Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Vis Exp ; (193)2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-37602854

RESUMEN

ARTICLES DISCUSSED: Wilson, W. A. Spectrophotometric methods for the study of eukaryotic glycogen metabolism. Journal of Visualized Experiments. (174), e63046 (2021). Wang, J. J. et al. A non-degradative extraction method for molecular structure characterization of bacterial glycogen particles. Journal of Visualized Experiments. (180), e63016 (2022). Wang, Z., Liu, Q., Wang, L., Gilbert, R. G., Sullivan, M. A. The extraction of liver glycogen molecules for glycogen structure determination. Journal of Visualized Experiments. (180), e63088 (2022). Jensen, R., Ortenblad, N., di Benedetto, C., Qvortrup, K., Nielsen, J. Quantification of subcellular glycogen distribution in skeletal muscle fibers using transmission electron microscopy. Journal of Visualized Experiments. (180), e63347 (2022). Fermont, L., Szydlowski, N., Colleoni, C. Determination of glucan chain length distribution of glycogen using the fluorophore-assisted carbohydrate electrophoresis (FACE) method. Journal of Visualized Experiments. (181), e63392 (2022).


Asunto(s)
Eucariontes , Células Eucariotas , Citoplasma , Colorantes Fluorescentes , Glucógeno
2.
Ann Clin Transl Neurol ; 7(11): 2186-2198, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33034425

RESUMEN

OBJECTIVE: Adult polyglucosan body disease (APBD) is an adult-onset neurological variant of glycogen storage disease type IV. APBD is caused by recessive mutations in the glycogen branching enzyme gene, and the consequent accumulation of poorly branched glycogen aggregates called polyglucosan bodies in the nervous system. There are presently no treatments for APBD. Here, we test whether downregulation of glycogen synthesis is therapeutic in a mouse model of the disease. METHODS: We characterized the effects of knocking out two pro-glycogenic proteins in an APBD mouse model. APBD mice were crossed with mice deficient in glycogen synthase (GYS1), or mice deficient in protein phosphatase 1 regulatory subunit 3C (PPP1R3C), a protein involved in the activation of GYS1. Phenotypic and histological parameters were analyzed and glycogen was quantified. RESULTS: APBD mice deficient in GYS1 or PPP1R3C demonstrated improvements in life span, morphology, and behavioral assays of neuromuscular function. Histological analysis revealed a reduction in polyglucosan body accumulation and of astro- and micro-gliosis in the brains of GYS1- and PPP1R3C-deficient APBD mice. Brain glycogen quantification confirmed the reduction in abnormal glycogen accumulation. Analysis of skeletal muscle, heart, and liver found that GYS1 deficiency reduced polyglucosan body accumulation in all three tissues and PPP1R3C knockout reduced skeletal muscle polyglucosan bodies. INTERPRETATION: GYS1 and PPP1R3C are effective therapeutic targets in the APBD mouse model. These findings represent a critical step toward the development of a treatment for APBD and potentially other glycogen storage disease type IV patients.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno/metabolismo , Glucógeno Sintasa/deficiencia , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Enfermedades del Sistema Nervioso/metabolismo , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Enfermedad del Almacenamiento de Glucógeno/fisiopatología , Enfermedad del Almacenamiento de Glucógeno/terapia , Ratones , Ratones Noqueados , Enfermedades del Sistema Nervioso/fisiopatología , Enfermedades del Sistema Nervioso/terapia
3.
Adv Neurobiol ; 23: 83-123, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31667806

RESUMEN

Brain glycogen synthesis is a regulated, multi-step process that begins with glucose transport across the blood brain barrier and culminates with the actions of glycogen synthase and the glycogen branching enzyme to elongate glucose chains and introduce branch points in a growing glycogen molecule. This review focuses on the synthesis of glycogen in the brain, with an emphasis on glycogen synthase, but draws on salient studies in mammalian muscle and liver as well as baker's yeast, with the goal of providing a more comprehensive view of glycogen synthesis and highlighting potential areas for further study in the brain. In addition, deficiencies in the glycogen biosynthetic enzymes which lead to glycogen storage diseases in humans are discussed, highlighting effects on the brain and discussing findings in genetically modified animal models that recapitulate these diseases. Finally, implications of glycogen synthesis in neurodegenerative and other diseases that impact the brain are presented.


Asunto(s)
Encéfalo/enzimología , Glucógeno Sintasa/química , Glucógeno Sintasa/metabolismo , Animales , Glucosa/metabolismo , Glucógeno/metabolismo , Humanos
4.
Cell Rep ; 29(6): 1410-1418.e6, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693883

RESUMEN

Browning induction or transplantation of brown adipose tissue (BAT) or brown/beige adipocytes derived from progenitor or induced pluripotent stem cells (iPSCs) can represent a powerful strategy to treat metabolic diseases. However, our poor understanding of the mechanisms that govern the differentiation and activation of brown adipocytes limits the development of such therapy. Various genetic factors controlling the differentiation of brown adipocytes have been identified, although most studies have been performed using in vitro cultured pre-adipocytes. We investigate here the differentiation of brown adipocytes from adipose progenitors in the mouse embryo. We demonstrate that the formation of multiple lipid droplets (LDs) is initiated within clusters of glycogen, which is degraded through glycophagy to provide the metabolic substrates essential for de novo lipogenesis and LD formation. Therefore, this study uncovers the role of glycogen in the generation of LDs.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis/genética , Tejido Adiposo Pardo/metabolismo , Embrión de Mamíferos/metabolismo , Glucógeno/metabolismo , Gotas Lipídicas/metabolismo , Adipocitos Marrones/ultraestructura , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/ultraestructura , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Glucógeno/ultraestructura , Humanos , Gotas Lipídicas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , PPAR gamma/genética , PPAR gamma/metabolismo , ARN Interferente Pequeño , Transcriptoma
5.
Ann Neurol ; 74(2): 297-300, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23913475

RESUMEN

Lafora disease (LD) is a fatal progressive myoclonus epilepsy characterized neuropathologically by aggregates of abnormally structured glycogen and proteins (Lafora bodies [LBs]), and neurodegeneration. Whether LBs could be prevented by inhibiting glycogen synthesis and whether they are pathogenic remain uncertain. We genetically eliminated brain glycogen synthesis in LD mice. This resulted in long-term prevention of LB formation, neurodegeneration, and seizure susceptibility. This study establishes that glycogen synthesis is requisite for LB formation and that LBs are pathogenic. It opens a therapeutic window for potential treatments in LD with known and future small molecule inhibitors of glycogen synthesis.


Asunto(s)
Glucógeno/antagonistas & inhibidores , Glucógeno/biosíntesis , Enfermedad de Lafora/prevención & control , Animales , Modelos Animales de Enfermedad , Fosfatasas de Especificidad Dual/genética , Técnicas de Inactivación de Genes , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Enfermedad de Lafora/patología , Enfermedad de Lafora/fisiopatología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Tirosina Fosfatasas no Receptoras
6.
J Neurosci Res ; 89(4): 585-91, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21259334

RESUMEN

Brain glycogen is proposed to function under both physiological and pathological conditions. Pharmacological elevation of this glucose polymer in brain is hypothesized to protect neurons against hypoglycemia-induced cell death. Elevation of brain glycogen levels due to prior hypoglycemia is postulated to contribute to the development of hypoglycemia-associated autonomic failure (HAAF) in insulin-treated diabetic patients. This latter mode of elevating glycogen levels is termed "supercompensation." We tested whether brain glycogen supercompensation occurs in healthy, conscious mice after recovery from insulin-induced acute or recurrent hypoglycemia. Blood glucose levels were lowered to less than 2.2 mmol/liter for 90 min by administration of insulin. Brain glucose levels decreased at least 80% and brain glycogen levels decreased approximately 50% after episodes of either acute or recurrent hypoglycemia. After these hypoglycemic episodes, mice were allowed access to food for 6 or 27 hr. After 6 hr, blood and brain glucose levels were restored but brain glycogen levels were elevated by 25% in mice that had been subjected to either acute or recurrent hypoglycemia compared with saline-treated controls. After a 27-hr recovery period, the concentration of brain glycogen had returned to baseline levels in mice previously subjected to either acute or recurrent hypoglycemia. We conclude that brain glycogen supercompensation occurs in healthy mice, but its functional significance remains to be established.


Asunto(s)
Encéfalo/metabolismo , Glucógeno/metabolismo , Hipoglucemia/metabolismo , Animales , Encéfalo/fisiopatología , Glucosa/metabolismo , Hipoglucemia/inducido químicamente , Hipoglucemia/fisiopatología , Hipoglucemiantes/farmacología , Insulina/farmacología , Masculino , Ratones
7.
PLoS Med ; 5(1): e27, 2008 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-18232732

RESUMEN

BACKGROUND: Stored glycogen is an important source of energy for skeletal muscle. Human genetic disorders primarily affecting skeletal muscle glycogen turnover are well-recognised, but rare. We previously reported that a frameshift/premature stop mutation in PPP1R3A, the gene encoding RGL, a key regulator of muscle glycogen metabolism, was present in 1.36% of participants from a population of white individuals in the UK. However, the functional implications of the mutation were not known. The objective of this study was to characterise the molecular and physiological consequences of this genetic variant. METHODS AND FINDINGS: In this study we found a similar prevalence of the variant in an independent UK white population of 744 participants (1.46%) and, using in vivo (13)C magnetic resonance spectroscopy studies, demonstrate that human carriers (n = 6) of the variant have low basal (65% lower, p = 0.002) and postprandial muscle glycogen levels. Mice engineered to express the equivalent mutation had similarly decreased muscle glycogen levels (40% lower in heterozygous knock-in mice, p < 0.05). In muscle tissue from these mice, failure of the truncated mutant to bind glycogen and colocalize with glycogen synthase (GS) decreased GS and increased glycogen phosphorylase activity states, which account for the decreased glycogen content. CONCLUSIONS: Thus, PPP1R3A C1984DeltaAG (stop codon 668) is, to our knowledge, the first prevalent mutation described that directly impairs glycogen synthesis and decreases glycogen levels in human skeletal muscle. The fact that it is present in approximately 1 in 70 UK whites increases the potential biomedical relevance of these observations.


Asunto(s)
Codón sin Sentido , Mutación del Sistema de Lectura , Glucógeno/biosíntesis , Músculo Esquelético/enzimología , Fosfoproteínas Fosfatasas/fisiología , Adulto , Animales , Diabetes Mellitus Tipo 2/enzimología , Femenino , Frecuencia de los Genes , Glucógeno/análisis , Glucógeno Fosforilasa/metabolismo , Glucógeno Sintasa/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Datos de Secuencia Molecular , Músculo Esquelético/química , Fosfoproteínas Fosfatasas/genética , Periodo Posprandial , Relación Estructura-Actividad , Reino Unido , Población Blanca/genética
8.
Biochem J ; 395(1): 137-45, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16356168

RESUMEN

Glycogen, a branched polymer of glucose, forms an energy re-serve in numerous organisms. In mammals, the two largest glyco-gen stores are in skeletal muscle and liver, which express tissue-specific glycogen synthase isoforms. MGSKO mice, in which mGys1 (mouse glycogen synthase) is disrupted, are devoid of muscle glycogen [Pederson, Chen, Schroeder, Shou, DePaoli-Roach and Roach (2004) Mol. Cell. Biol. 24, 7179-7187]. The GSL30 mouse line hyper-accumulates glycogen in muscle [Manchester, Skurat, Roach, Hauschka and Lawrence (1996) Proc. Natl. Acad. Sci. U.S.A. 93, 10707-10711]. We performed a microarray analysis of mRNA from the anterior tibialis, medial gastrocnemius and liver of MGSKO mice, and from the gastroc-nemius of GSL30 mice. In MGSKO mice, transcripts of 79 genes varied in their expression in the same direction in both the anterior tibialis and gastrocnemius. These included several genes encoding proteins proximally involved in glycogen metabolism. The Ppp1r1a [protein phosphatase 1 regulatory (inhibitor) sub-unit 1A] gene underwent the greatest amount of downregulation. In muscle, the downregulation of Pfkfb1 and Pfkfb3, encoding isoforms of 6-phosphofructo-2-kinase/fructose-2,6-bisphospha-tase, is consistent with decreased glycolysis. Pathways for branched-chain amino acid, and ketone body utilization appear to be downregulated, as is the capacity to form the gluconeogenic precursors alanine, lactate and glutamine. Expression changes among several members of the Wnt signalling pathway were identified, suggesting an as yet unexplained role in glycogen meta-bolism. In liver, the upregulation of Pfkfb1 and Pfkfb3 expression is consistent with increased glycolysis, perhaps as an adaptation to altered muscle metabolism. By comparing changes in muscle expression between MGSKO and GSL30 mice, we found a subset of 44 genes, the expression of which varied as a function of muscle glycogen content. These genes are candidates for regulation by glycogen levels. Particularly interesting is the observation that 11 of these genes encode cardiac or slow-twitch isoforms of muscle contractile proteins, and are upregulated in muscle that has a greater oxidative capacity in MGSKO mice.


Asunto(s)
Perfilación de la Expresión Génica , Glucógeno/metabolismo , Músculo Esquelético/metabolismo , Animales , Regulación de la Expresión Génica , Glucógeno Sintasa/deficiencia , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Músculo Esquelético/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Proteínas Wnt/genética
9.
Diabetes ; 54(12): 3466-73, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16306363

RESUMEN

Glycogen is an important component of whole-body glucose metabolism. MGSKO mice lack skeletal muscle glycogen due to disruption of the GYS1 gene, which encodes muscle glycogen synthase. MGSKO mice were 5-10% smaller than wild-type littermates with less body fat. They have more oxidative muscle fibers and, based on the activation state of AMP-activated protein kinase, more capacity to oxidize fatty acids. Blood glucose in fed and fasted MGSKO mice was comparable to wild-type littermates. Serum insulin was lower in fed but not in fasted MGSKO animals. In a glucose tolerance test, MGSKO mice disposed of glucose more effectively than wild-type animals and had a more sustained elevation of serum insulin. This result was not explained by increased conversion to serum lactate or by enhanced storage of glucose in the liver. However, glucose infusion rate in a euglycemic-hyperinsulinemic clamp was normal in MGSKO mice despite diminished muscle glucose uptake. During the clamp, MGSKO animals accumulated significantly higher levels of liver glycogen as compared with wild-type littermates. Although disruption of the GYS1 gene negatively affects muscle glucose uptake, overall glucose tolerance is actually improved, possibly because of a role for GYS1 in tissues other than muscle.


Asunto(s)
Glucosa/metabolismo , Glucógeno Sintasa/deficiencia , Animales , Grasas de la Dieta/farmacología , Femenino , Prueba de Tolerancia a la Glucosa , Crecimiento , Insulina/sangre , Glucógeno Hepático/metabolismo , Masculino , Ratones , Ratones Noqueados , Fibras Musculares Esqueléticas/enzimología , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/enzimología , Músculo Esquelético/patología
10.
Biochem Biophys Res Commun ; 331(2): 491-6, 2005 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-15850786

RESUMEN

Skeletal muscle glycogen is considered to be an important source of energy for contraction and increasing the level of the glucose polymer is generally thought to improve exercise performance in humans. A genetically modified mouse model (GSL30), which overaccumulates glycogen due to overexpression of a hyperactive form of glycogen synthase, was used to examine whether increasing the level of the polysaccharide enhances the ability of mice to run on a treadmill. The skeletal muscle of the GSL30 mice had large deposits of glycogen. There were no significant increases in the work performed by GSL30 mice as compared to their respective wild type littermates when exercised to exhaustion. The amount of muscle glycogen utilized by GSL30 mice, however, was greater, while the amount of liver glycogen consumed during exhaustive exercise was less than wild type animals. This result suggests that increased muscle glycogen stores do not necessarily improve exercise performance in mice.


Asunto(s)
Glucógeno/metabolismo , Músculo Esquelético/metabolismo , Condicionamiento Físico Animal/fisiología , Animales , Glucemia/análisis , Prueba de Esfuerzo , Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Ácido Láctico/sangre , Hígado/metabolismo , Ratones , Músculo Esquelético/citología , Músculo Esquelético/fisiología
11.
J Biol Chem ; 280(17): 17260-5, 2005 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-15711014

RESUMEN

The glucose storage polymer glycogen is generally considered to be an important source of energy for skeletal muscle contraction and a factor in exercise endurance. A genetically modified mouse model lacking muscle glycogen was used to examine whether the absence of the polysaccharide affects the ability of mice to run on a treadmill. The MGSKO mouse has the GYS1 gene, encoding the muscle isoform of glycogen synthase, disrupted so that skeletal muscle totally lacks glycogen. The morphology of the soleus and quadriceps muscles from MGSKO mice appeared normal. MGSKO-null mice, along with wild type littermates, were exercised to exhaustion. There were no significant differences in the work performed by MGSKO mice as compared with their wild type littermates. The amount of liver glycogen consumed during exercise was similar for MGSKO and wild type animals. Fasting reduced exercise endurance, and after overnight fasting, there was a trend to reduced exercise endurance for the MGSKO mice. These studies provide genetic evidence that in mice muscle glycogen is not essential for strenuous exercise and has relatively little effect on endurance.


Asunto(s)
Glucógeno Sintasa/genética , Glucógeno Sintasa/metabolismo , Músculos/enzimología , Condicionamiento Físico Animal , Animales , Glucemia/metabolismo , Peso Corporal , Femenino , Glucógeno/química , Glucógeno/metabolismo , Glucólisis , Heterocigoto , Lactatos/metabolismo , Hígado/metabolismo , Glucógeno Hepático/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Polímeros/química , Polisacáridos/química , Factores de Tiempo
12.
Mol Cell Biol ; 24(16): 7179-87, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15282316

RESUMEN

Glycogen serves as a repository of glucose in many mammalian tissues. Mice lacking this glucose reserve in muscle, heart, and several other tissues were generated by disruption of the GYS1 gene, which encodes an isoform of glycogen synthase. Crossing mice heterozygous for the GYS1 disruption resulted in a significant underrepresentation of GYS1-null mice in the offspring. Timed matings established that Mendelian inheritance was followed for up to 18.5 days postcoitum (dpc) and that approximately 90% of GYS1-null animals died soon after birth due to impaired cardiac function. Defects in cardiac development began between 11.5 and 14.5 dpc. At 18.5 dpc, the hearts were significantly smaller, with reduced ventricular chamber size and enlarged atria. Consistent with impaired cardiac function, edema, pooling of blood, and hemorrhagic liver were seen. Glycogen synthase and glycogen were undetectable in cardiac muscle and skeletal muscle from the surviving null mice, and the hearts showed normal morphology and function. Congenital heart disease is one of the most common birth defects in humans, at up to 1 in 50 live births. The results provide the first direct evidence that the ability to synthesize glycogen in cardiac muscle is critical for normal heart development and hence that its impairment could be a significant contributor to congenital heart defects.


Asunto(s)
Glucógeno Sintasa/metabolismo , Glucógeno/metabolismo , Corazón/crecimiento & desarrollo , Miocardio/metabolismo , Isoformas de Proteínas/metabolismo , Animales , Ecocardiografía , Femenino , Glucógeno Sintasa/genética , Corazón/anatomía & histología , Corazón/fisiología , Humanos , Hígado/citología , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/química , Miocardio/citología , Isoformas de Proteínas/genética , Tasa de Supervivencia , Transgenes
13.
J Biol Chem ; 279(14): 13764-8, 2004 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-14742447

RESUMEN

Glycogen is a storage form of glucose utilized as an energy reserve by many organisms. Glycogen synthase, which is essential for synthesizing this glucose polymer, is regulated by both covalent phosphorylation and the concentration of glucose-6-P. With the yeast glycogen synthase Gsy2p, we recently identified two mutants, R579A/R580A/R582A [corrected] and R586A/R588A/R591A, in which multiple arginine residues were mutated to alanine that were completely insensitive to activation by glucose-6-P in vitro (Pederson, B. A., Cheng, C., Wilson, W. A., and Roach, P. J. (2000) J. Biol. Chem. 275, 27753-27761). We report here the expression of these mutants in Saccharomyces cerevisiae and, as expected from our findings in vitro, they were not activated by glucose-6-P. The R579A/R580A/R582A [corrected] mutant, which is also resistant to inhibition by phosphorylation, caused hyperaccumulation of glycogen. In contrast, the mutant R586A/R588A/R591A, which retains the ability to be inactivated by phosphorylation, resulted in lower glycogen accumulation when compared with wild-type cells. When intracellular glucose-6-P levels were increased by mutating the PFK2 gene, glycogen storage due to the wild-type enzyme was increased, whereas that associated with R579A/R580A/R582A [corrected] was not greatly changed. This is the first direct demonstration that activation of glycogen synthase by glucose-6-P in vivo is necessary for normal glycogen accumulation.


Asunto(s)
Glucosa-6-Fosfato/metabolismo , Glucógeno Sintasa/metabolismo , Glucógeno/metabolismo , Saccharomyces cerevisiae/enzimología , Glucógeno Sintasa/genética , Mutagénesis , Fosforilación , Plásmidos , Saccharomyces cerevisiae/genética
14.
Biochem Biophys Res Commun ; 305(4): 826-30, 2003 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12767905

RESUMEN

Glycogen, a branched polymer of glucose, serves as an energy reserve in many organisms. The degree of branching likely reflects the balance between the activities of glycogen synthase and branching enzyme. Mice overexpressing constitutively active glycogen synthase in skeletal muscle (GSL30) have elevated muscle glycogen. To test whether excess glycogen synthase activity affected glycogen branching, we examined the glycogen from skeletal muscle of GSL30 mice. The absorption spectrum of muscle glycogen determined in the presence of iodine was shifted to higher wavelengths in the GSL30 animals, consistent with a decrease in the degree of branching. As judged by Western blotting, the levels of glycogenin and the branching enzyme were also elevated. Branching enzyme activity also increased approximately threefold. However, this compared with an increase in glycogen synthase of some 50-fold, so that the increase in branching enzyme in response to overexpression of glycogen synthase was insufficient to synthesize normally branched glycogen.


Asunto(s)
Glucógeno Sintasa/metabolismo , Glucógeno/biosíntesis , Glucógeno/química , Músculo Esquelético/enzimología , Enzima Ramificadora de 1,4-alfa-Glucano/metabolismo , Animales , Glucosiltransferasas , Glucógeno Sintasa/genética , Glicoproteínas/metabolismo , Ratones , Músculo Esquelético/metabolismo , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...