Abstract
Glycogen is a biologically essential macromolecule that is directly involved in multiple human diseases. While its primary role in carbohydrate storage and energy metabolism in the liver and muscle is well characterized, recent research has highlighted critical metabolic and non-metabolic roles for glycogen in the brain. In this review, the emerging roles of glycogen homeostasis in the healthy and diseased brain are discussed with a focus on advancing our understanding of the role of glycogen in the brain. Innovative technologies that have led to novel insights into glycogen functions are detailed. Key insights into how the cellular localization impacts neuronal and glial function are discussed. Perturbed glycogen functions are observed in multiple disorders of the brain, including where it serves as a disease driver in the emerging category of neurological glycogen storage diseases (n-GSDs). n-GSDs include Lafora disease (LD), adult polyglucosan body disease (APBD), Cori disease, Glucose transporter type 1 deficiency syndrome (G1D), GSD0b, and late onset Pompe disease (PD). They are neurogenetic disorders characterized by aberrant glycogen which results in devastating neurological and systemic symptoms. In the most severe cases, rapid neurodegeneration coupled with dementia results in death soon after diagnosis. Finally, we discuss current treatment strategies that are currently being developed and have the potential to be of great benefit to n-GSD patients. Taken together, novel technologies and biological insights have resulted in a renaissance in the brain glycogen that dramatically advanced our understanding of both biology and disease. Future studies are needed to expand our understanding and the multifaceted roles of glycogen and effectively apply these insights in human disease.
Keywords: Glycogen storage disease, Congenital disorders of glycosylation, Glycogen, Epilepsy, N-linked Glycosylation, Brain metabolism
Graphical Abstract

Glycogen is an essential carbohydrate storage molecule involved in multiple tissue-specific processes. This review provides a comprehensive summary of our understanding of brain glycogen and the multifaceted roles of glycogen proposed by recent articles. The roles are described in health and disease with a focus on current evidence linking glycogen and glycosylation. Finally, we highlight the therapeutic potential of targeting glycogen metabolism and potential avenues for future research.
Glycogen is one of the fundamental biological molecules and is critical for primary carbohydrate storage, metabolism, and emerging novel roles. Its central role in energy homeostasis has long been studied (Bernard 1857; Foster 1899; Carpenter, Smith, and Meneses 1876). Glycogen is found in high levels in the liver and muscle, where it serves as systemic and local carbohydrate reserves. However, the machinery for glycogen synthesis and degradation is widely expressed throughout the body, and other tissues and organs have been increasingly recognized to critically depend on cellular glycogen (Brewer and Gentry 2019a; Dienel and Carlson 2019; Obel et al. 2012). Through the lenses of newly developed technologies, spatially unique glycogen distribution in the brain, lung, testis, kidney, and bone marrow has been increasingly studied (Oe et al. 2016; Young et al. 2020; 2022; Sun et al. 2019; 2021).
Interestingly, glycogen displays heterogeneous patterns of distribution within the brain (Oe, Akther, and Hirase 2019) and is observed in diverse cell types within the central nervous system (CNS), including neurons, astrocytes, and microglia (A. M. Brown and Ransom 2007; Hertz, Peng, and Dienel 2007). This has significance for key aspects of brain biology as well as implications for diseases that affect the brain. Currently, our understanding of brain glycogen includes its pivotal roles in learning and memory, signaling events, neurotransmitter metabolism, and protein glycosylation. Here, we review multiple roles of glycogen in the brain. We also highlight key new technologies and insights into the biology of glycogen that may have profound implications for the brain. Finally, we discuss the role of glycogen in neurological disorders and address therapeutic strategies targeting glycogen metabolism to alleviate major symptoms of neurological disorders where glycogen is the primary disease driver.
Brain Glycogen – architecture
Glycogen consists of glucose molecules linked via glycoside bonds with α-1,4 chains and α-1,6 branches. This design allows the storage of up to ~50,000 glucose units (Shearer and Graham 2002; Meléndez, Meléndez-Hevia, and Cascante 1997; Marchand et al. 2002). Glycogen synthase is responsible for elongation of the α-1,4 chains and glycogen branching enzyme catalyzes the α-1,6 branches. Conversely, glycogen degradation in the cytosol is carried out by glycogen debranching enzyme and glycogen phosphorylase, while lysosomal glycogen is degraded by glucosidases.
In addition to glucose moieties, recent work revealed other key covalent components in brain glycogen. Brain glycogen contains covalently attached phosphate. Glycogen phosphate content is regulated by laforin, a glycogen phosphatase (Tagliabracci et al. 2007; Gentry, Worby, and Dixon 2005; Raththagala et al. 2015). More recently, work revealed the incorporation of monosaccharides other than glucose in brain glycogen, which is discussed in more detail below. Glycogen structure is defined by branching pattern, glucose chain length, and composition including covalent phosphate (Brewer and Gentry 2019b). Differences in these key factors modulate glycogen solubility, accessibility to interacting enzymes, and granular size (Brewer and Gentry 2019a; Gentry et al. 2018). Multiple glycogen storage disorders (GSDs) present with increased phosphate content and/or abnormal branching and chain elongation (Sullivan et al. 2019; Nitschke et al. 2017; B. I. Brown and Brown 1966; Mercier and Whelan 1973). Tissue- and context-specific glycogen architecture with respect to biology and disease remains a critical knowledge gap in the field.
Brain Glycogen – distribution
Determination of glycogen distribution in the brain was previously challenging due to technical limitations and the detection limits of methods available at the time. However, the development of specific glycogen antibodies and improved rapid fixation methods have allowed visualization of brain glycogen in two dimensional space with cellular details (Oe et al. 2016; Oe, Akther, and Hirase 2019; Baba 1993). Brain glycogen distribution exhibits high spatial and cellular heterogeneity, being present in the hippocampus, cerebellum, layers of the neocortex, striatum, substantia nigra, and pons; regions surrounding the vasculature, and white matter regions such as the hypothalamus and thalamus (Oe, Akther, and Hirase 2019; A. Brown, Baltan, and Ransom 2009; A. M. Brown, Rich, and Ransom 2019). Interestingly, glycogen distribution exhibits spatial heterogeneity within white and gray matter regions, which may be coupled to functional differences. This spatial heterogeneity raises interesting questions regarding the integrative roles of glycogen in these respective brain regions.
Glycogen is observed in the cytosol of multiple cell types in the brain. Astrocytes store the largest quantities of glycogen, with neurons and microglia also observed to have significant glycogen stores (Saez et al. 2014; Oe et al. 2016). Of note, neurons possess the enzymatic machinery for glycogen metabolism and have been shown to actively engage in glycogen turnover. Indeed, neuronal glycogen is essential for neuronal survival under hypoxic conditions in vitro, and it plays important roles for memory formation and hypoxia tolerance in vivo (Duran et al. 2013; Saez et al. 2014). However, the relative fraction of glycogen and glycogen turnover in neurons and glia cells remains to be further investigated.
Interestingly, brain glycogen can be further compartmentalized on a subcellular level and the concentration and turnover may vary according to specific compartments. For instance, elevated glycogen has been observed in the brain in cellular processes including synaptic endfeet (Dienel and Carlson 2019; Richter, Hamprecht, and Scheich 1996; Pfeiffer-Guglielmi et al. 2003; Bak and Walls 2018). In peripheral organs, glycogen is also compartmentalized at the subcellular level, with glycogen being identified in the nucleus, mitochondria, and in close proximity to the endoplasmic reticulum (Baird and Fisher 1957; De Man, Blok, and Beens 1966; Cardell 1977; Nielsen et al. 2010; Ishikawa and Pel 1965). The first description of nuclear glycogen was reported in hepatocytes in the 1940s and subsequently confirmed by multiple laboratories (Chipps and Duff 1942; Baird and Fisher 1957; Bogoch et al. 1955). The recently identified role of nuclear glycogen in histone acetylation suggests that subcellular localization of glycogen plays key roles in fundamental biology which have not been previously appreciated (Sun et al. 2019). To this end, the subcellular localization of glycogen is an exciting area for future scientific advances and remains to be further elucidated within brain cells.
Brain Glycogen – detection and localization
Multiple methods are widely utilized to analyze and quantify glycogen, including periodic acid-Schiff (PAS) staining, alcohol or trichloroacetic acid (TCA) precipitation, followed by biochemical enzymatic assay, and electron microscopy (EM) (Figure 1) (Brewer and Gentry 2019a). Glycogen was first purified by ethanol and TCA precipitation from 1909 to the 1930s, but this method required a substantial amount of tissue (Pflüger 1909; Somogyi 1934; Willstätter and Rohdewald 1934). In the 1930s-1940s, direct visualization of glycogen was achieved through the implementation of electron microscopy (EM). EM provided key insights into glycogen structure/architecture (scanning EM) and sub-cellular locations within a cell (transmission EM).
Figure 1: Glycogen and glycan detection methods.
Overview of common methods to detect glycogen and glycans, including overlapping methods. MALDI-MSI can be utilized to detect both simultaneously. Created with BioRender.com. Abbreviations: EM – electron microscopy, PAS – periodic acid-Schiff, HPAEC-PAD – high performance anion-exchange chromatography with pulsed amperometric detection, GCMS – Gas chromatography mass spectrometry, MALDI-IMS – Matrix-assisted laser desorption/ionization imaging mass spectrometry, MS – mass spectrometry, SDS-PAGE – sodium dodecyl sulfate–polyacrylamide gel electrophoresis, NMR – nuclear magnetic resonance.
One of the first techniques to provide histological annotation of glycogen came with the development of PAS staining by McManus in 1946 (McManus 1946; 1948; Aterman and Norkin 1963). PAS is easy to perform, and coupled with a rapid turnaround time, it is an ideal method for clinical pathology. Even today, PAS is utilized in clinical settings for the diagnosis of diverse diseases, including Ewing’s sarcoma (Mizuguchi et al. 2016; Hartman et al. 1991). As a research tool, PAS staining has several limitations due to low specificity caused by reactivity with a wide range of polysaccharides, including glycoproteins, glycolipids, glycogen, and mucins. Additionally, while PAS in combination with diastase treatment (amylase) can improve selectivity, the technique is not sensitive enough to detect physiological glycogen levels in many tissues and cell types.
The development of glycogen antibodies dramatically improved our knowledge of regional glycogen distribution (Baba 1993; Nakamura-Tsuruta et al. 2012). Two mouse monoclonal IgM glycogen antibodies, IV58B6 and ESG1A9, have been verified to detect glycogen by immunohistochemical methods. While both antibodies detect glycogen, ESG1A9 possesses higher affinity for larger glycogen molecules, and IV58B6 displays a more uniform affinity for glycogen of all sizes (Oe, Akther, and Hirase 2019; Oe et al. 2016). Additionally, brain glycogen is difficult to quantify and study due to its rapid post-mortem degradation (Wu, Wong, and Swanson 2019; Pontén, Ratcheson, and Siesjö 1973). In fact, research on glycogen extraction methods and degradation time suggest that up to 95% of glycogen may be degraded during improper experimental- and extraction conditions (Cruz and Dienel 2002; Dienel 2020; Hsu et al. 2021). The implementation of focused microwave fixation in situ has provided a significant advantage for the accurate detection of physiological glycogen compared to other approaches (Oe et al. 2016; Juras et al. 2022).
Importantly, recent technological advances are allowing multi-scale definition of the cellular, spatial, and systemic metabolism of the central nervous system. Multiple techniques for accurate and sensitive glycogen detection have been established and widely implemented to measure glycogen from mouse to man. These techniques include gas chromatography mass spectrometry (GCMS), matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI), magnetic resonance spectroscopy (MRS), and magnetic resonance imaging (MRI) (Figure 1) (Soares, Gruetter, and Lei 2017). The development of GCMS-based methods to detect glycogen dramatically improved accuracy and sensitivity (Young et al. 2020; Andres et al. 2020). MALDI-MSI combines the accuracy and sensitivity of mass spectrometry with added spatial information and glycogen architecture (Conroy, Allison, et al. 2021; Young et al. 2022; Hawkinson and Sun 2022). This technique offers crucial quantification of regional- and architectural information and is approaching single-cell resolution. While MS-based assays focus on endpoint analysis, glycogen quantitation in living organisms is made possible through recent development in MRI (Y. Zhou et al. 2020). While having reduced sensitivity compared to MS-based methods, these methods have the significant advantage of being non-invasive and applicable to imaging the entire human brain, allowing live imaging of glycogen. One such MRI method relies on magnetic coupling between glycogen and water through nuclear Overhauser enhancement (i.e., glycoNOE), and represents a promising technique for glycogen detection and treatment evaluation for GSDs (Y. Zhou et al. 2020). Collectively, utilizing these newer techniques will be crucial for deepening our understanding of brain glycogen with respect to spatial resolution and imaging glycogen in live organisms.
The pleiotropic roles of glycogen
The brain is an energy-demanding organ relying on a continuous supply of glucose (Bordone et al. 2019). Interestingly, the brain is also uniquely susceptible to damage caused by fluctuating glucose and oxygen levels. Brain glycogen promptly responds to changing cerebral energy demands and is degraded for energy production as an emergency fuel in response to low glucose levels. However, brain glycogen cannot sustain metabolic demands for long due to the high energy demands of the brain and low glycogen levels. Illustrating this, glycogen is very rapidly degraded post-mortem (Dienel and Carlson 2019; Dienel 2019b; Pontén, Ratcheson, and Siesjö 1973; Wu, Wong, and Swanson 2019). Furthermore, brain glycogen quickly reacts to changing energy demands and is rapidly degraded during a hypoglycemic episode or ischemia in both astrocytes and neurons (Guo et al. 2021; Bastian et al. 2019; Dienel and Rothman 2019). Importantly, neurons possess active glycogen metabolism proteins, and these enzymes are needed to protect neurons from hypoxia (Saez et al. 2014). Indeed, cultured neurons both de-novo synthesize and degrade glycogen in response to hypoxia by activation of glycogen synthase and glycogen phosphorylase (Vilchez et al. 2007). Additionally, it has been increasingly appreciated in recent years that glycogen has pleiotropic roles that support normal brain function, and dysregulation of glycogen metabolism contributes to multiple diseases (Waitt et al. 2017; Obel et al. 2012; Bak and Walls 2018; Dienel 2019b; DiNuzzo and Schousboe 2019) (Figure 2).
Figure 2: Integrated and proposed novel roles of Glycogen.
Glycogen is a diverse molecule rapidly responding to changing energy levels and serve multiple vital functions in several bodily organs. Created with BioRender.com.
Further, glycogen can be utilized in a coupled multi-cellular context. Work by Magistretti and colleagues demonstrated a relationship between glycogen and lactate, suggesting that astrocytes degrade glycogen to produce lactate for neuronal energy demands known as the Astrocyte-Neuron-Lactate-Shuttle (ANLS) (Magistretti and Allaman 2015) (Figure 3). However, the ANLS remains a controversial hypothesis due to stoichiometric disagreements and the ability of neurons to utilize lactate. Thus, other hypotheses explaining the relationship between glucose/glycogen metabolism and neurotransmission have been proposed, including the Glucose Sparing by Glycogenolysis (GSG) hypothesis (Rothman et al. 2022; Dinuzzo et al. 2012; Swanson 1992). The GSG hypothesis suggests that astrocytes utilize glycogen to spare glucose and thereby increase glucose availability for neurons during high energy demands. This hypothesis also provides an explanation for the stoichiometry and energy demands of the astrocytic glutamate/GABA oxidation that the ANLS does not fully explain. Yet, both hypotheses include limitations that must be further elucidated. Collectively, the complexity of metabolic pathways and interrelationship between neurons and astrocytes awaits further investigation, but the central importance of glycogen in neurons and astrocyte remains clear (Dienel 2019b; Bélanger, Allaman, and Magistretti 2011).
Figure 3. Glycogen is a critical component of cerebral metabolism.
Glycogen metabolism directly supports neuronal and astrocytic function. Glycogen is also found in microglia. Glycogen supports brain energy metabolism, but controversies exist regarding the underlying mechanisms and energetic coupling between neurons and astrocytes. Created with BioRender.com. Abbreviations: Glu – glutamate, gln – glutamine, ATP – adenosine triphosphate.
Cognitive function and memory formation
The earliest descriptions of glycogen in memory and learning were demonstrated in chickens and based on impaired learning after injections of a glycogen phosphorylase inhibitor 1,4-dideoxy-1,4-imino-d-arabinitol (DAB) (Gibbs, Anderson, and Hertz 2006). These findings have been supported by other studies demonstrating that impaired memory and learning in rat astrocytes and neurons, utilizing DAB or lactate transporter inhibition/deletion to perturb mobilization of astrocytic glycogen-derived lactate (Alberini et al. 2018). Furthermore, multiple other studies suggest that astrocytes contribute to memory formation as well and are crucial for reward location (Doron et al. 2022). While physiologically important, mechanistic insights were limited, thus leaving knowledge gaps regarding the corroboration of cell types. Emerging evidence supports a role for both astrocytic and neuronal glycogen in memory formation that calls for further investigation (Duran et al. 2020; Duran, Gruart, Varea, et al. 2019).
Astrocytic glycogen can support multiple functions, including lactate release, glutamate uptake, and astrocytic glutamine synthesis (Suzuki et al. 2011; Sickmann et al. 2009; Andersen and Schousboe 2022). Glutamine is a precursor for glutamate, and biosynthesis of glutamine occurs primarily in astrocytes and can be synthesized from glycogen. Interestingly, pharmacological inhibition of glutamine synthesis resulted in glycogen accumulation in astrocytes and perturbed memory consolidation in chickens and mice (Folbergrová et al. 1969; Swanson et al. 1989; Gibbs, Anderson, and Hertz 2006). Additionally, glutamine supplementation can improve impaired memory and learning caused by perturbed glycogen metabolism in chickens (Gibbs et al. 2007; Gibbs, Anderson, and Hertz 2006). Thus, glutamine is intimately linked to memory formation and glutamatergic neurotransmission awaiting further elucidation.
Glutamate is the major excitatory neurotransmitter in the brain, and glutamatergic neurotransmission is closely coupled to energy metabolism and tightly regulated to maintain homeostasis and prevent seizure generation (Bordone et al. 2019). Multiple laboratories demonstrated that glutamatergic neurotransmission is coupled to glycogen metabolism by employing inhibitors of glycogen metabolism enzymes in cultures, co-cultures, and slices (Dienel 2019b; Sickmann et al. 2009; Drulis-Fajdasz et al. 2015; Mozrzymas, Szczęsny, and Rakus 2011). Indeed, it is well-established that inhibition of glycogenolysis results in disrupted excitatory signaling and impaired long-term potentiation (Suzuki et al. 2011; Mozrzymas, Szczęsny, and Rakus 2011; Duran, Gruart, Varea, et al. 2019). However, the mechanism for synaptic perturbations caused by inhibition of glycogenolysis is not fully elucidated and has been linked to both neuronal and astrocytic processes.
Astrocytes provide another link to glycogen and glutamatergic neurotransmission through their clearance of extracellular K+. Strikingly, astrocytic clearance of extracellular K+ directly causes glycogen degradation. The current model for this degradation is the energy demands of the Na+/K+-ATPase that stimulates glycogenolysis and respiration for ATP production (Hof, Pascale, and Magistretti 1988; Xu et al. 2013; Choi et al. 2012). Thus, clearance of K+ depends on glycogen, and inability in clearance of K+ could lead to seizure generation and hyperexcitability (Hof, Pascale, and Magistretti 1988; Choi et al. 2012). Taken together, glycogen directly impacts glutamatergic neurotransmission by multiple processes, including providing substrate for glutamate production and K+ clearance.
More recent, studies with transgenic models have demonstrated that genetically altered mice devoid of CNS glycogen, i.e., Nestin-Cre-Gys1 knockout, survive and have a normal lifespan, but present with impaired memory formation and long-term memory consolidation (Duran et al. 2013; Gibbs, Anderson, and Hertz 2006). Follow-up studies based on genetic manipulation of neuronal glycogen metabolism demonstrated that animals devoid of glycogen in hippocampal neurons have impaired synaptic plasticity and memory formation, but did not exhibit increased susceptibility to hippocampal seizures (Duran, Gruart, Varea, et al. 2019). Additionally, studies utilizing genetic strategies to specifically target and ablate neuronal and astrocytic glycogen suggested that astrocytes are more linked to neurodegeneration and inflammation, while neurons contribute more to epileptic seizure susceptibility (Duran et al. 2021; Duran, Gruart, López-Ramos, et al. 2019). Thus, data from these genetic models suggest neuronal glycogen as a primary contributor to glycogen-derived memory perturbations but the potential adaptation occurring in transgenic mice should be kept in mind.
Thermoregulation, viscosity, and phase separation
Glycogen as a macromolecule has also been shown to contribute to diverse biological processes directly affecting intermolecular processes and homeostasis, including osmolarity, crowding, temperature regulation, and viscosity (N A Chebotareva 2007; Natalia A Chebotareva et al. 2004; Persson, Ambati, and Brandman 2020; Kerly 1929). Early studies determined that the viscosity of a liquid decreases in response to increasing temperatures (Longsworth 1954). This decreased viscosity at higher temperatures consequently leads to increased diffusion rates and molecular interaction frequency, affecting signal transduction and kinetics of complex assembly (Longsworth 1954; M. E. Johnson 2018; Stojanovski et al. 2017). Interestingly, glycogen increases heat stress tolerance, analogous to homeoviscous adaptation in the cell membrane (Wilson, Wang, and Roach 2002; Persson et al. 2020).
Work in the 1970s, pioneered by Sinensky and colleagues, elegantly demonstrated that cells adapt membrane lipid content in response to temperature changes and thereby allow the cell to control intermolecular interactions (Budin et al. 2018; Sinensky 1974). However, the mechanism for thermoregulation and viscosity in the cytosol remained unclear until it was recently demonstrated that glycogen and trehalose, a disaccharide of glucose, contribute to these processes (Persson, Ambati, and Brandman 2020). Through a combination of in vitro and in vivo techniques, it was demonstrated that yeast cells adjust glycogen and trehalose synthesis in response to temperature changes to prevent viscosity perturbations and maintain constant diffusion rates. Furthermore, data suggest that this viscoadaption impacts phase separation and the solubility of biomolecules in the cell. Indeed, it has been reported that glycogen undergoes liquid-liquid phase separation, creating large dynamic droplets in vitro that require the glycogen phosphatase laforin for activation of cell survival pathways (Liu et al. 2021). Collectively, glycogen appears to be crucial for thermoregulation, viscosity, and phase separation which may have specific impacts in the brain.
Glycogen in gene expression via histone acetylation
The nucleus is a highly specialized cellular organelle in eukaryotes that serves as a key cellular processing and DNA regulatory center. Histone acetylation in the nucleus impacts cell proliferation and growth, which are well-established hallmarks of aberrant cancer metabolism (Gujral et al. 2020; Unnikrishnan, Gafken, and Tsukiyama 2010; Z. Zhou et al. 2019; Zois, Favaro, and Harris 2014). Strikingly, increased glycogen in the nucleus is also a hallmark of some cancers, as first reported nearly 80 years ago by microscopy studies (Chipps and Duff 1942). However, the physiological function of nuclear glycogen remained largely unknown until recent work demonstrated that nuclear glycogen is linked to histone acetylation (Sun et al. 2019; Dong 2019; Donohue, Gentry, and Sun 2020). It was demonstrated that de novo nuclear glycogen synthesis and the metabolic fate of the glycogenolysis product, pyruvate, directly controls epigenetic regulation through histone acetylation. Furthermore, decreased levels of the E3 ubiquitin ligase malin impaired nuclear glycogenolysis by directly decreasing glycogen phosphorylase levels in the nucleus, causing nuclear glycogen accumulation (Sun et al. 2019; Donohue, Gentry, and Sun 2020). This novel molecular mechanism and role for nuclear glycogen has fascinating implications for brain function and is the subject of ongoing research.
Glycogen and protein glycosylation
Protein glycosylation is a highly regulated posttranslational modification that is critical for multiple cellular functions including protein folding, cell-cell interactions, cell adhesion, proliferation, and inflammation (Schwarz and Aebi 2011; Ng and Freeze 2018; Sun et al. 2021). Its importance is emphasized by the fact that >2% of the genome encodes genes involved in glycan metabolism, and >100 human diseases arise from glycosylation defects (Sun et al. 2021; Reily et al. 2019; Conroy, Hawkinson, et al. 2021; Ng and Freeze 2018). Glycogen and N-linked glycans share significant biochemical connections. Both are branched polysaccharides required for life which share the common substrate UDP-glucose. Additionally, glycogenolysis has been shown to preferably supply the hexosamine pathway for the biosynthesis of UDP-GlcNAc. Finally, brain glycogen has been shown to be a rich source of glucosamine, that can be directly converted to UDP-GlcNAc upon release from glycogen (Sun et al. 2021).
Asparagine- or N-linked glycosylation is the most common form of protein glycosylation (Eichler 2019). The most abundant extracellular monosaccharides and building blocks for glycosylation in the brain are glucose, glucosamine, fructose, mannose, fucose, and galactose. These monosaccharides enter cells by facilitated diffusion through the glucose transporter (GLUT) family of membrane transporters encoded in the SLC2 genes. Historically, the chemical heterogeneity of glycosylation has proven to be a barrier to study. However, emerging methods utilizing mass spectrometry and in situ enzymatic degradation of glycans known as MALDI-MSI provides improved sensitivity and spatial resolution (McDowell et al. 2021; Blaschke et al. 2020; Powers et al. 2013; Drake et al. 2018; Stanback et al. 2021; Hawkinson and Sun 2022).
Early studies suggested that muscle and liver glycogen contain small amounts of covalently bound glucosamine and provided an early connection to protein glycosylation. These studies utilized radiolabeled glucosamine and galactosamine to prove that glycogen synthase catalyzes the in vitro incorporation of UDP-glucosamine into glycogen (Tarentino and Maley 1976; Maley, McGarrahan, and DelGiacco 1966). Furthermore, Kirkman and Whelan demonstrated that glucosamine is a normal, albeit <1%, component of pig and rabbit liver glycogen, where it replaces α-1,4-linked glucose residues (Kirkman and Whelan 1986). More recently, a key pathway in brain carbohydrate metabolism that directly connects brain glycogen and N-linked protein glycosylation has been reported (Sun et al. 2021). By a multidisciplinary approach, it was demonstrated that N-linked protein glycosylation in the brain is critically and directly connected to glycogenolysis via the metabolite glucosamine. Strikingly, brain glycogen is composed of 25% glucosamine, while liver and muscle glycogen contain <1% glucosamine. Indeed, cells in the CNS leverage glycogen-associated glucosamine as a substantial substrate pool to produce UDP-acetylglucosamine (UDP-GlcNAc) and modulating glycogenolysis impacts N-linked glycan biosynthesis. Inhibiting glycogen utilization restricted UDP-GlcNAc pools and thereby total N-linked glycan levels in the brain. This mechanism was confirmed both in vitro and in vivo using two separate genetic mouse models lacking different glycogen metabolism enzymes. This study biochemically demonstrated that glucosamine is incorporated into glycogen by glycogen synthase and released from glycogen as glucosamine-1-phopshate by glycogen phosphorylase and glycogen debranching enzyme. The development of innovative multiplexed MALDI-MSI techniques allowed simultaneous visualization of glycogen and N-linked glycans in multiple disease models and revealed increases in glycogen and decreases in N-linked glycans (Conroy, Stanback, et al. 2021; Young et al. 2022). This technology provides an exciting new avenue to study spatial brain metabolism in the context of health and diseases.
It is well-established that both glycogen metabolism and N-linked protein glycosylation are crucial for synaptic modeling and memory formation. Perturbations in either pathway delay neuronal outgrowth and initiate neuro-inflammation that leads to cognitive delay, memory loss, and epilepsy (Dienel 2019a; Scott and Panin 2014; Gibbs, Anderson, and Hertz 2006). In fact, multiple disorders are either directly caused by or have perturbed glycosylation as a hallmark of the disease, including congenital disorders of glycosylation (CDG), some cancers, Alzheimer’s disease, Pompe disease, and Lafora disease (Gaunitz, Tjernberg, and Schedin-weiss 2020; Hawkinson et al. 2021; Sun et al. 2021; de-Souza-Ferreira, Ferreira, and de-Freitas-Junior 2023; Ng and Freeze 2018; Conroy, Stanback, et al. 2021; Wens et al. 2014). Strikingly, there are clinical studies reporting N-linked hypo-glycosylation in GSD patients; and aberrant glycogen aggregates in CDG patients (Papi et al. 2023; Sun et al. 2021; Tegtmeyer et al. 2014; McMahon and Frost 1996). Indeed, aortic stiffness in Pompe disease is caused by advanced glycan formation in aortic smooth muscle (Wens et al. 2014). Furthermore, multiple GSD and CDG patients share many neurological symptoms, including epilepsy and cognitive impairment (Piedade et al. 2022; Nitschke et al. 2018b; Korlimarla et al. 2019). While the novel route of hexosamine metabolism in the brain provides a direct metabolic connection between glycogen and N-glycans, key insights remain to be determined. Excitingly, these novel avenues of research have the potential to be therapeutically actionable for key glycogen-driven diseases of the CNS.
Glycogen in disease
GSDs are a group of inherited metabolic disorders and the first subtype was described in 1929 by von Gierke (Moses 2002; von Gierke 1929). This initial description of two patients with increased renal glycogen was later named GSD type I (Gierke’s disease), and more than 16 types of GSDs have subsequently been identified (Yeo, Moawad, and Grunewald 2023; Ellingwood and Cheng 2018). GSDs are a heterogeneous group of diseases driven by pathogenic glycogen with a wide range of symptoms, clinical onset, and life expectancy.
GSDs are caused by mutations in a wide range of genes encoding glycogen metabolism enzymes, leading to glycogen accumulation in most of the diseases (Figure 4A). However, there are at least two known GSDs that do not present with increased glycogen levels, including GSD type 0 (GSD0) and Glucose transporter 1 (Glut1) deficiency syndrome (G1D). These diseases are caused by mutations in genes encoding glycogen synthase and Glut1, preventing normal production of glycogen from glucose (Rajasekaran et al. 2023; Yeo, Moawad, and Grunewald 2023; Ellingwood and Cheng 2018). GSD0 and G1D mouse brains exhibit decreased glycogen and have similar symptoms to other GSDs (Rajasekaran et al. 2023; Chen and Weinstein 2016).
Figure 4. Neurological glycogen storage diseases and emerging treatments.
Overview of neurological glycogen storage diseases (n-GSDs) and key treatment strategies under development. A) Schematic representation of glycogen metabolism with affected enzymes in n-GSD and disease names. B) Current clinical status of treatment strategies for Lafora and Pompe disease and administration route for each treatment with green check signifying published and/or presented work and yellow check signifying ongoing investigation. ASOs targeting GYS1 have been tested via IT delivery for Lafora disease and siRNAs targeting GYS1 have been tested via IV delivery for Pompe disease. Preclinical work using AEFs has been performed for both diseases with multiple administration routes tested for Lafora disease. Created with BioRender.com. Abbreviations: ASO – anti-sense oligonucleotides, AEF – antibody-enzyme fusion, IT – Intrathecal, ICV – Intracerebroventricular, IV – Intravenous, siRNA – Small interfering ribonucleic acid, AAV – Adeno-associated virus, LD – Lafora disease, G1D – glucose transporter type 1 deficiency syndrome, Glc– glucose, UDP-Glc – uridine diphosphate glucose, P – phosphate, G1P – glucose-1-phosphate, G6P – glucose-6-phosphate, PGM1 – phosphoglucomutase 1, G6PASE - glucose-6-phosphatase, GSD0 – glycogen storage disease type 0, GP – glycogen phosphorylase, GYS – glycogen synthase, LA – laforin, GDE – glycogen debranching enzyme, GBE – glycogen branching enzyme, APBD – adult polyglucosan body disease.
Neurological glycogen storage diseases (n-GSDs)
GSDs can be classified by the primary affected organ. n-GSDs primarily affect the brain and CNS. Further, a number of disorders caused by inborn errors of metabolism present with a wide range of neurological symptoms, with disease severity ranging from mild delay in intellectual development to severe seizures, loss of cognition, and death. Additionally, it is increasingly recognized that a subset of inborn errors of metabolism share the pathogenic feature of aberrant glycogen-like aggregates and overlap with known n-GSDs. These diseases are particularly deadly and provide a unique window into metabolic perturbations in the nervous system.
Collectively n-GSDs, including Lafora disease (LD) (Gentry et al. 2018), adult polyglucosan body disease (APBD) (Zebhauser et al. 2022), Cori disease (Hobson-Webb et al. 2010), Glucose transporter type 1 deficiency syndrome (G1D) (Wang, Pascual, and De Vivo 1993), GSD0b, and late onset Pompe disease (PD) (Korlimarla et al. 2019), are neurogenetic disorders that are clinically characterized by aberrant glycogen most commonly manifested as pathogenic glycogen-like accumulations termed polyglucosan bodies (PGB) or dramatically decreased levels of glycogen in the CNS (Cafferty et al. 1991; Nitschke et al. 2018a). n-GSD patients can display progressive paraparesis, sensory deficit, peripheral neuropathy, myoclonic seizure and in the most severe cases, rapid neurodegeneration coupled with dementia and death can occur within 5 years of disease onset (Nitschke et al. 2018a; Zebhauser et al. 2022; Boulan-Predseil et al. 1995). Many n-GSD patients are adolescents and young adults and thus far there is no effective cure or targeted therapy. These diseases highlight the importance of brain glycogen, and research to date has provided a unique window into studying how neurological glycogen impacts the CNS in health and disease (Figure 4).
Glycogen as a therapeutic target
Given the severity and consequences of perturbed glycogen and glycan metabolism, several treatment strategies are being developed (Liu et al. 2021). Multiple strategies are being pursued that directly target enzymes involved in glycogen metabolism to prevent glycogen accumulation or promote degradation of aberrant glycogen aggregates. Further, a wide range of therapeutics platforms and techniques are being explored, including small molecule, biologic, and genetic approaches (Brewer et al. 2019; Ahonen et al. 2021; Gumusgoz et al. 2022; Markussen et al. 2021; Tang et al. 2020; Austin et al. 2019a). While some strategies remain in early preclinical drug development stages, a few treatments have already provided encouraging data in clinical trials (Figure 4B).
Small molecule inhibitors of glycogen synthase are one of the very promising therapies being translated into the clinical. These substrate reducing therapies (SRTs) inhibit GYS1 and decrease glycogen levels. The small molecule MZE001 from Maze Therapeutics recently completed Phase I clinical trials (NCT05249621). The selective inhibition of muscle glycogen synthase by MZE001 renders it a promising drug for Pompe disease and multiple other GSDs. MZE001 was granted Orphan Drug Designation by the FDA, and initial reports from a large Phase I are promising (Therapeutics 2022). However, any SRT must cross the blood brain barrier for therapeutic efficacy in n-GSD therapeutics.
Anti-sense oligonucleotides (ASOs) targeting GYS1 developed in collaboration with Ionis Pharmaceuticals have also displayed promising results in pre-clinical Lafora disease models (Ahonen et al. 2021). The GYS1 ASO reduced brain glycogen synthase levels in both young and old LD mouse models, halted aberrant glycogen formation, and reduced neuroinflammation. In this study, the earlier treatment in younger mice provided the most robust response. Similarly, the GYS1 siRNA ABX1100 from Aro Biotherapeutics received FDA orphan drug designation for treatment of Pompe disease (Beyer 2022).
Antibody Enzyme Fusions (AEFs) are another promising class of drugs under development for GSDs from Parasail, LLC (Brewer et al. 2019; Austin et al. 2019a). VAL-0417 is a promising AEF comprised of a cell-penetrating antibody fragment (Fab) fused with pancreatic α-amylase, allowing in vivo degradation of glycogen aggregates and normalization of both brain metabolism and N-linked glycosylation (Sun et al. 2021). Importantly, the utilized VAL-0417 antibody fragment efficiently delivers the drug by entering cells via the equilibrative nucleoside transporter 2 (ENT2, SLC29A2) and localizes to the cytosol and nucleus (Hansen et al. 2013; Weisbart et al. 2012; Hansen, Weisbart, and Nishimura 2005; Weisbart et al. 2015; Hansen et al. 2007). Since ENT2 is ubiquitously expressed on cell membranes in mice and humans, VAL-0417 has potential applications in multiple diseases (Crawford et al. 1998; Lu, Chen, and Klaassen 2004; Hansen et al. 2007). As with other modalities, key aspects of brain delivery remain to be resolved for clinical translation, with initial data demonstrating that both intrathecal and intracerebroventricular administration of VAL-0417 is effective (Austin et al. 2019b). AEF therapy may be particularly beneficial for late-stage symptomatic patients who require degradation of already formed aggregates (Brewer et al. 2019; Austin et al. 2019a; Sun et al. 2021).
Adeno-associated virus (AAV)-mediated gene therapy is a promising approach for treating both the systemic and the neurological symptoms of Pompe disease and potentially multiple other GSDs. Key components to gene therapy strategies include the choice of vector, promoter, and route of administration. To target the neurological symptoms of the disease, intrathecal (IT), intracisternal magna (ICM) (Hordeaux et al. 2017), and intracerebroventricular (ICV) (Lee et al. 2018) routes have been considered (Salabarria et al. 2020). One group treated adult GAA −/− mice with a single ICM injection of either AAV9 or AAVrh10-CAG-hGAA (Hordeaux et al. 2017). The authors demonstrated that the glycogen buildup was completely cleared four months post-ICM throughout the CNS, including brain cerebellum, brainstem, glia cells, myelin sheaths, and the proximal and distal spinal cords. These changes corresponded to improved motor function and reduction of hypertrophic cardiomyopathy one-year post-treatment (Hordeaux et al. 2017). Similarly, ICM administration of AAV5-GAA in Gaa−/− mice decreased intraneuronal glycogen content and improved ventilation (Qiu 2012). Another group (Lee et al. 2018) treated Pompe mice using rAAVN-GAA driven by a synapsis promoter. Treated animals exhibited increased GAA activity, decreased glycogen content in the CNS but not in skeletal muscle or the liver, improvement in motor coordination and balance with reduced astrogliosis and improved myelination. Two ongoing clinical trials focus on liver-directed gene therapy approaches ((Smith et al. 2023) and NCT04093349)) by creating a liver depot for GAA production. The trial by SPARK Therapeutics using an AAVRh74 derived capsid is currently on clinical hold due to adverse events (NCT04093349). Recently, the preliminary results were reported in three subjects receiving AAV8-LSPhGAA at a dose of 1.6 x 1012vg/Kg (Smith et al. 2023). The authors demonstrated safety; however, the lack of glycogen lowering suggested that despite the presence of increased GAA activity in skeletal muscle, the efficacy of gene therapy at this dose was not sufficient to replace ERT. While there is some potential overlap between different modalities, there are also significant differences in terms of cellular targeting, suitability for use in the CNS, and potentially synergistic effects that make the parallel development and testing of different modalities particularly critical for n-GSD patients.
Future directions and concluding remarks
Recent research into glycogen biology has provided key insights into brain biology and human disease. These include insights into the localization and direct utilization of glycogen in the brain and key metabolic and non-metabolic roles for brain glycogen with direct relevance to human health. Further, recent general insights into glycogen biology in other organs have revealed novel ongoing research avenues to broaden and deepen our understanding of glycogen function in the brain.
Intriguingly, multiple groups have established that glycogen is a key molecular component in many aspects of brain health. Multiple laboratories have demonstrated that brain glycogen increases in response to disease or injury, including interictal stages of epilepsy (Seo et al. 2022), stroke (Cai et al. 2020), and anesthesia (A. M. Brown, Rich, and Ransom 2019). Additionally, brain glycogen likely impacts neuronal plasticity during aging and dementia (Riba et al. 2021; Mann et al. 1987; E. C. B. Johnson et al. 2020). Thus, glycogen may have direct causal effects on diseases of the brain well beyond what is currently considered.
Continued progress on n-GSDs makes it evident that there are likely connections with more common neurological diseases. As the role of glycogen metabolism is defined in these other diseases, it is possible that treatments for n-GSDs could be more broadly utilized. Thus, the overlap and potential common therapeutic approaches between these groups of diseases is of great interest.
Funding Statement and acknowledgement:
Research reported in this publication was supported by the National Institute of Neurological Disorders and Stroke of the National Institutes of Health under Award Numbers R35 NS116824 (MSG) and P01 NS097197 (M.S.G). NIH grant 1R01AG066653, 1R01AG078702, 1R01CA266004, V-Scholar Grant, to R.C.S. R01 HL139708-02 and U01-NS116752-01A1 to MC and BJB.
Abbreviations:
- AAV
Adeno-associated virus
- AEF
Antibody-enzyme fusion
- ASO
Antisense oligonucleotide
- ANLS
Astrocyte-Neuron-Lactate-Shuttle
- APBD
adult polyglucosan body disease
- ATP
adenosine triphosphate
- CDG
congenital disorders of glycosylation
- CNS
Central nervous system
- DAB
1,4-dideoxy-1,4-imino-d-arabinitol
- DNA
Deoxyribonucleic acid
- EM
Electron microscopy
- G1D
Glucose transporter type 1 deficiency syndrome
- G1P
glucose-1-phosphate
- G6P
glucose-6-phosphate
- G6PASE
glucose-6-phosphatase
- GBE
glycogen branching enzyme
- GCMS
Gas chromatography mass spectrometry
- GDE
glycogen debranching enzyme
- Glc
glucose
- Gln
Glutamine
- Glu
Glutamate
- GLUT1
Glucose transporter type 1
- GP
glycogen phosphorylase
- GS
glycogen synthase
- GSD
Glycogen storage disorder
- GSG
Glucose Sparing by Glycogenolysis
- HPAEC-PAD
high performance anion-exchange chromatography with pulsed amperometric detection
- ICM
intracisternal magna
- ICV
intracerebroventricular
- IT
intrathecal
- LA
laforin
- LD
Lafora disease
- MALDI-IMS
Matrix-assisted laser desorption/ionization imaging mass spectrometry
- MRS
magnetic resonance spectroscopy
- MRI
magnetic resonance imaging
- MS
mass spectrometry
- n-GSDs
neurological glycogen storage diseases
- NMR
nuclear magnetic resonance
- P
phosphate
- PAS
periodic acid-Schiff
- PD
Pompe disease
- PGB
Polyglucosan
- PGM1
phosphoglucomutase 1
- SDS-PAGE
sodium dodecyl sulfate–polyacrylamide gel electrophoresis
- siRNA
Small interfering ribonucleic acid
- SRT
Substrate reducing therapy
- TCA
trichloroacetic acid
- UDP
Uridine diphosphate
- UDP-Glc
uridine diphosphate glucose
- UDP-GlcNac
Uridine diphosphate N-acetylglucosamine
Footnotes
Conflict of interest and disclosures.
RCS has received research support and received consultancy fees from Maze Therapeutics and is a member of the Medical Advisory Board for Little Warrior Foundation. MSG has research support, research compounds, or consultancy fees from Maze Therapeutics, Valerion Therapeutics, Ionis Pharmaceuticals, PTC Therapeutics, and the Glut1-Deficiency Syndrome Foundation. CWVK, RCS, and MSG are co-founders of Attrogen LLC. MC has received research support from Sanofi, Friedreich Ataxia Research Alliance (FARA), Amicus, AavantiBio, Lacerta, Provention Bio, Sarepta, Duchenne Research Fund, Muscular Dystrophy Association (MDA), GoFAR, Cydan, Audentes. MC has received consulting fees from AavantiBio, Reata, Lilly, Avexis and Gilbert foundation, SwanBio and PTC Therapeutics. BJB has received research support from SolidBio, ProventionBio, Barth Syndrome Foundation. BJB has received consulting fees from AavantiBio, Amicus Therapeutics, Rocket Pharma, Pfizer, Sanofi, and Sarepta Therapeutics. MC and BB are co-founders of AavantiBio, LLC and Ventura, LLC.
References
- Ahonen Saija, Nitschke Silvia, Grossman Tamar R, Kordasiewicz Holly, Wang Peixiang, Zhao Xiaochu, Guisso Dikran R, Kasiri Sahba, Nitschke Felix, and Minassian Berge A. 2021. “Gys1 Antisense Therapy Rescues Neuropathological Bases of Murine Lafora Disease.” Brain : A Journal of Neurology 144 (10): 2985–93. 10.1093/brain/awab194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alberini Cristina M, Cruz Emmanuel, Descalzi Giannina, Bessières Benjamin, and Gao Virginia. 2018. “Astrocyte Glycogen and Lactate: New Insights into Learning and Memory Mechanisms.” Glia 66 (6): 1244–62. 10.1002/glia.23250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andersen Jens V., and Schousboe Arne. 2022. “Glial Glutamine Homeostasis in Health and Disease.” Neurochemical Research. 10.1007/s11064-022-03771-1. [DOI] [PubMed] [Google Scholar]
- Andres Douglas A, Young Lyndsay E A, Veeranki X Sudhakar, Hawkinson Tara R, Levitan Bryana M, He Daheng, Wang Chi, Satin Jonathan, and Sun Ramon C. 2020. “Cro Improved Workflow for Mass Spectrometry – Based Metabolomics Analysis of the Heart” 295: 2676–86. 10.1074/jbc.RA119.011081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aterman K, and Norkin S. 1963. “The Periodic Acid – Schiff Reaction.” Nature 197 (4874): 1306. 10.1038/1971306a0. [DOI] [PubMed] [Google Scholar]
- Austin Grant L., Simmons Zoe R., Klier Jack E., Rondon Alberto, Hodges Brad L., Shaffer Robert, Aziz Nadine M., et al. 2019a. “Central Nervous System Delivery and Biodistribution Analysis of an Antibody-Enzyme Fusion for the Treatment of Lafora Disease.” Molecular Pharmaceutics 16 (9): 3791–3801. 10.1021/acs.molpharmaceut.9b00396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Austin Grant L, Simmons Zoe R, Klier Jack E, Rondon Alberto, Hodges Brad L, Shaffer Robert, Aziz Nadine M, et al. 2019b. “Central Nervous System Delivery and Biodistribution Analysis of an Antibody-Enzyme Fusion for the Treatment of Lafora Disease.” Molecular Pharmaceutics 16 (9): 3791–3801. 10.1021/acs.molpharmaceut.9b00396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baba O. 1993. “Production of monoclonal antibody that recognizes glycogen and its application for immunohistochemistry.” Kokubyo Gakkai zasshi. The Journal of the Stomatological Society, Japan 60 (2): 264–87. 10.5357/koubyou.60.264. [DOI] [PubMed] [Google Scholar]
- Baird WF, and Fisher ER. 1957. “Observations Concerning Vacuolation and Deposition of Glycogen in Nuclei of Hepatic Cells.” Laboratory Investigation; a Journal of Technical Methods and Pathology 6 (4): 324–33. [PubMed] [Google Scholar]
- Bak Lasse K., and Walls Anne B.. 2018. “Astrocytic Glycogen Metabolism in the Healthy and Diseased Brain.” Journal of Biological Chemistry 293 (19): 7108–16. 10.1074/jbc.R117.803239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bastian Chinthasagar, Quinn John, Doherty Christine, Franke Caroline, Faris Anna, Brunet Sylvain, and Baltan Selva. 2019. “Role of Brain Glycogen During Ischemia, Aging and Cell-to-Cell Interactions BT - Brain Glycogen Metabolism.” In , edited by DiNuzzo Mauro and Schousboe Arne, 347–61. Cham: Springer International Publishing. 10.1007/978-3-030-27480-1_12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bélanger Mireille, Allaman Igor, and Magistretti Pierre J. 2011. “Brain Energy Metabolism: Focus on Astrocyte-Neuron Metabolic Cooperation.” Cell Metabolism 14 (6): 724–38. 10.1016/j.cmet.2011.08.016. [DOI] [PubMed] [Google Scholar]
- Bernard C 1857. “Sur Le Mécanisme Physiologique de La Formation Du Sucre Dans Le Foie.” C R Acad Sci (Paris) 44: 578–586. [Google Scholar]
- Beyer Mike. 2022. “Aro Biotherapeutics Receives FDA Orphan Drug Designation for ABX1100 for the Treatment of Pompe Disease.” 2022. https://www.arobiotx.com/aro-receives-fda-orphan-drug-designation-for-abx1100. [Google Scholar]
- Blaschke Calvin, Black Alyson, Mehta Anand S, Angel Peggi M, Drake Richard R, and Drake Richard R. 2020. “Rapid N-Glycan Profiling of Serum and Plasma by a Novel Slide Based Imaging Mass Spectrometry Workflow Running Title : Rapid N-Glycan Profiling of Serum and Plasma Address Reprint Requests To :” 10.1021/jasms.0c00213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bogoch A, Casselman WGB, Kaplan A, and Bockus HL. 1955. “Studies of Hepatic Function in Diabetes Mellitus, Portal Cirrhosis and Other Liver Diseases: A Correlation of Clinical, Biochemical and Liver Needle Biopsy Findings I. Diabetes Mellitus.” The American Journal of Medicine 18 (3): 354–84. https://doi.org/ 10.1016/0002-9343(55)90217-5. [DOI] [PubMed] [Google Scholar]
- Bordone Melina Paula, Salman Mootaz M., Titus Haley E., Amini Elham, Andersen Jens V., Chakraborti Barnali, Diuba Artem V., et al. 2019. The Energetic Brain – A Review from Students to Students. Journal of Neurochemistry. Vol. 151. 10.1111/jnc.14829. [DOI] [PubMed] [Google Scholar]
- Boulan-Predseil P, Vital A, Brochet B, Darriet D, Henry P, and Vital C. 1995. “Dementia of Frontal Lobe Type Due to Adult Polyglucosan Body Disease.” Journal of Neurology 242 (8): 512–16. 10.1007/BF00867422. [DOI] [PubMed] [Google Scholar]
- Brewer M. Kathryn, and Gentry Matthew S.. 2019a. “Brain Glycogen Structure and Its Associated Proteins: Past, Present and Future.” Advances in Neurobiology 23: 17–81. 10.1007/978-3-030-27480-1_2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brewer M Kathryn, and Gentry Matthew S. 2019b. “Brain Glycogen Structure and Its Associated Proteins: Past, Present and Future.” Advances in Neurobiology 23: 17–81. 10.1007/978-3-030-27480-1_2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brewer M Kathryn, Uittenbogaard Annette, Austin Grant L, Segvich Dyann M, DePaoli-Roach Anna, Roach Peter J, McCarthy John J, et al. 2019. “Targeting Pathogenic Lafora Bodies in Lafora Disease Using an Antibody-Enzyme Fusion.” Cell Metabolism 30 (4): 689–705.e6. 10.1016/j.cmet.2019.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brown A, Baltan S, and Ransom BR. 2009. “Glycogen Metabolism in CNS White Matter.” In , edited by Larry RBT - Encyclopedia of Neuroscience Squire, 929–34. Oxford: Academic Press. https://doi.org/ 10.1016/B978-008045046-9.01732-0. [DOI] [Google Scholar]
- Brown Angus M, and Ransom Bruce R. 2007. “Astrocyte Glycogen and Brain Energy Metabolism.” Glia 55 (12): 1263–71. 10.1002/glia.20557. [DOI] [PubMed] [Google Scholar]
- Brown Angus M, Rich Laura R, and Ransom Bruce R. 2019. “Metabolism of Glycogen in Brain White Matter.” Advances in Neurobiology 23: 187–207. 10.1007/978-3-030-27480-1_7. [DOI] [PubMed] [Google Scholar]
- Brown BI, and Brown DH. 1966. “Lack of an Alpha-1,4-Glucan: Alpha-1,4-Glucan 6-Glycosyl Transferase in a Case of Type IV Glycogenosis.” Proceedings of the National Academy of Sciences of the United States of America 56 (2): 725–29. 10.1073/pnas.56.2.725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Budin Itay, de Rond Tristan, Chen Yan, Chan Leanne Jade G, Petzold Christopher J, and Keasling Jay D. 2018. “Viscous Control of Cellular Respiration by Membrane Lipid Composition.” Science 362 (6419): 1186–89. 10.1126/science.aat7925. [DOI] [PubMed] [Google Scholar]
- Cafferty MS, Lovelace RE, Hays AP, Servidei S, Dimauro S, and Rowland LP. 1991. “Polyglucosan Body Disease.” Muscle & Nerve 14 (2): 102–7. 10.1002/mus.880140203. [DOI] [PubMed] [Google Scholar]
- Cai Yanhui, Guo Haiyun, Fan Ze, Zhang Xinlei, Wu Di, Tang Wenhong, Gu Tingting, et al. 2020. “Glycogenolysis Is Crucial for Astrocytic Glycogen Accumulation and Brain Damage after Reperfusion in Ischemic Stroke.” IScience 23 (5). 10.1016/j.isci.2020.101136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cardell Robert R. 1977. “Smooth Endoplasmic Reticulum in Rat Hepatocytes during Glycogen Deposition and Depletion.” In , edited by Bourne GH, Danielli JF, and K W B T - International Review of Cytology Jeon, 48:221–79. Academic Press. https://doi.org/ 10.1016/S0074-7696(08)61746-5. [DOI] [PubMed] [Google Scholar]
- Carpenter WB, Smith FG, and Meneses HP. 1876. Principles of Human Physiology. Philadelphia, PA: Henry C. [Google Scholar]
- Chebotareva NA. 2007. “Effect of Molecular Crowding on the Enzymes of Glycogenolysis” 72 (13): 1478–90. [DOI] [PubMed] [Google Scholar]
- Chebotareva Natalia A, Andreeva Iraida E, Makeeva Valentina F, Livanova Natalia B, and Kurganov Boris I. 2004. “Effect of Molecular Crowding on Self-Association of Phosphorylase Kinase and Its Interaction with Phosphorylase b and Glycogen.” Journal of Molecular Recognition : JMR 17 (5): 426–32. 10.1002/jmr.696. [DOI] [PubMed] [Google Scholar]
- Chen Margaret A, and Weinstein David A. 2016. “Glycogen Storage Diseases: Diagnosis, Treatment and Outcome.” Translational Science of Rare Diseases 1: 45–72. 10.3233/TRD-160006. [DOI] [Google Scholar]
- Chipps HD, and Duff GL. 1942. “Glycogen Infiltration of the Liver Cell Nuclei.” The American Journal of Pathology 18 (4): 645–59. [PMC free article] [PubMed] [Google Scholar]
- Choi Hyun B, Gordon Grant R J, Zhou Ning, Tai Chao, Rungta Ravi L, Martinez Jennifer, Milner Teresa A, et al. 2012. “Metabolic Communication between Astrocytes and Neurons via Bicarbonate-Responsive Soluble Adenylyl Cyclase.” Neuron 75 (6): 1094–1104. 10.1016/j.neuron.2012.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conroy Lindsey R, Allison Derek B, Sun Qi, Young Lyndsay E A, Hawkinson Tara R, Clarke Harrison A, Ferreira Juanita E, et al. 2021. “Multi-Dimensional Reduction Clustering of Complex Carbohydrates Reveal Tissue Metabolism, Heterogeneity and Histopathology.” BioRxiv, January, 2021.06.14.448358. 10.1101/2021.06.14.448358. [DOI] [Google Scholar]
- Conroy Lindsey R, Hawkinson Tara R, Young Lyndsay E A, Gentry Matthew S, and Sun Ramon C. 2021. “Emerging Roles of N-Linked Glycosylation in Brain Physiology and Disorders.” Trends in Endocrinology and Metabolism: TEM 32 (12): 980–93. 10.1016/j.tem.2021.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conroy Lindsey R, Stanback Alexandra E, Young Lyndsay E A, Clarke Harrison A, Austin Grant L, Liu Jinze, Allison Derek B, and Sun Ramon C. 2021. “In Situ Analysis of N-Linked Glycans as Potential Biomarkers of Clinical Course in Human Prostate Cancer.” Molecular Cancer Research : MCR 19 (10): 1727–38. 10.1158/1541-7786.MCR-20-0967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crawford CR, Patel DH, Naeve C, and Belt JA. 1998. “Cloning of the Human Equilibrative, Nitrobenzylmercaptopurine Riboside (NBMPR)-Insensitive Nucleoside Transporter Ei by Functional Expression in a Transport-Deficient Cell Line.” The Journal of Biological Chemistry 273 (9): 5288–93. 10.1074/jbc.273.9.5288. [DOI] [PubMed] [Google Scholar]
- Cruz Nancy F, and Dienel Gerald A. 2002. “High Glycogen Levels in Brains of Rats with Minimal Environmental Stimuli: Implications for Metabolic Contributions of Working Astrocytes.” Journal of Cerebral Blood Flow and Metabolism : Official Journal of the International Society of Cerebral Blood Flow and Metabolism 22 (12): 1476–89. 10.1097/01.WCB.0000034362.37277.C0. [DOI] [PubMed] [Google Scholar]
- de-Souza-Ferreira Michelle, Ferreira Érika Elias, and de-Freitas-Junior Julio Cesar Madureira. 2023. “Aberrant N-Glycosylation in Cancer: MGAT5 and B1,6-GlcNAc Branched N-Glycans as Critical Regulators of Tumor Development and Progression.” Cellular Oncology. 10.1007/s13402-023-00770-4. [DOI] [PubMed] [Google Scholar]
- Dienel Gerald A. 2019a. “Brain Glucose Metabolism: Integration of Energetics with Function.” Physiological Reviews 99 (1): 949–1045. 10.1152/physrev.00062.2017. [DOI] [PubMed] [Google Scholar]
- Dienel Gerald A. 2019b. “Brain Glucose Metabolism: Integration of Energetics with Function.” Physiological Reviews 99 (1): 949–1045. 10.1152/physrev.00062.2017. [DOI] [PubMed] [Google Scholar]
- ———. 2020. “Metabolomic and Imaging Mass Spectrometric Assays of Labile Brain Metabolites: Critical Importance of Brain Harvest Procedures.” Neurochemical Research 45 (11): 2586–2606. 10.1007/s11064-020-03124-w. [DOI] [PubMed] [Google Scholar]
- Dienel Gerald A, and Carlson Gerald M. 2019. “Major Advances in Brain Glycogen Research: Understanding of the Roles of Glycogen Have Evolved from Emergency Fuel Reserve to Dynamic, Regulated Participant in Diverse Brain Functions.” Advances in Neurobiology 23: 1–16. 10.1007/978-3-030-27480-1_1. [DOI] [PubMed] [Google Scholar]
- Dienel Gerald A, and Rothman Douglas L. 2019. “Glycogenolysis in Cerebral Cortex During Sensory Stimulation, Acute Hypoglycemia, and Exercise: Impact on Astrocytic Energetics, Aerobic Glycolysis, and Astrocyte-Neuron Interactions BT - Brain Glycogen Metabolism.” In , edited by DiNuzzo Mauro and Schousboe Arne, 209–67. Cham: Springer International Publishing. 10.1007/978-3-030-27480-1_8. [DOI] [PubMed] [Google Scholar]
- Dinuzzo Mauro, Mangia Silvia, Maraviglia Bruno, and Giove Federico. 2012. “The Role of Astrocytic Glycogen in Supporting the Energetics of Neuronal Activity.” Neurochemical Research 37 (11): 2432–38. 10.1007/s11064-012-0802-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DiNuzzo Mauro, and Schousboe Arne. 2019. Brain Glycogen Metabolism. Advances in Neurobiology. Vol. 23. [DOI] [PubMed] [Google Scholar]
- Dong Xifeng. 2019. “Nuclear Glycogenolysis Modulates Histone Acetylation: A Novel Mechanism of Epigenetic Regulation in Cancer.” Cancer Communications 39 (1): 71. 10.1186/s40880-019-0420-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Donohue Katherine J., Gentry Matthew S., and Sun Ramon C.. 2020. “The E3 Ligase Malin Plays a Pivotal Role in Promoting Nuclear Glycogenolysis and Histone Acetylation.” Annals of Translational Medicine 8 (5): 254–254. 10.21037/atm.2020.01.130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doron Adi, Rubin Alon, Benmelech-Chovav Aviya, Benaim Netai, Carmi Tom, Refaeli Ron, Novick Nechama, Kreisel Tirzah, Ziv Yaniv, and Goshen Inbal. 2022. “Hippocampal Astrocytes Encode Reward Location.” Nature 609 (7928): 772–78. 10.1038/s41586-022-05146-6. [DOI] [PubMed] [Google Scholar]
- Drake Richard R, Powers Thomas W, Norris-Caneda Kim, Mehta Anand S, and Angel Peggi M. 2018. “In Situ Imaging of N-Glycans by MALDI Imaging Mass Spectrometry of Fresh or Formalin-Fixed Paraffin-Embedded Tissue.” Current Protocols in Protein Science 94 (1): e68. https://doi.org/ 10.1002/cpps.68. [DOI] [PubMed] [Google Scholar]
- Drulis-Fajdasz Dominika, Wójtowicz Tomasz, Wawrzyniak Marcin, Wlodarczyk Jakub, Mozrzymas Jerzy W, and Rakus Dariusz. 2015. “Involvement of Cellular Metabolism in Age-Related LTP Modifications in Rat Hippocampal Slices.” Oncotarget 6 (16): 14065–81. 10.18632/oncotarget.4188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duran Jordi, Brewer M Kathryn, Hervera Arnau, Gruart Agnès, Del Rio Jose Antonio, Delgado-García José M, and Guinovart Joan J. 2020. “Lack of Astrocytic Glycogen Alters Synaptic Plasticity but Not Seizure Susceptibility.” Molecular Neurobiology 57 (11): 4657–66. 10.1007/s12035-020-02055-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duran Jordi, Gruart Agnès, López-Ramos Juan Carlos, Delgado-García José M, and Guinovart Joan J. 2019. “Glycogen in Astrocytes and Neurons: Physiological and Pathological Aspects.” Advances in Neurobiology 23: 311–29. 10.1007/978-3-030-27480-1_10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duran Jordi, Gruart Agnès, Varea Olga, López-Soldado Iliana, Delgado-García José M., and Guinovart Joan J.. 2019. “Lack of Neuronal Glycogen Impairs Memory Formation and Learning-Dependent Synaptic Plasticity in Mice.” Frontiers in Cellular Neuroscience 13 (August): 1–11. 10.3389/fncel.2019.00374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duran Jordi, Hervera Arnau, Kia H Markussen Olga Varea, López-Soldado Iliana, Sun Ramon C, del Río Jose Antonio, Gentry Matthew S, and Guinovart Joan J. 2021. “Astrocytic Glycogen Accumulation Drives the Pathophysiology of Neurodegeneration in Lafora Disease.” Brain, no. 2021. 10.1093/brain/awab110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duran Jordi, Saez Isabel, Gruart Agnès, Guinovart Joan J, and Delgado-García José M. 2013. “Impairment in Long-Term Memory Formation and Learning-Dependent Synaptic Plasticity in Mice Lacking Glycogen Synthase in the Brain.” Journal of Cerebral Blood Flow and Metabolism : Official Journal of the International Society of Cerebral Blood Flow and Metabolism 33 (4): 550–56. 10.1038/jcbfm.2012.200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eichler Jerry. 2019. “Protein Glycosylation.” Current Biology : CB 29 (7): R229–31. 10.1016/j.cub.2019.01.003. [DOI] [PubMed] [Google Scholar]
- Ellingwood Sara S., and Cheng Alan. 2018. “Biochemical and Clinical Aspects of Glycogen Storage Diseases.” Journal of Endocrinology 238 (3): R131–41. 10.1530/JOE-18-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Folbergrová J, Passonneau JV, Lowry OH, and Schulz DW. 1969. “Glycogen, Ammonia and Related Metabolities in the Brain during Seizures Evoked by Methionine Sulphoximine.” Journal of Neurochemistry 16 (2): 191–203. 10.1111/j.1471-4159.1969.tb05937.x. [DOI] [PubMed] [Google Scholar]
- Foster M 1899. “Claude Bernard.” Unwin, London. [Google Scholar]
- Gaunitz Stefan, Tjernberg Lars O, and Schedin-weiss Sophia. 2020. “The N-Glycan Profile in Cortex and Hippocampus Is Altered in Alzheimer Disease.” Journal of Neurochemistry, no. September 2020: 292–304. 10.1111/jnc.15202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gentry Matthew S., Worby Carolyn A., and Dixon Jack E.. 2005. “Insights into Lafora Disease: Malin Is an E3 Ubiquitin Ligase That Ubiquitinates and Promotes the Degradation of Laforin.” Proceedings of the National Academy of Sciences of the United States of America 102 (24): 8501–6. 10.1073/pnas.0503285102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gentry Matthew S, Guinovart Joan J, Minassian Berge A, Roach Peter J, and Serratosa Jose M. 2018. “Lafora Disease Offers a Unique Window into Neuronal Glycogen Metabolism.” The Journal of Biological Chemistry 293 (19): 7117–25. 10.1074/jbc.R117.803064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gibbs Marie E, Anderson Damian G, and Hertz Leif. 2006. “Inhibition of Glycogenolysis in Astrocytes Interrupts Memory Consolidation in Young Chickens.” Glia 54 (3): 214–22. 10.1002/glia.20377. [DOI] [PubMed] [Google Scholar]
- Gibbs Marie E, Lloyd Hilary G E, Santa Thomas, and Hertz Leif. 2007. “Glycogen Is a Preferred Glutamate Precursor during Learning in 1-Day-Old Chick: Biochemical and Behavioral Evidence.” Journal of Neuroscience Research 85 (15): 3326–33. 10.1002/jnr.21307. [DOI] [PubMed] [Google Scholar]
- von Gierke E. 1929. “Hepato-Nephro-Megalia Glycogenica (Glykogenspeicher-Krankheit Der Leber Und Nieren).” Beitr Pathol Anat 497-513 82: 497–513. [Google Scholar]
- Gujral Palak, Mahajan Vishakha, Lissaman Abbey C, and Ponnampalam Anna P. 2020. “Histone Acetylation and the Role of Histone Deacetylases in Normal Cyclic Endometrium.” Reproductive Biology and Endocrinology 18 (1): 84. 10.1186/s12958-020-00637-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gumusgoz Emrah, Kasiri Sahba, Guisso Dikran R., Wu Jun, Dear Matthew, Verhalen Brandy, and Minassian Berge A.. 2022. “AAV-Mediated Artificial MiRNA Reduces Pathogenic Polyglucosan Bodies and Neuroinflammation in Adult Polyglucosan Body and Lafora Disease Mouse Models.” Neurotherapeutics 19 (3): 982–93. 10.1007/s13311-022-01218-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo Haiyun, Fan Ze, Wang Shiquan, Ma Lina, Wang Jin, Yu Doutong, Zhang Zhen, et al. 2021. “Astrocytic A1/A2 Paradigm Participates in Glycogen Mobilization Mediated Neuroprotection on Reperfusion Injury after Ischemic Stroke.” Journal of Neuroinflammation 18 (1): 230. 10.1186/s12974-021-02284-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansen James E, Chan Grace, Liu Yanfeng, Denise C Hegan Shibani Dalal, Kwon Youngho, Xu Yuanyuan, et al. 2013. “Targeting Cancer with a Lupus Autoantibody #” 4 (157). 10.1126/scitranslmed.3004385.Targeting. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansen James E, Tse Chung-Ming, Chan Grace, Heinze Emil R, Nishimura Robert N, and Weisbart Richard H. 2007. “Intranuclear Protein Transduction through a Nucleoside Salvage Pathway.” The Journal of Biological Chemistry 282 (29): 20790–93. 10.1074/jbc.C700090200. [DOI] [PubMed] [Google Scholar]
- Hansen James E, Weisbart Richard H, and Nishimura Robert N. 2005. “Antibody Mediated Transduction of Therapeutic Proteins into Living Cells.” TheScientificWorldJournal 5 (September): 782–88. 10.1100/tsw.2005.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hartman KR, T J Triche, Kinsella TJ, and Miser JS. 1991. “Prognostic Value of Histopathology in Ewing’s Sarcoma. Long-Term Follow-up of Distal Extremity Primary Tumors.” Cancer 67 (1): 163–71. . [DOI] [PubMed] [Google Scholar]
- Hawkinson Tara R, Clarke Harrison A, Young Lyndsay E A, Conroy Lindsey R, Markussen Kia H, Kerch Kayla M, Johnson Lance A, et al. 2021. “In Situ Spatial Glycomic Imaging of Mouse and Human Alzheimer’s Disease Brains.” Alzheimer’s & Dementia : The Journal of the Alzheimer’s Association, December. 10.1002/alz.12523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hawkinson Tara R, and Sun Ramon C. 2022. “Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry Imaging of Glycogen In Situ.” Methods in Molecular Biology (Clifton, N.J.) 2437: 215–28. 10.1007/978-1-0716-2030-4_15. [DOI] [PubMed] [Google Scholar]
- Hertz Leif, Peng Liang, and Dienel Gerald A. 2007. “Energy Metabolism in Astrocytes: High Rate of Oxidative Metabolism and Spatiotemporal Dependence on Glycolysis/Glycogenolysis.” Journal of Cerebral Blood Flow and Metabolism : Official Journal of the International Society of Cerebral Blood Flow and Metabolism 27 (2): 219–49. 10.1038/sj.jcbfm.9600343. [DOI] [PubMed] [Google Scholar]
- Hobson-Webb Lisa D, Austin Stephanie L, Bali Deeksha S, and Kishnani Priya S. 2010. “The Electrodiagnostic Characteristics of Glycogen Storage Disease Type III.” Genetics in Medicine : Official Journal of the American College of Medical Genetics 12 (7): 440–45. 10.1097/GIM.0b013e3181cd735b. [DOI] [PubMed] [Google Scholar]
- Hof PR, Pascale E, and Magistretti PJ. 1988. “K+ at Concentrations Reached in the Extracellular Space during Neuronal Activity Promotes a Ca2+-Dependent Glycogen Hydrolysis in Mouse Cerebral Cortex.” The Journal of Neuroscience 8 (6): 1922 LP – 1928. 10.1523/JNEUROSCI.08-06-01922.1988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hordeaux J, Dubreil L, Robveille C, Deniaud J, Pascal Q, Dequéant B, Pailloux J, et al. 2017. “Long-Term Neurologic and Cardiac Correction by Intrathecal Gene Therapy in Pompe Disease.” Acta Neuropathologica Communications 5 (1): 66. 10.1186/s40478-017-0464-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hsu Chao-Hsiung, Lin Stephen, Ho Ai-Chen, Johnson T Derek, Wang Paul C, Scafidi Joseph, and Tu Tsang-Wei. 2021. “Comparison of in Vivo and in Situ Detection of Hippocampal Metabolites in Mouse Brain Using (1) H-MRS.” NMR in Biomedicine 34 (2): e4451. 10.1002/nbm.4451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishikawa T, and Pel YF. 1965. “Intramitochondrial Glycogen Particles in Rat Retinal Receptor Cells.” The Journal of Cell Biology 25 (2): 402–7. 10.1083/jcb.25.2.402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson Erik C.B., Dammer Eric B., Duong Duc M., Ping Lingyan, Zhou Maotian, Yin Luming, Higginbotham Lenora A., et al. 2020. “Large-Scale Proteomic Analysis of Alzheimer’s Disease Brain and Cerebrospinal Fluid Reveals Early Changes in Energy Metabolism Associated with Microglia and Astrocyte Activation.” Nature Medicine 26 (5): 769–80. 10.1038/s41591-020-0815-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Johnson Margaret E. 2018. “Modeling the Self-Assembly of Protein Complexes through a Rigid- Body Rotational Reaction – Di Ff Usion Algorithm.” 10.1021/acs.jpcb.8b08339. [DOI] [PubMed] [Google Scholar]
- Juras Jelena A, Webb Madison B, Young Lyndsay E A, Markussen Kia H, Hawkinson Tara R, Buoncristiani Michael D, Bolton Kayli E, et al. 2022. “In Situ Microwave Fixation to Define the Terminal Rodent Brain Metabolome.” BioRxiv, January, 2022.08.16.504166. 10.1101/2022.08.16.504166. [DOI] [Google Scholar]
- Kerly B Y Margaret. 1929. “X . THE SOLUBILITY OF GLYCOGEN . IN the Course of an Investigation on Glycogen , the Solubility of a Sample with That of a Sample Prepared from an Aqueous Extract of Tissue Dehydrated” 250 (curve c). [Google Scholar]
- Kirkman BR, and Whelan WJ. 1986. “Glucosamine Is a Normal Component of Liver Glycogen.” FEBS Letters 194 (1): 6–11. 10.1016/0014-5793(86)80041-2. [DOI] [PubMed] [Google Scholar]
- Korlimarla Aditi, Lim Jeong-A, Kishnani Priya S., and Sun Baodong. 2019. “An Emerging Phenotype of Central Nervous System Involvement in Pompe Disease: From Bench to Bedside and Beyond.” Annals of Translational Medicine 7 (13): 289–289. 10.21037/atm.2019.04.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee Ni Chung, Hwu Wuh Liang, Muramatsu Shin Ichi, Falk Darin J., Byrne Barry J., Cheng Chia Hao, Shih Nien Chu, Chang Kai Ling, Tsai Li Kai, and Chien Yin Hsiu. 2018. “A Neuron-Specific Gene Therapy Relieves Motor Deficits in Pompe Disease Mice.” Molecular Neurobiology 55 (6): 5299–5309. 10.1007/s12035-017-0763-4. [DOI] [PubMed] [Google Scholar]
- Liu Qingxu, Li Jiaxin, Zhang Weiji, Xiao Chen, Zhang Shihao, Nian Cheng, Li Junhong, et al. 2021. “Glycogen Accumulation and Phase Separation Drives Liver Tumor Initiation.” Cell 184 (22): 5559–5576.e19. 10.1016/j.cell.2021.10.001. [DOI] [PubMed] [Google Scholar]
- Longsworth LG. 1954. “Temperature Dependence of Diffusion in Aqueous Solutions.” The Journal of Physical Chemistry 58 (9): 770–73. 10.1021/j150519a017. [DOI] [Google Scholar]
- Lu Hong, Chen Chuan, and Klaassen Curtis. 2004. “Tissue Distribution of Concentrative and Equilibrative Nucleoside Transporters in Male and Female Rats and Mice.” Drug Metabolism and Disposition: The Biological Fate of Chemicals 32 (12): 1455–61. 10.1124/dmd.104.001123. [DOI] [PubMed] [Google Scholar]
- Magistretti Pierre J., and Allaman Igor. 2015. “A Cellular Perspective on Brain Energy Metabolism and Functional Imaging.” Neuron 86 (4): 883–901. 10.1016/j.neuron.2015.03.035. [DOI] [PubMed] [Google Scholar]
- Maley F, McGarrahan JF, and DelGiacco R. 1966. “Galactosamine: A Precursor of Glycogen Glucosamine.” Biochemical and Biophysical Research Communications 23 (1): 85–91. 10.1016/0006-291x(66)90273-7. [DOI] [PubMed] [Google Scholar]
- De Man JCH, Blok APR, and Beens W. 1966. “Relationship between Gllycogen and Agranular Endoplasmic Reticulum in Rat Hepatic Cells.” Journal of Histochemistry & Cytochemistry 14 (2): 135–46. 10.1177/14.2.135. [DOI] [PubMed] [Google Scholar]
- Mann DMA, Sumpter PQ, Davies CA, and Yates PO. 1987. “Glycogen Accumulations in the Cerebral Cortex in Alzheimer’s Disease.” Acta Neuropathologica 73 (2): 181–84. 10.1007/BF00693786. [DOI] [PubMed] [Google Scholar]
- Marchand I, Chorneyko K, Tarnopolsky M, Hamilton S, Shearer J, Potvin J, and Graham TE. 2002. “Quantification of Subcellular Glycogen in Resting Human Muscle: Granule Size, Number, and Location.” Journal of Applied Physiology (Bethesda, Md. : 1985) 93 (5): 1598–1607. 10.1152/japplphysiol.00585.2001. [DOI] [PubMed] [Google Scholar]
- Markussen Kia H, Macedo Jessica K A, Machío María, Dolce Alison, Goldberg Y Paul, Kooi Craig W Vander, and Gentry Matthew S. 2021. “The 6th International Lafora Epilepsy Workshop: Advances in the Search for a Cure.” Epilepsy & Behavior : E&B 119 (June): 107975. 10.1016/j.yebeh.2021.107975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McDowell Colin T., Lu Xiaowei, Mehta Anand S., Angel Peggi M., and Drake Richard R.. 2021. “Applications and Continued Evolution of Glycan Imaging Mass Spectrometry.” Mass Spectrometry Reviews, no. July: 1–32. 10.1002/mas.21725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McMahon Robert J., and Frost Susan C.. 1996. “Glycogen: A Carbohydrate Source for GLUT-1 Glycosylation during Glucose Deprivation of 3T3-L1 Adipocytes.” American Journal of Physiology - Endocrinology and Metabolism 270 (4 33-4). 10.1152/ajpendo.1996.270.4.e640. [DOI] [PubMed] [Google Scholar]
- McManus JFA. 1946. “Histological Demonstration of Mucin after Periodic Acid.” Nature 158 (4006): 202. 10.1038/158202a0. [DOI] [PubMed] [Google Scholar]
- ———. 1948. “Histological and Histochemical Uses of Periodic Acid.” Stain Technology 23 (3): 99–108. 10.3109/10520294809106232. [DOI] [PubMed] [Google Scholar]
- Meléndez R, Meléndez-Hevia E, and Cascante M. 1997. “How Did Glycogen Structure Evolve to Satisfy the Requirement for Rapid Mobilization of Glucose? A Problem of Physical Constraints in Structure Building.” Journal of Molecular Evolution 45 (4): 446–55. 10.1007/pl00006249. [DOI] [PubMed] [Google Scholar]
- Mercier C, and Whelan WJ. 1973. “Further Characterization of Glycogen from Type-IV Glycogen-Storage Disease.” European Journal of Biochemistry 40 (1): 221–23. 10.1111/j.1432-1033.1973.tb03189.x. [DOI] [PubMed] [Google Scholar]
- Mizuguchi Keishi, Minato Hiroshi, Onishi Hitomi, Mitani Yuki, and Kawai Jun. 2016. “Cytopathological Findings of Primary Pulmonary Ewing Family of Tumors with EWSR1 Translocation: A Case Report.” Thoracic Cancer. Singapore. 10.1111/1759-7714.12347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moses Shimon W. 2002. “Historical Highlights and Unsolved Problems in Glycogen Storage Disease Type 1.” European Journal of Pediatrics 161 (1): S2–9. 10.1007/BF02679988. [DOI] [PubMed] [Google Scholar]
- Mozrzymas Jerzy, Szczęsny Tomasz, and Rakus Darek. 2011. “The Effect of Glycogen Phosphorolysis on Basal Glutaminergic Transmission.” Biochemical and Biophysical Research Communications 404 (2): 652–55. https://doi.org/ 10.1016/j.bbrc.2010.12.033. [DOI] [PubMed] [Google Scholar]
- Nakamura-Tsuruta Sachiko, Yasuda Michiko, Nakamura Toshiyuki, Shinoda Eri, Furuyashiki Takashi, Kakutani Ryo, Takata Hiroki, Kato Yoji, and Ashida Hitoshi. 2012. “Comparative Analysis of Carbohydrate-Binding Specificities of Two Anti-Glycogen Monoclonal Antibodies Using ELISA and Surface Plasmon Resonance.” Carbohydrate Research 350 (March): 49–54. 10.1016/j.carres.2011.12.029. [DOI] [PubMed] [Google Scholar]
- Ng Bobby G, and Freeze Hudson H. 2018. “Perspectives on Glycosylation and Its Congenital Disorders.” Trends in Genetics 34 (6): 466–76. https://doi.org/ 10.1016/j.tig.2018.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nielsen Joachim, Suetta Charlotte, Hvid Lars G, Schrøder Henrik D, Aagaard Per, and Ortenblad Niels. 2010. “Subcellular Localization-Dependent Decrements in Skeletal Muscle Glycogen and Mitochondria Content Following Short-Term Disuse in Young and Old Men.” American Journal of Physiology. Endocrinology and Metabolism 299 (6): E1053–60. 10.1152/ajpendo.00324.2010. [DOI] [PubMed] [Google Scholar]
- Nitschke Felix, Ahonen Saija J., Nitschke Silvia, Mitra Sharmistha, and Minassian Berge A.. 2018a. “Lafora Disease — from Pathogenesis to Treatment Strategies.” Nature Reviews Neurology 14 (10): 606–17. 10.1038/s41582-018-0057-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nitschke Felix, Ahonen Saija J, Nitschke Silvia, Mitra Sharmistha, and Minassian Berge A. 2018b. “Lafora Disease - from Pathogenesis to Treatment Strategies.” Nature Reviews. Neurology 14 (10): 606–17. 10.1038/s41582-018-0057-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nitschke Felix, Sullivan Mitchell A, Wang Peixiang, Zhao Xiaochu, Chown Erin E, Perri Ami M, Israelian Lori, et al. 2017. “Abnormal Glycogen Chain Length Pattern, Not Hyperphosphorylation, Is Critical in Lafora Disease.” EMBO Molecular Medicine 9 (7): 906–17. 10.15252/emmm.201707608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Obel Linea F., Müller Margit S., Walls Anne B., Sickmann Helle M., Bak Lasse K., Waagepetersen Helle S., and Schousboe Arne. 2012. “Brain Glycogen - New Perspectives on Its Metabolic Function and Regulation at the Subcellular Level.” Frontiers in Neuroenergetics 4 (MAR): 1–15. 10.3389/fnene.2012.00003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oe Yuki, Akther Sonam, and Hirase Hajime. 2019. “Regional Distribution of Glycogen in the Mouse Brain Visualized by Immunohistochemistry BT - Brain Glycogen Metabolism.” In , edited by DiNuzzo Mauro and Schousboe Arne, 147–68. Cham: Springer International Publishing. 10.1007/978-3-030-27480-1_5. [DOI] [PubMed] [Google Scholar]
- Oe Yuki, Baba Otto, Ashida Hitoshi, Nakamura Kouichi C., and Hirase Hajime. 2016. “Glycogen Distribution in the Microwave-Fixed Mouse Brain Reveals Heterogeneous Astrocytic Patterns.” Glia 64 (9): 1532–45. 10.1002/glia.23020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Papi Atefe, Zamani Mina, Shariati Gholamreza, Sedaghat Alireza, Seifi Tahere, Negahdari Samira, Sedighzadeh Sahar Sadat, et al. 2023. “Whole Exome Sequencing Reveals Several Novel Variants in Congenital Disorders of Glycosylation and Glycogen Storage Diseases in Seven Patients from Iran.” Molecular Genetics & Genomic Medicine, 1–6. 10.1002/mgg3.2099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Persson Laura B, Ambati Vardhaan S, and Brandman Onn. 2020. “Cellular Control of Viscosity Counters Changes in Temperature and Energy Availability.” Cell 183 (6): 1572–1585.e16. 10.1016/j.cell.2020.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Persson Laura B, Ambati Vardhaan S, Persson Laura B, Ambati Vardhaan S, and Brandman Onn. 2020. “Article Cellular Control of Viscosity Counters Changes in Temperature and Energy Availability Ll Ll Article Cellular Control of Viscosity Counters Changes in Temperature and Energy Availability.” Cell 183 (6): 1572–1585.e16. 10.1016/j.cell.2020.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pfeiffer-Guglielmi Brigitte, Fleckenstein Burkhard, Jung Günther, and Hamprecht Bernd. 2003. “Immunocytochemical Localization of Glycogen Phosphorylase Isozymes in Rat Nervous Tissues by Using Isozyme-Specific Antibodies.” Journal of Neurochemistry 85 (1): 73–81. 10.1046/j.1471-4159.2003.01644.x. [DOI] [PubMed] [Google Scholar]
- Pflüger E. 1909. “Meine Methode Der Quantitativen Analyse Des Glykogenes Und Die Arteigenthümlichkeit Der Substanzen Des Thierleibes.” Archiv Für Die Gesamte Physiologie Des Menschen Und Der Tiere 129 (6–7): 362–378. [Google Scholar]
- Piedade Ana, Francisco Rita, Jaeken Jaak, Sarkhail Peymaneh, Brasil Sandra, Ferreira Carlos R, Rijoff Tatiana, et al. 2022. “Epidemiology of Congenital Disorders of Glycosylation ( CDG )— Overview and Perspectives.” Journal of Rare Diseases, 1–19. 10.1007/s44162-022-00003-6. [DOI] [Google Scholar]
- Pontén U, Ratcheson RA, and Siesjö BK. 1973. “Metabolic Changes in the Brains of Mice Frozen in Liquid Nitrogen.” Journal of Neurochemistry 21 (5): 1211–16. [PubMed] [Google Scholar]
- Powers Thomas W, Jones E Ellen, Betesh Lucy R, Romano Patrick R, Gao Peng, Copland John A, Mehta Anand S, and Drake Richard R. 2013. “Matrix Assisted Laser Desorption Ionization Imaging Mass Spectrometry Workflow for Spatial Profiling Analysis of N-Linked Glycan Expression in Tissues.” Analytical Chemistry 85 (20): 9799–9806. 10.1021/ac402108x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rajasekaran Karthik, Ma Qian, Levi B Good Gauri Kathote, Jakkamsetti Vikram, Liu Peiying, Avila Adrian, et al. 2023. “Metabolic Modulation of Synaptic Failure and Thalamocortical Hypersynchronization with Preserved Consciousness in Glut1 Deficiency.” Science Translational Medicine 14 (665): eabn2956. 10.1126/scitranslmed.abn2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raththagala Madushi, Brewer M. Kathryn, Parker Matthew W., Sherwood Amanda R., Wong Brian K., Hsu Simon, Bridges Travis M., et al. 2015. “Structural Mechanism of Laforin Function in Glycogen Dephosphorylation and Lafora Disease.” Molecular Cell 57 (2): 261–72. https://doi.org/ 10.1016/j.molcel.2014.11.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reily Colin, Stewart Tyler J., Renfrow Matthew B., and Novak Jan. 2019. “Glycosylation in Health and Disease.” Nature Reviews Nephrology 15 (6): 346–66. 10.1038/s41581-019-0129-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riba Marta, Jaume del Valle Elisabet Augé, Vilaplana Jordi, and Pelegrí Carme. 2021. “From Corpora Amylacea to Wasteosomes: History and Perspectives.” Ageing Research Reviews 72: 101484. https://doi.org/ 10.1016/j.arr.2021.101484. [DOI] [PubMed] [Google Scholar]
- Richter K, Hamprecht B, and Scheich H. 1996. “Ultrastructural Localization of Glycogen Phosphorylase Predominantly in Astrocytes of the Gerbil Brain.” Glia 17 (4): 263–73. . [DOI] [PubMed] [Google Scholar]
- Rothman Douglas L, Dienel Gerald A, Behar Kevin L, Hyder Fahmeed, DiNuzzo Mauro, Giove Federico, and Mangia Silvia. 2022. “Glucose Sparing by Glycogenolysis (GSG) Determines the Relationship between Brain Metabolism and Neurotransmission.” Journal of Cerebral Blood Flow and Metabolism : Official Journal of the International Society of Cerebral Blood Flow and Metabolism 42 (5): 844–60. 10.1177/0271678X211064399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saez Isabel, Duran Jordi, Sinadinos Christopher, Beltran Antoni, Yanes Oscar, María F Tevy Carlos Martínez-Pons, Milán Marco, and Guinovart Joan J. 2014. “Neurons Have an Active Glycogen Metabolism That Contributes to Tolerance to Hypoxia.” Journal of Cerebral Blood Flow and Metabolism : Official Journal of the International Society of Cerebral Blood Flow and Metabolism 34 (6): 945–55. 10.1038/jcbfm.2014.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salabarria SM, Nair J, Clement N, Smith BK, Raben N, Fuller DD, Byrne BJ, and Corti M. 2020. “Advancements in AAV-Mediated Gene Therapy for Pompe Disease.” Journal of Neuromuscular Diseases 7 (1): 15–31. 10.3233/JND-190426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwarz Flavio, and Aebi Markus. 2011. “Mechanisms and Principles of N-Linked Protein Glycosylation.” Current Opinion in Structural Biology 21 (5): 576–82. 10.1016/j.sbi.2011.08.005. [DOI] [PubMed] [Google Scholar]
- Scott Hilary, and Panin Vladislav M. 2014. “N-Glycosylation in Regulation of the Nervous System BT - Glycobiology of the Nervous System.” In , edited by Yu Robert K and Schengrund Cara-Lynne, 367–94. New York, NY: Springer New York. 10.1007/978-1-4939-1154-7_17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seo Gi Young, Elliott S Neal Felicity Han, Vidovic Diana, Nooru-Mohamed Fathima, Dienel Gerald A, Sullivan Mitchell A, and Borges Karin. 2022. “Brain Glycogen Content Is Increased in the Acute and Interictal Chronic Stages of the Mouse Pilocarpine Model of Epilepsy.” Epilepsia Open 7 (2): 361–67. 10.1002/epi4.12599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shearer Jane, and Graham Terry E. 2002. “New Perspectives on the Storage and Organization of Muscle Glycogen.” Canadian Journal of Applied Physiology = Revue Canadienne de Physiologie Appliquee 27 (2): 179–203. 10.1139/h02-012. [DOI] [PubMed] [Google Scholar]
- Sickmann Helle M, Walls Anne B, Schousboe Arne, Bouman Stephan D, and Waagepetersen Helle S. 2009. “Functional Significance of Brain Glycogen in Sustaining Glutamatergic Neurotransmission.” Journal of Neurochemistry 109 (s1): 80–86. https://doi.org/ 10.1111/j.1471-4159.2009.05915.x. [DOI] [PubMed] [Google Scholar]
- Sinensky Michael. 1974. “Homeoviscous Adaptation-A Homeostatic . Process That Regulates the Viscosity of Membrane Lipids in Escherichia Coli” 71 (2): 522–25. 10.1073/pnas.71.2.522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith Edward C, Hopkins Sam, Case Laura E, Xu Ming, Walters Crista, Dearmey Stephanie, Han Sang-Oh, et al. 2023. “Phase I Study of Liver Depot Gene Therapy in Late-Onset Pompe Disease.” Molecular Therapy : The Journal of the American Society of Gene Therapy, February. 10.1016/j.ymthe.2023.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soares Ana Francisca, Gruetter Rolf, and Lei Hongxia. 2017. “Technical and Experimental Features of Magnetic Resonance Spectroscopy of Brain Glycogen Metabolism.” Analytical Biochemistry 529: 117–26. https://doi.org/ 10.1016/j.ab.2016.12.023. [DOI] [PubMed] [Google Scholar]
- Somogyi Michael. 1934. “The Solubility and Preparation of Phosphorous- and Nitrogen-Free Glycogen.” Journal of Biological Chemistry 104 (2): 245–53. 10.1016/s0021-9258(18)75759-1. [DOI] [Google Scholar]
- Stanback Alexandra E, Conroy Lindsey R, Young Lyndsay E A, Hawkinson Tara R, Markussen Kia H, Clarke Harrison A, Allison Derek B, and Sun Ramon C. 2021. “Regional N-Glycan and Lipid Analysis from Tissues Using MALDI-Mass Spectrometry Imaging.” STAR Protocols 2 (1): 100304. 10.1016/j.xpro.2021.100304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stojanovski Klement, Ferrar Tony, Benisty Hannah, Uschner Friedemann, Delgado Javier, Jimenez Javier, Solé Carme, et al. 2017. “Interaction Dynamics Determine Signaling and Output Pathway Responses.” Cell Reports 19 (1): 136–49. https://doi.org/ 10.1016/j.celrep.2017.03.029. [DOI] [PubMed] [Google Scholar]
- Sullivan Mitchell A, Nitschke Silvia, Skwara Evan P, Wang Peixiang, Zhao Xiaochu, Pan Xiao S, Chown Erin E, et al. 2019. “Skeletal Muscle Glycogen Chain Length Correlates with Insolubility in Mouse Models of Polyglucosan-Associated Neurodegenerative Diseases.” Cell Reports 27 (5): 1334–1344.e6. https://doi.org/ 10.1016/j.celrep.2019.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun Ramon C, Dukhande Vikas V, Zhou Zhengqiu, Young Lyndsay E A, Emanuelle Shane, Brainson Christine Fillmore, and Gentry Matthew S. 2019. “Nuclear Glycogenolysis Modulates Histone Acetylation in Human Non-Small Cell Lung Cancers.” Cell Metabolism 30 (5): 903–916.e7. 10.1016/j.cmet.2019.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun Ramon C, Young Lyndsay E A, Bruntz Ronald C, Markussen Kia H, Zhou Zhengqiu, Conroy Lindsey R, Hawkinson Tara R, et al. 2021. “Brain Glycogen Serves as a Critical Glucosamine Cache Required for Protein Glycosylation.” Cell Metabolism, May. 10.1016/j.cmet.2021.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki Akinobu, Stern Sarah A, Bozdagi Ozlem, Huntley George W, Walker Ruth H, Magistretti Pierre J, and Alberini Cristina M. 2011. “Astrocyte-Neuron Lactate Transport Is Required for Long-Term Memory Formation.” Cell 144 (5): 810–23. 10.1016/j.cell.2011.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swanson RA. 1992. “Physiologic Coupling of Glial Glycogen Metabolism to Neuronal Activity in Brain.” Canadian Journal of Physiology and Pharmacology 70 Suppl: S138–44. 10.1139/y92-255. [DOI] [PubMed] [Google Scholar]
- Swanson RA, Yu AC, Sharp FR, and Chan PH. 1989. “Regulation of Glycogen Content in Primary Astrocyte Culture: Effects of Glucose Analogues, Phenobarbital, and Methionine Sulfoximine.” Journal of Neurochemistry 52 (5): 1359–65. 10.1111/j.1471-4159.1989.tb09180.x. [DOI] [PubMed] [Google Scholar]
- Tagliabracci Vincent S, Turnbull Julie, Wang Wei, Girard Jean-Marie, Zhao Xiaochu, Skurat Alexander V, Delgado-Escueta Antonio V, Minassian Berge A, DePaoli-Roach Anna A, and Roach Peter J. 2007. “Laforin Is a Glycogen Phosphatase, Deficiency of Which Leads to Elevated Phosphorylation of Glycogen in Vivo.” Proceedings of the National Academy of Sciences 104 (49): 19262–66. 10.1073/pnas.0707952104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang Buyun, Frasinyuk Mykhaylo S, Chikwana Vimbai M, Mahalingan Krishna K, Morgan Cynthia A, Segvich Dyann M, Bondarenko Svitlana P, et al. 2020. “Discovery and Development of Small-Molecule Inhibitors of Glycogen Synthase.” Journal of Medicinal Chemistry 63 (7): 3538–51. 10.1021/acs.jmedchem.9b01851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tarentino AL, and Maley F. 1976. “Direct Evidence That D-Galactosamine Incorporation into Glycogen Occurs via UDP-Glucosamine.” FEBS Letters 69 (1): 175–78. 10.1016/0014-5793(76)80680-1. [DOI] [PubMed] [Google Scholar]
- Tegtmeyer Laura C, Rust Stephan, van Scherpenzeel Monique, Ng Bobby G, Losfeld Marie-Estelle, Timal Sharita, Raymond Kimiyo, et al. 2014. “Multiple Phenotypes in Phosphoglucomutase 1 Deficiency.” The New England Journal of Medicine 370 (6): 533–42. 10.1056/NEJMoa1206605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Therapeutics Maze. 2022. “Maze Therapeutics to Present Preclinical Data Highlighting MZE001 as a Potential Treatment for Pompe Disease at 2022 WORLD Symposium.” 2022. https://mazetx.com/maze-therapeutics-to-present-preclinical-data-highlighting-mze001-as-a-potential-treatment-for-pompe-disease-at-2022-world-symposium/. [Google Scholar]
- Unnikrishnan Ashwin, Gafken Philip R, and Tsukiyama Toshio. 2010. “Dynamic Changes in Histone Acetylation Regulate Origins of DNA Replication.” Nature Structural & Molecular Biology 17 (4): 430–37. 10.1038/nsmb.1780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vilchez David, Ros Susana, Cifuentes Daniel, Pujadas Lluís, Jordi Vallès Belén García-Fojeda, Criado-García Olga, et al. 2007. “Mechanism Suppressing Glycogen Synthesis in Neurons and Its Demise in Progressive Myoclonus Epilepsy.” Nature Neuroscience 10 (11): 1407–13. 10.1038/nn1998. [DOI] [PubMed] [Google Scholar]
- Waitt Alice E, Reed Liam, Ransom Bruce R, and Brown Angus M. 2017. “Emerging Roles for Glycogen in the CNS.” Frontiers in Molecular Neuroscience 10: 73. 10.3389/fnmol.2017.00073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Dong, Pascual Juan M, and De Vivo Darryl. 1993. “Glucose Transporter Type 1 Deficiency Syndrome.” In , edited by Adam Margaret P, Everman David B, Mirzaa Ghayda M, Pagon Roberta A, Wallace Stephanie E, Bean Lora J H, Gripp Karen W, and Amemiya Anne. Seattle (WA). [PubMed] [Google Scholar]
- Weisbart Richard H, Chan Grace, Jordaan Gwen, Noble Philip W, Liu Yanfeng, Glazer Peter M, Nishimura Robert N, and Hansen James E. 2015. “DNA-Dependent Targeting of Cell Nuclei by a Lupus Autoantibody.” Scientific Reports 5 (July): 12022. 10.1038/srep12022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weisbart Richard H, Gera Joseph F, Chan Grace, Hansen James E, Li Erica, Cloninger Cheri, Levine Arnold J, and Nishimura Robert N. 2012. “A Cell-Penetrating Bispecific Antibody for Therapeutic Regulation of Intracellular Targets.” Molecular Cancer Therapeutics 11 (10): 2169–73. 10.1158/1535-7163.MCT-12-0476-T. [DOI] [PubMed] [Google Scholar]
- Wens Stephan C A, Kuperus Esther, Mattace-Raso Francesco U S, Kruijshaar Michelle E, Brusse Esther, van Montfort Kees C A G M, Scheltens- de Boer Marjan, Sijbrands Eric J G, van der Ploeg Ans T, and van Doorn Pieter A. 2014. “Increased Aortic Stiffness and Blood Pressure in Non-Classic Pompe Disease.” Journal of Inherited Metabolic Disease 37 (3): 391–97. 10.1007/s10545-013-9667-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willstätter R, and Rohdewald M. 1934. “Über Den Zustand Des Glykogens in Der Leber, Im Muskel Und in Leukocyten.” Hoppe-Seyleŕs Zeitschrift Für Physiologische Chemie (Zur Kenntnis Der Proteinbindung Physiologisch Wichtiger Stoffe.). 225 (2–3): 103–124. [Google Scholar]
- Wilson Wayne A, Wang Zhong, and Roach Peter J. 2002. “Systematic Identification of the Genes Affecting Glycogen Storage in the Yeast Saccharomyces Cerevisiae.” Molecular and Cellular Proteomics 1 (3): 232–42. 10.1074/mcp.M100024-MCP200. [DOI] [PubMed] [Google Scholar]
- Wu Long, Wong Candance P, and Swanson Raymond A. 2019. “Methodological Considerations for Studies of Brain Glycogen.” Journal of Neuroscience Research 97 (8): 914–22. 10.1002/jnr.24412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Junnan, Song Dan, Xue Zhanxia, Gu Li, Hertz Leif, and Peng Liang. 2013. “Requirement of Glycogenolysis for Uptake of Increased Extracellular K+ in Astrocytes: Potential Implications for K+ Homeostasis and Glycogen Usage in Brain.” Neurochemical Research 38 (3): 472–85. 10.1007/s11064-012-0938-3. [DOI] [PubMed] [Google Scholar]
- Yeo Mildrid, Moawad Hager, and Grunewald Stephanie. 2023. “Disorders of Carbohydrate Metabolism: A Review of Glycogen Storage Disorders.” Paediatrics and Child Health. https://doi.org/ 10.1016/j.paed.2022.12.007. [DOI] [Google Scholar]
- Young Lyndsay E A, Brizzee Corey O, Macedo Jessica K A, Murphy Robert D, Contreras Christopher J, DePaoli-Roach Anna A, Roach Peter J, Gentry Matthew S, and Sun Ramon C. 2020. “Accurate and Sensitive Quantitation of Glucose and Glucose Phosphates Derived from Storage Carbohydrates by Mass Spectrometry.” Carbohydrate Polymers 230 (February): 115651. 10.1016/j.carbpol.2019.115651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young Lyndsay E A, Conroy Lindsey R, Clarke Harrison A, Hawkinson Tara R, Bolton Kayli E, Sanders William C, Chang Josephine E, et al. 2022. “In Situ Mass Spectrometry Imaging Reveals Heterogeneous Glycogen Stores in Human Normal and Cancerous Tissues.” EMBO Molecular Medicine 14 (11): e16029. 10.15252/emmm.202216029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zebhauser Paul Theo, Cordts Isabell, Hengel Holger, Haslinger Bernhard, Lingor Paul, Akman Hasan Orhan, Haack Tobias B, and Deschauer Marcus. 2022. “Characterization of Cognitive Impairment in Adult Polyglucosan Body Disease.” Journal of Neurology 269 (6): 2854–61. 10.1007/s00415-022-10960-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Yang, van Zijl Peter C.M., Xu Xiang, Xu Jiadi, Li Yuguo, Chen Lin, and Yadav Nirbhay N.. 2020. “Magnetic Resonance Imaging of Glycogen Using Its Magnetic Coupling with Water.” Proceedings of the National Academy of Sciences of the United States of America 117 (6): 3144–49. 10.1073/pnas.1909921117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Zhengqiu, Kinslow Connor J, Hibshoosh Hanina, Guo Hua, Cheng Simon K, He Chunyan, Gentry Matthew S, and Sun Ramon C. 2019. “Clinical Features, Survival and Prognostic Factors of Glycogen-Rich Clear Cell Carcinoma (GRCC) of the Breast in the U.S. Population.” Journal of Clinical Medicine 8 (2). 10.3390/jcm8020246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zois Christos E, Favaro Elena, and Harris Adrian L. 2014. “Glycogen Metabolism in Cancer.” Biochemical Pharmacology 92 (1): 3–11. 10.1016/j.bcp.2014.09.001. [DOI] [PubMed] [Google Scholar]




