12082101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
The prolonged storage of glucose as glycogen can promote the quiescence of tumor cells, whereas the accumulation of an aberrant form of glycogen without the primer protein glycogenin can induce the metabolic switch towards a glycolytic phenotype. Here, we analyzed the expression of n = 67 genes involved in glycogen metabolism on the uveal melanoma (UM) cohort of the Cancer Genome Atlas (TCGA) study and validated the differentially expressed genes in an independent cohort. We also evaluated the glycogen levels with regard to the prognostic factors via a differential periodic acid-Schiff (PAS) staining. UMs with monosomy-3 exhibited a less glycogenetic and more insulin-resistant gene expression profile, together with the reduction of glycogen levels, which were associated with the metastases. Expression of glycogenin-1 (Locus: 3q24) was lower in the monosomy-3 tumors, whereas the complementary isoform glycogenin-2 (Locus: Xp22.33) was upregulated in females. Remarkably, glycogen was more abundant in the monosomy-3 tumors of male versus female patients. We therefore provide the first evidence to the dysregulation of glycogen metabolism as a novel factor that may be aggravating the course of UM particularly in males.
Collapse
|
38
|
Roberts GAG, Tunster SJ. Characterising the dynamics of placental glycogen stores in the mouse. Placenta 2020; 99:131-140. [PMID: 32798765 DOI: 10.1016/j.placenta.2020.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/26/2020] [Accepted: 07/13/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The placenta performs a range of functions to support fetal growth. In addition to facilitating nutrient transport, the placenta also stores glucose as glycogen, which is thought to maintain fetal glucose supply during late gestation. However, evidence to support such a role is currently lacking. Similarly, our understanding of the dynamics of placental glycogen metabolism in normal mouse pregnancy is limited. METHODS We quantified the placental glycogen content of wild type C57BL/6JOlaHsd mouse placentas from mid (E12.5) to late (E18.5) gestation, alongside characterising the temporal expression pattern of genes encoding glycogenesis and glycogenolysis pathway enzymes. To assess the potential of the placenta to produce glucose, we investigated the spatiotemporal expression of glucose 6-phosphatase by qPCR and in situ hybridisation. Separate analyses were undertaken for placentas of male and female conceptuses to account for potential sexual dimorphism. RESULTS Placental glycogen stores peak at E15.5, having increased over 5-fold from E12.5, before declining by a similar extent by E18.5. Glycogen stores were 17% higher in male placentas than in females at E15.5. Expression of glycogen branching enzyme (Gbe1) was reduced ~40% towards term. Expression of the glucose 6-phosphatase isoform G6pc3 was enriched in glycogen trophoblast cells and increased towards term. DISCUSSION Reduced expression of Gbe1 suggests a decline in glycogen branching towards term. Expression of G6pc3 by glycogen trophoblasts is consistent with an ability to produce and release glucose from glycogen stores. However, the ultimate destination of the glucose generated from placental glycogen remains to be elucidated.
Collapse
Affiliation(s)
- George A G Roberts
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - Simon J Tunster
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.
| |
Collapse
|
39
|
Mandalasi M, Kim HW, Thieker D, Sheikh MO, Gas-Pascual E, Rahman K, Zhao P, Daniel NG, van der Wel H, Ichikawa HT, Glushka JN, Wells L, Woods RJ, Wood ZA, West CM. A terminal α3-galactose modification regulates an E3 ubiquitin ligase subunit in Toxoplasma gondii. J Biol Chem 2020; 295:9223-9243. [PMID: 32414843 PMCID: PMC7335778 DOI: 10.1074/jbc.ra120.013792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/14/2020] [Indexed: 12/29/2022] Open
Abstract
Skp1, a subunit of E3 Skp1/Cullin-1/F-box protein ubiquitin ligases, is modified by a prolyl hydroxylase that mediates O2 regulation of the social amoeba Dictyostelium and the parasite Toxoplasma gondii The full effect of hydroxylation requires modification of the hydroxyproline by a pentasaccharide that, in Dictyostelium, influences Skp1 structure to favor assembly of Skp1/F-box protein subcomplexes. In Toxoplasma, the presence of a contrasting penultimate sugar assembled by a different glycosyltransferase enables testing of the conformational control model. To define the final sugar and its linkage, here we identified the glycosyltransferase that completes the glycan and found that it is closely related to glycogenin, an enzyme that may prime glycogen synthesis in yeast and animals. However, the Toxoplasma enzyme catalyzes formation of a Galα1,3Glcα linkage rather than the Glcα1,4Glcα linkage formed by glycogenin. Kinetic and crystallographic experiments showed that the glycosyltransferase Gat1 is specific for Skp1 in Toxoplasma and also in another protist, the crop pathogen Pythium ultimum The fifth sugar is important for glycan function as indicated by the slow-growth phenotype of gat1Δ parasites. Computational analyses indicated that, despite the sequence difference, the Toxoplasma glycan still assumes an ordered conformation that controls Skp1 structure and revealed the importance of nonpolar packing interactions of the fifth sugar. The substitution of glycosyltransferases in Toxoplasma and Pythium by an unrelated bifunctional enzyme that assembles a distinct but structurally compatible glycan in Dictyostelium is a remarkable case of convergent evolution, which emphasizes the importance of the terminal α-galactose and establishes the phylogenetic breadth of Skp1 glycoregulation.
Collapse
Affiliation(s)
- Msano Mandalasi
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Hyun W Kim
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - David Thieker
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - M Osman Sheikh
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Elisabet Gas-Pascual
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Kazi Rahman
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Peng Zhao
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Nitin G Daniel
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Hanke van der Wel
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - H Travis Ichikawa
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - John N Glushka
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Lance Wells
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA; Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Robert J Woods
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA; Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Zachary A Wood
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Christopher M West
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA; Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA.
| |
Collapse
|
40
|
Chokeshaiusaha K, Puthier D, Sananmuang T, Olanratmanee EO, Nguyen C, Kedkovid R. Differential DNA methylation analysis across the promoter regions using methylated DNA immunoprecipitation sequencing profiling of porcine loin muscle. Vet World 2020; 13:1113-1125. [PMID: 32801562 PMCID: PMC7396332 DOI: 10.14202/vetworld.2020.1113-1125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Background and Aim: Pork leanness and marbling are among the essential traits of consumer preference. To acquire knowledge about universal epigenetic regulations for improving breed selection, a meta-analysis of methylated DNA immunoprecipitation sequencing (MeDIP-seq) profiling data of mixed loin muscle types was performed in this study. Materials and Methods: MeDIP-seq profiling datasets of longissimus dorsi muscle and psoas major muscles from male and female pigs of Landrace and Tibetan breeds were preprocessed and aligned to the porcine genome. Analysis of differential methylated DNA regions (DMRs) between the breeds was performed by focusing on transcription start sites (TSSs) of known genes (−20,000-3000 bases from TSS). All associated genes were further reviewed for their functions and predicted for transcription factors (TF) possibly associated with their TSSs. Results: When the methylation levels of DMRs in TSS regions of Landrace breed were compared to those of Tibetan breed, 10 DMRs were hypomethylated (Landrace < Tibetan), and 19 DMRs were hypermethylated (Landrace > Tibetan), accordingly (p≤0.001). According to the reviews about gene functions, all associated genes were pieces of evidence for their roles in a variety of muscle and lipid metabolisms. Prediction of the binding TFs revealed the six most abundant binding TFs to such DMRs-associated TSS (p≤0.0001) as follows: ZNF384, Foxd3, IRF1, KLF9, EWSR1-FLI1, HES5, and TFAP2A. Conclusion: Common DMRs-associated TSS between the lean-type and the marbled-type loin muscles were identified in this study. Interestingly, the genes associated with such regions were strongly evidenced for their possible roles on the muscle trait characteristics by which further novel research topics could be focused on them in the future.
Collapse
Affiliation(s)
- Kaj Chokeshaiusaha
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chon Buri, Thailand
| | - Denis Puthier
- Aix-Marseille University, INSERM UMR 1090, TAGC, Marseille, France
| | - Thanida Sananmuang
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chon Buri, Thailand
| | - Em-On Olanratmanee
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chon Buri, Thailand
| | - Catherine Nguyen
- Aix-Marseille University, INSERM UMR 1090, TAGC, Marseille, France
| | - Roongtham Kedkovid
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Swine Reproduction Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
41
|
Mair D, Biskup S, Kress W, Abicht A, Brück W, Zechel S, Knop KC, Koenig FB, Tey S, Nikolin S, Eggermann K, Kurth I, Ferbert A, Weis J. Differential diagnosis of vacuolar myopathies in the NGS era. Brain Pathol 2020; 30:877-896. [PMID: 32419263 PMCID: PMC8017999 DOI: 10.1111/bpa.12864] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/10/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Altered autophagy accompanied by abnormal autophagic (rimmed) vacuoles detectable by light and electron microscopy is a common denominator of many familial and sporadic non-inflammatory muscle diseases. Even in the era of next generation sequencing (NGS), late-onset vacuolar myopathies remain a diagnostic challenge. We identified 32 adult vacuolar myopathy patients from 30 unrelated families, studied their clinical, histopathological and ultrastructural characteristics and performed genetic testing in index patients and relatives using Sanger sequencing and NGS including whole exome sequencing (WES). We established a molecular genetic diagnosis in 17 patients. Pathogenic mutations were found in genes typically linked to vacuolar myopathy (GNE, LDB3/ZASP, MYOT, DES and GAA), but also in genes not regularly associated with severely altered autophagy (FKRP, DYSF, CAV3, COL6A2, GYG1 and TRIM32) and in the digenic facioscapulohumeral muscular dystrophy 2. Characteristic histopathological features including distinct patterns of myofibrillar disarray and evidence of exocytosis proved to be helpful to distinguish causes of vacuolar myopathies. Biopsy validated the pathogenicity of the novel mutations p.(Phe55*) and p.(Arg216*) in GYG1 and of the p.(Leu156Pro) TRIM32 mutation combined with compound heterozygous deletion of exon 2 of TRIM32 and expanded the phenotype of Ala93Thr-caveolinopathy and of limb-girdle muscular dystrophy 2i caused by FKRP mutation. In 15 patients no causal variants were detected by Sanger sequencing and NGS panel analysis. In 12 of these cases, WES was performed, but did not yield any definite mutation or likely candidate gene. In one of these patients with a family history of muscle weakness, the vacuolar myopathy was eventually linked to chloroquine therapy. Our study illustrates the wide phenotypic and genotypic heterogeneity of vacuolar myopathies and validates the role of histopathology in assessing the pathogenicity of novel mutations detected by NGS. In a sizable portion of vacuolar myopathy cases, it remains to be shown whether the cause is hereditary or degenerative.
Collapse
Affiliation(s)
- Dorothea Mair
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany.,Department of Neurology, Kassel School of Medicine, Klinikum Kassel, Kassel, Germany.,University of Southampton, Southampton, UK
| | - Saskia Biskup
- Centre for Genomics and Transcriptomics CeGaT, Tübingen, Germany
| | - Wolfram Kress
- Institute of Human Genetics, University Würzburg, Würzburg, Germany
| | | | - Wolfgang Brück
- Institute of Neuropathology, Göttingen University, Göttingen, Germany
| | - Sabrina Zechel
- Institute of Neuropathology, Göttingen University, Göttingen, Germany
| | | | | | - Shelisa Tey
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Stefan Nikolin
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Katja Eggermann
- Institute of Human Genetics, RWTH Aachen University, Aachen, Germany
| | - Ingo Kurth
- Institute of Human Genetics, RWTH Aachen University, Aachen, Germany
| | - Andreas Ferbert
- Department of Neurology, Kassel School of Medicine, Klinikum Kassel, Kassel, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
42
|
Visuttijai K, Hedberg-Oldfors C, Thomsen C, Glamuzina E, Kornblum C, Tasca G, Hernandez-Lain A, Sandstedt J, Dellgren G, Roach P, Oldfors A. Glycogenin is Dispensable for Glycogen Synthesis in Human Muscle, and Glycogenin Deficiency Causes Polyglucosan Storage. J Clin Endocrinol Metab 2020; 105:5599738. [PMID: 31628455 PMCID: PMC7046021 DOI: 10.1210/clinem/dgz075] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023]
Abstract
CONTEXT Glycogenin is considered to be an essential primer for glycogen biosynthesis. Nevertheless, patients with glycogenin-1 deficiency due to biallelic GYG1 (NM_004130.3) mutations can store glycogen in muscle. Glycogenin-2 has been suggested as an alternative primer for glycogen synthesis in patients with glycogenin-1 deficiency. OBJECTIVE The objective of this article is to investigate the importance of glycogenin-1 and glycogenin-2 for glycogen synthesis in skeletal and cardiac muscle. DESIGN, SETTING, AND PATIENTS Glycogenin-1 and glycogenin-2 expression was analyzed by Western blot, mass spectrometry, and immunohistochemistry in liver, heart, and skeletal muscle from controls and in skeletal and cardiac muscle from patients with glycogenin-1 deficiency. RESULTS Glycogenin-1 and glycogenin-2 both were found to be expressed in the liver, but only glycogenin-1 was identified in heart and skeletal muscle from controls. In patients with truncating GYG1 mutations, neither glycogenin-1 nor glycogenin-2 was expressed in skeletal muscle. However, nonfunctional glycogenin-1 but not glycogenin-2 was identified in cardiac muscle from patients with cardiomyopathy due to GYG1 missense mutations. By immunohistochemistry, the mutated glycogenin-1 colocalized with the storage of glycogen and polyglucosan in cardiomyocytes. CONCLUSIONS Glycogen can be synthesized in the absence of glycogenin, and glycogenin-1 deficiency is not compensated for by upregulation of functional glycogenin-2. Absence of glycogenin-1 leads to the focal accumulation of glycogen and polyglucosan in skeletal muscle fibers. Expression of mutated glycogenin-1 in the heart is deleterious, and it leads to storage of abnormal glycogen and cardiomyopathy.
Collapse
Affiliation(s)
- Kittichate Visuttijai
- Department of Pathology and Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carola Hedberg-Oldfors
- Department of Pathology and Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christer Thomsen
- Department of Pathology and Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emma Glamuzina
- National Metabolic Service, Starship Children’s Hospital, Auckland, New Zealand
| | | | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Dipartimento di Scienze dell’Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Joakim Sandstedt
- Department of Clinical Chemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Göran Dellgren
- Department of Cardiothoracic Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Peter Roach
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Anders Oldfors
- Department of Pathology and Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Correspondence and Reprint Requests: Anders Oldfors, Department of Pathology and Genetics, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, 41345, Gothenburg, Sweden. E-mail:
| |
Collapse
|
43
|
Li C, Hu Z. Is liver glycogen fragility a possible drug target for diabetes? FASEB J 2019; 34:3-15. [PMID: 31914592 DOI: 10.1096/fj.201901463rr] [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: 06/12/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/14/2022]
Abstract
Liver glycogen α particles are molecularly fragile in diabetic mice, and readily form smaller β particles, which degrade more rapidly to glucose. This effect is well associated with the loss of blood-glucose homeostasis in diabetes. The biological mechanism of such fragility is still unknown; therefore, there are perceived opportunities that could eventually lead to new means to manage type 2 diabetes. The hierarchical structures of glycogen particles are controlled by the underlying biosynthesis/degradation process that involves various enzymes, including, for example, glycogen synthase (GS) and glycogen-branching enzyme (GBE). Recent studies have shown that fragile glycogen α particles in diabetic mice have longer chains and a higher molecular density compared to wild-type mice, indicating an enhanced enzymatic activity ratio of GS to GBE in diabetes. Furthermore, it has been shown that with an improved blood glucose homeostasis, the glycogen fragility in diabetic mice can be restored by treatment with active ingredients from traditional Chinese medicine, yet the underlying mechanism is unknown. In this review, we summarize recent advances in understandings glycogen fragility from the perspectives of glycogen biosynthesis/degradation, glycogen hierarchical structures, and its relation to diabetes. Importantly, we for the first time set GS/GBE activity ratio as the therapeutic target for diabetes.
Collapse
Affiliation(s)
- Cheng Li
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China.,School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Zhenxia Hu
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
44
|
From the seminal discovery of proteoglycogen and glycogenin to emerging knowledge and research on glycogen biology. Biochem J 2019; 476:3109-3124. [DOI: 10.1042/bcj20190441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/10/2019] [Accepted: 10/14/2019] [Indexed: 11/17/2022]
Abstract
AbstractAlthough the discovery of glycogen in the liver, attributed to Claude Bernard, happened more than 160 years ago, the mechanism involved in the initiation of glucose polymerization remained unknown. The discovery of glycogenin at the core of glycogen's structure and the initiation of its glucopolymerization is among one of the most exciting and relatively recent findings in Biochemistry. This review focuses on the initial steps leading to the seminal discoveries of proteoglycogen and glycogenin at the beginning of the 1980s, which paved the way for subsequent foundational breakthroughs that propelled forward this new research field. We also explore the current, as well as potential, impact this research field is having on human health and disease from the perspective of glycogen storage diseases. Important new questions arising from recent studies, their links to basic mechanisms involved in the de novo glycogen biogenesis, and the pervading presence of glycogenin across the evolutionary scale, fueled by high throughput -omics technologies, are also addressed.
Collapse
|
45
|
Hedberg-Oldfors C, De Ridder W, Kalev O, Böck K, Visuttijai K, Caravias G, Töpf A, Straub V, Baets J, Oldfors A. Functional characterization of GYG1 variants in two patients with myopathy and glycogenin-1 deficiency. Neuromuscul Disord 2019; 29:951-960. [PMID: 31791869 DOI: 10.1016/j.nmd.2019.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 09/23/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022]
Abstract
Glycogen storage disease XV is caused by variants in the glycogenin-1 gene, GYG1, and presents as a predominant skeletal myopathy or cardiomyopathy. We describe two patients with late-onset myopathy and biallelic GYG1 variants. In patient 1, the novel c.144-2A>G splice acceptor variant and the novel frameshift variant c.631delG (p.Val211Cysfs*30) were identified, and in patient 2, the previously described c.304G>C (p.Asp102His) and c.487delG (p.Asp163Thrfs*5) variants were found. Protein analysis showed total absence of glycogenin-1 expression in patient 1, whereas in patient 2 there was reduced expression of glycogenin-1, with the residual protein being non-functional. Both patients showed glycogen and polyglucosan storage in their muscle fibers, as revealed by PAS staining and electron microscopy. Age at onset of the myopathy phenotype was 53 years and 70 years respectively, with the selective pattern of muscle involvement on MRI corroborating the pattern of weakness. Cardiac evaluation of patient 1 and 2 did not show any specific abnormalities linked to the glycogenin-1 deficiency. In patient 2, who was shown to express the p.Asp102His mutated glycogenin-1, cardiac evaluation was still normal at age 77 years. This contrasts with the association of the p.Asp102His variant in homozygosity with a severe cardiomyopathy in several cases with an onset age between 30 and 50 years. This finding might indicate that the level of p.Asp102His mutated glycogenin-1 determines if a patient will develop a cardiomyopathy.
Collapse
Affiliation(s)
- Carola Hedberg-Oldfors
- Department of Pathology and Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Willem De Ridder
- Neurogenetics Group, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Ognian Kalev
- Institute of Pathology, Kepler University Hospital, Neuromed Campus, Linz, Austria
| | - Klaus Böck
- Department of Neurology 1, Kepler University Hospital, Neuromed Campus, Linz, Austria
| | - Kittichate Visuttijai
- Department of Pathology and Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Georg Caravias
- Department of Neurology 1, Kepler University Hospital, Neuromed Campus, Linz, Austria; Department of Neurology 2, Kepler University Hospital, Linz, Austria
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Jonathan Baets
- Neurogenetics Group, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium; Neuromuscular Reference Centre, Department of Neurology, Antwerp University Hospital, Antwerp, Belgium
| | - Anders Oldfors
- Department of Pathology and Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
46
|
Abstract
Lafora disease is a severe, autosomal recessive, progressive myoclonus epilepsy. The disease usually manifests in previously healthy adolescents, and death commonly occurs within 10 years of symptom onset. Lafora disease is caused by loss-of-function mutations in EPM2A or NHLRC1, which encode laforin and malin, respectively. The absence of either protein results in poorly branched, hyperphosphorylated glycogen, which precipitates, aggregates and accumulates into Lafora bodies. Evidence from Lafora disease genetic mouse models indicates that these intracellular inclusions are a principal driver of neurodegeneration and neurological disease. The integration of current knowledge on the function of laforin-malin as an interacting complex suggests that laforin recruits malin to parts of glycogen molecules where overly long glucose chains are forming, so as to counteract further chain extension. In the absence of either laforin or malin function, long glucose chains in specific glycogen molecules extrude water, form double helices and drive precipitation of those molecules, which over time accumulate into Lafora bodies. In this article, we review the genetic, clinical, pathological and molecular aspects of Lafora disease. We also discuss traditional antiseizure treatments for this condition, as well as exciting therapeutic advances based on the downregulation of brain glycogen synthesis and disease gene replacement.
Collapse
|
47
|
Torres AG, Reina O, Stephan-Otto Attolini C, Ribas de Pouplana L. Differential expression of human tRNA genes drives the abundance of tRNA-derived fragments. Proc Natl Acad Sci U S A 2019; 116:8451-8456. [PMID: 30962382 PMCID: PMC6486751 DOI: 10.1073/pnas.1821120116] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The human genome encodes hundreds of transfer RNA (tRNA) genes but their individual contribution to the tRNA pool is not fully understood. Deep sequencing of tRNA transcripts (tRNA-Seq) can estimate tRNA abundance at single gene resolution, but tRNA structures and posttranscriptional modifications impair these analyses. Here we present a bioinformatics strategy to investigate differential tRNA gene expression and use it to compare tRNA-Seq datasets from cultured human cells and human brain. We find that sequencing caveats affect quantitation of only a subset of human tRNA genes. Unexpectedly, we detect several cases where the differences in tRNA expression among samples do not involve variations at the level of isoacceptor tRNA sets (tRNAs charged with the same amino acid but using different anticodons), but rather among tRNA genes within the same isodecoder set (tRNAs having the same anticodon sequence). Because isodecoder tRNAs are functionally equal in terms of genetic translation, their differential expression may be related to noncanonical tRNA functions. We show that several instances of differential tRNA gene expression result in changes in the abundance of tRNA-derived fragments (tRFs) but not of mature tRNAs. Examples of differentially expressed tRFs include PIWI-associated RNAs, tRFs present in tissue samples but not in cells cultured in vitro, and somatic tissue-specific tRFs. Our data support that differential expression of tRNA genes regulate noncanonical tRNA functions performed by tRFs.
Collapse
Affiliation(s)
- Adrian Gabriel Torres
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, 08028 Barcelona, Catalonia, Spain
| | - Oscar Reina
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, 08028 Barcelona, Catalonia, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, 08028 Barcelona, Catalonia, Spain
| | - Lluís Ribas de Pouplana
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, 08028 Barcelona, Catalonia, Spain;
- Catalan Institution for Research and Advanced Studies, 08010 Barcelona, Catalonia, Spain
| |
Collapse
|
48
|
Blackwood SJ, Hanya E, Katz A. Effect of postexercise temperature elevation on postexercise glycogen metabolism of isolated mouse soleus muscle. J Appl Physiol (1985) 2019; 126:1103-1109. [PMID: 30730817 DOI: 10.1152/japplphysiol.01121.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effects of temperature elevation after intense repeated contractions on glycogen and energy metabolism as well as contractile function of isolated mouse soleus muscle (slow twitch, oxidative) were investigated. Muscles were stimulated electrically to perform repeated tetanic contractions for 10 min at 25°C, which reduced tetanic force by ~85% and glycogen by 50%. After 120-min recovery at 25°C glycogen was fully restored (~125% of basal), whereas after recovery at 35°C glycogen decreased further (~25% of basal). Glycogen synthase fractional activity averaged 31.8 ± 3.1% (baseline = 33.8 ± 3.4%) after 120-min recovery at 25°C but was increased after recovery at 35°C (63.8 ± 4.8%; P < 0.001 vs. 25°C). Phosphorylase fractional and total activities were not affected by the higher temperature. However, recovery at 35°C resulted in a significantly higher content of the phosphorylase substrate inorganic phosphate (~20%; P < 0.01 vs. 25°C). Finally, fatigue development during a subsequent bout of repeated contractions at 25°C was similar after 120-min recovery at 25°C and 35°C. These data demonstrate that after intense contractions elevated temperature inhibits glycogen accumulation, likely by increasing the availability of the phosphorylase substrate inorganic phosphate, but has no effect on fatigue development. Thus after heat exposure phosphorylase plays a significant role in glycogen accumulation, and glycogen does not limit muscle performance in isolated mouse soleus muscle after recovery from elevated temperature. NEW & NOTEWORTHY Whether elevated temperature affects glycogen biogenesis and contractile performance of isolated slow-twitch muscle is not known. Here we show that after a bout of repeated contractions in isolated mouse soleus muscle at 25°C, increasing muscle temperature during recovery to 35°C blocked glycogen accumulation compared with recovery at 25°C. Surprisingly, during a subsequent bout of repeated contractions at 25°C, the rate of fatigue was not different between groups after recovery at the two temperatures.
Collapse
Affiliation(s)
- Sarah J Blackwood
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| | - Ester Hanya
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| | - Abram Katz
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
49
|
Wu L, Wong CP, Swanson RA. Methodological considerations for studies of brain glycogen. J Neurosci Res 2019; 97:914-922. [PMID: 30892752 DOI: 10.1002/jnr.24412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 01/02/2023]
Abstract
Glycogen stores in the brain have been recognized for decades, but the underlying physiological function of this energy reserve remains elusive. This uncertainty stems in part from several technical challenges inherent in the study of brain glycogen metabolism. These include low glycogen content in the brain, non-homogeneous labeling of glycogen by radiotracers, rapid glycogenolysis during postmortem tissue handling, and effects of the stress response on brain glycogen turnover. Here we briefly review the aspects of the glycogen structure and metabolism that bear on these technical challenges and present ways they can be addressed.
Collapse
Affiliation(s)
- Long Wu
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Candance P Wong
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Health Care System, San Francisco, California
| | - Raymond A Swanson
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Health Care System, San Francisco, California
| |
Collapse
|
50
|
Xu W, Zhou H, Xuan H, Saha P, Wang G, Chen W. Novel metabolic disorders in skeletal muscle of Lipodystrophic Bscl2/Seipin deficient mice. Mol Cell Endocrinol 2019; 482:1-10. [PMID: 30521848 PMCID: PMC6340772 DOI: 10.1016/j.mce.2018.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 01/23/2023]
Abstract
Bscl2-/- mice recapitulate many of the major metabolic manifestations in Berardinelli-Seip congenital lipodystrophy type 2 (BSCL2) individuals, including lipodystrophy, hepatosteatosis, muscular hypertrophy, and insulin resistance. Metabolic defects in Bscl2-/- mice with regard to glucose and lipid metabolism in skeletal muscle have never been investigated. Here, we identified Bscl2-/- mice displayed reduced intramyocellular triglyceride (IMTG) content but increased glycogen storage predominantly in oxidative type I soleus muscle (SM). These changes were associated with increased incomplete fatty acid oxidation and glycogen synthesis. Interestingly, SM in Bscl2-/- mice demonstrated a fasting duration induced insulin sensitivity which was further confirmed by hyperinsulinemic-euglycemic clamp in SM of overnight fasted Bscl2-/- mice but reversed by raising circulating NEFA levels through intralipid infusion. Furthermore, mice with skeletal muscle-specific inactivation of BSCL2 manifested no changes in muscle deposition of lipids and glycogen, suggesting BSCL2 does not play a cell-autonomous role in muscle lipid and glucose homeostasis. Our study uncovers a novel link between muscle metabolic defects and insulin resistance, and underscores an important role of circulating NEFA in regulating oxidative muscle insulin signaling in BSCL2 lipodystrophy.
Collapse
Affiliation(s)
- Wenqiong Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, PR China; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hongyi Zhou
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hongzhuan Xuan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA; School of Life Science, Liaocheng University, Liaocheng, Shandong Province, 252059, PR China
| | - Pradip Saha
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, PR China.
| | - Weiqin Chen
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|