Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Gastroenterology ; 160(5): 1725-1740.e2, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33309778

RESUMO

BACKGROUND & AIMS: We recently showed that alcoholic hepatitis (AH) is characterized by dedifferentiation of hepatocytes and loss of mature functions. Glucose metabolism is tightly regulated in healthy hepatocytes. We hypothesize that AH may lead to metabolic reprogramming of the liver, including dysregulation of glucose metabolism. METHODS: We performed integrated metabolomic and transcriptomic analyses of liver tissue from patients with AH or alcoholic cirrhosis or normal liver tissue from hepatic resection. Focused analyses of chromatin immunoprecipitation coupled to DNA sequencing was performed. Functional in vitro studies were performed in primary rat and human hepatocytes and HepG2 cells. RESULTS: Patients with AH exhibited specific changes in the levels of intermediates of glycolysis/gluconeogenesis, the tricarboxylic acid cycle, and monosaccharide and disaccharide metabolism. Integrated analysis of the transcriptome and metabolome showed the used of alternate energetic pathways, metabolite sinks and bottlenecks, and dysregulated glucose storage in patients with AH. Among genes involved in glucose metabolism, hexokinase domain containing 1 (HKDC1) was identified as the most up-regulated kinase in patients with AH. Histone active promoter and enhancer markers were increased in the HKDC1 genomic region. High HKDC1 levels were associated with the development of acute kidney injury and decreased survival. Increased HKDC1 activity contributed to the accumulation of glucose-6-P and glycogen in primary rat hepatocytes. CONCLUSIONS: Altered metabolite levels and messenger RNA expression of metabolic enzymes suggest the existence of extensive reprogramming of glucose metabolism in AH. Increased HKDC1 expression may contribute to dysregulated glucose metabolism and represents a novel biomarker and therapeutic target for AH.


Assuntos
Desdiferenciação Celular , Metabolismo Energético , Perfilação da Expressão Gênica , Glucose/metabolismo , Hepatite Alcoólica/enzimologia , Hepatócitos/enzimologia , Hexoquinase/metabolismo , Fígado/enzimologia , Metabolômica , Injúria Renal Aguda/enzimologia , Injúria Renal Aguda/genética , Adaptação Fisiológica , Animais , Europa (Continente) , Feminino , Regulação Enzimológica da Expressão Gênica , Glucose-6-Fosfato/metabolismo , Glicogênio/metabolismo , Células Hep G2 , Hepatite Alcoólica/genética , Hepatite Alcoólica/patologia , Hepatócitos/patologia , Hexoquinase/genética , Humanos , Fígado/patologia , Masculino , Metaboloma , Pessoa de Meia-Idade , Ratos Wistar , Transcriptoma , Estados Unidos
2.
Proc Natl Acad Sci U S A ; 111(28): E2831-40, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982189

RESUMO

Glycogen is a primary form of energy storage in eukaryotes that is essential for glucose homeostasis. The glycogen polymer is synthesized from glucose through the cooperative action of glycogen synthase (GS), glycogenin (GN), and glycogen branching enzyme and forms particles that range in size from 10 to 290 nm. GS is regulated by allosteric activation upon glucose-6-phosphate binding and inactivation by phosphorylation on its N- and C-terminal regulatory tails. GS alone is incapable of starting synthesis of a glycogen particle de novo, but instead it extends preexisting chains initiated by glycogenin. The molecular determinants by which GS recognizes self-glucosylated GN, the first step in glycogenesis, are unknown. We describe the crystal structure of Caenorhabditis elegans GS in complex with a minimal GS targeting sequence in GN and show that a 34-residue region of GN binds to a conserved surface on GS that is distinct from previously characterized allosteric and binding surfaces on the enzyme. The interaction identified in the GS-GN costructure is required for GS-GN interaction and for glycogen synthesis in a cell-free system and in intact cells. The interaction of full-length GS-GN proteins is enhanced by an avidity effect imparted by a dimeric state of GN and a tetrameric state of GS. Finally, the structure of the N- and C-terminal regulatory tails of GS provide a basis for understanding phosphoregulation of glycogen synthesis. These results uncover a central molecular mechanism that governs glycogen metabolism.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans/enzimologia , Glucosiltransferases , Glicogênio Sintase , Glicoproteínas , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Sistema Livre de Células , Células Cultivadas , Cristalografia por Raios X , Glucosiltransferases/química , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Glicogênio/biossíntese , Glicogênio/química , Glicogênio/genética , Glicogênio Sintase/química , Glicogênio Sintase/genética , Glicogênio Sintase/metabolismo , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilação , Camundongos , Camundongos Knockout , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Relação Estrutura-Atividade
3.
Diabetologia ; 59(5): 1012-20, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26825527

RESUMO

AIMS/HYPOTHESIS: Glycogen accumulation occurs in beta cells of diabetic patients and has been proposed to partly mediate glucotoxicity-induced beta cell dysfunction. However, the role of glycogen metabolism in beta cell function and its contribution to diabetes pathophysiology remain poorly understood. We investigated the function of beta cell glycogen by studying glucose homeostasis in mice with (1) defective glycogen synthesis in the pancreas; and (2) excessive glycogen accumulation in beta cells. METHODS: Conditional deletion of the Gys1 gene and overexpression of protein targeting to glycogen (PTG) was accomplished by Cre-lox recombination using pancreas-specific Cre lines. Glucose homeostasis was assessed by determining fasting glycaemia, insulinaemia and glucose tolerance. Beta cell mass was determined by morphometry. Glycogen was detected histologically by periodic acid-Schiff's reagent staining. Isolated islets were used for the determination of glycogen and insulin content, insulin secretion, immunoblots and gene expression assays. RESULTS: Gys1 knockout (Gys1 (KO)) mice did not exhibit differences in glucose tolerance or basal glycaemia and insulinaemia relative to controls. Insulin secretion and gene expression in isolated islets was also indistinguishable between Gys1 (KO) and controls. Conversely, despite effective glycogen overaccumulation in islets, mice with PTG overexpression (PTG(OE)) presented similar glucose tolerance to controls. However, under fasting conditions they exhibited lower glycaemia and higher insulinaemia. Importantly, neither young nor aged PTG(OE) mice showed differences in beta cell mass relative to age-matched controls. Finally, a high-fat diet did not reveal a beta cell-autonomous phenotype in either model. CONCLUSIONS/INTERPRETATION: Glycogen metabolism is not required for the maintenance of beta cell function. Glycogen accumulation in beta cells alone is not sufficient to trigger the dysfunction or loss of these cells, or progression to diabetes.


Assuntos
Glucose/metabolismo , Glicogênio/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Feminino , Glicogênio/fisiologia , Glicogênio Sintase/genética , Glicogênio Sintase/metabolismo , Homeostase , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Masculino , Camundongos , Camundongos Knockout
4.
Hum Mol Genet ; 23(12): 3147-56, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24452334

RESUMO

Lafora disease is a fatal neurodegenerative condition characterized by the accumulation of abnormal glycogen inclusions known as Lafora bodies. It is an autosomal recessive disorder caused by mutations in either the laforin or malin gene. To study whether glycogen is primarily responsible for the neurodegeneration in Lafora disease, we generated malin knockout mice with impaired (totally or partially) glycogen synthesis. These animals did not show the increase in markers of neurodegeneration, the impairments in electrophysiological properties of hippocampal synapses, nor the susceptibility to kainate-induced epilepsy seen in the malin knockout model. Interestingly, the autophagy impairment that has been described in malin knockout animals was also rescued in this double knockout model. Conversely, two other mouse models in which glycogen is over-accumulated in the brain independently of the lack of malin showed impairment in autophagy. Our findings reveal that glycogen accumulation accounts for the neurodegeneration and functional consequences seen in the malin knockout model, as well as the impaired autophagy. These results identify the regulation of glycogen synthesis as a key target for the treatment of Lafora disease.


Assuntos
Autofagia , Fosfatases de Especificidade Dupla/metabolismo , Glicogênio Sintase/genética , Glicogênio/metabolismo , Doença de Lafora/fisiopatologia , Ubiquitina-Proteína Ligases/genética , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Sinapses Elétricas/metabolismo , Epilepsia/induzido quimicamente , Epilepsia/patologia , Glicogênio Sintase/metabolismo , Hipocampo/fisiologia , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Ácido Caínico/farmacologia , Doença de Lafora/metabolismo , Doença de Lafora/patologia , Camundongos , Camundongos Knockout , Mutação , Proteínas Tirosina Fosfatases não Receptoras , Ubiquitina-Proteína Ligases/metabolismo
5.
Am J Physiol Regul Integr Comp Physiol ; 311(2): R307-14, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27280431

RESUMO

McArdle disease (muscle glycogenosis type V) is a disease caused by myophosphorylase deficiency leading to "blocked" glycogen breakdown. A significant but varying glycogen accumulation in especially distal hind limb muscles of mice affected by McArdle disease has recently been demonstrated. In this study, we investigated how myophosphorylase deficiency affects glucose metabolism in hind limb muscle of 20-wk-old McArdle mice and vastus lateralis muscles from patients with McArdle disease. Western blot analysis and activity assay demonstrated that glycogen synthase was inhibited in glycolytic muscle from McArdle mice. The level and activation of proteins involved in contraction-induced glucose transport (AMPK, GLUT4) and glycogen synthase inhibition were increased in quadriceps muscle of McArdle mice. In addition, pCaMKII in quadriceps was reduced, suggesting lower insulin-induced glucose uptake, which could lead to lower glycogen accumulation. In comparison, tibialis anterior, extensor digitorum longus, and soleus had massive glycogen accumulation, but few, if any, changes or adaptations in glucose metabolism compared with wild-type mice. The findings suggest plasticity in glycogen metabolism in the McArdle mouse that is related to myosin heavy chain type IIB content in muscles. In patients, the level of GLUT4 was vastly increased, as were hexokinase II and phosphofructokinase, and glycogen synthase was more inhibited, suggesting that patients adapt by increasing capture of glucose for direct metabolism, thereby significantly reducing glycogen buildup compared with the mouse model. Hence, the McArdle mouse may be a useful tool for further comparative studies of disease mechanism caused by myophosphorylase deficiency and basic studies of metabolic adaptation in muscle.


Assuntos
Glucose/metabolismo , Doença de Depósito de Glicogênio Tipo V/metabolismo , Complexos Multienzimáticos , Músculo Esquelético/metabolismo , Adolescente , Adulto , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Especificidade da Espécie , Adulto Jovem
6.
Biochem J ; 472(2): 225-37, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26417114

RESUMO

Understanding how glucose metabolism is finely regulated at molecular and cellular levels in the liver is critical for knowing its relationship to related pathologies, such as diabetes. In order to gain insight into the regulation of glucose metabolism, we studied the liver-expressed isoforms aldolase B and fructose-1,6-bisphosphatase-1 (FBPase-1), key enzymes in gluconeogenesis, analysing their cellular localization in hepatocytes under different metabolic conditions and their protein-protein interaction in vitro and in vivo. We observed that glucose, insulin, glucagon and adrenaline differentially modulate the intracellular distribution of aldolase B and FBPase-1. Interestingly, the in vitro protein-protein interaction analysis between aldolase B and FBPase-1 showed a specific and regulable interaction between them, whereas aldolase A (muscle isozyme) and FBPase-1 showed no interaction. The affinity of the aldolase B and FBPase-1 complex was modulated by intermediate metabolites, but only in the presence of K(+). We observed a decreased association constant in the presence of adenosine monophosphate, fructose-2,6-bisphosphate, fructose-6-phosphate and inhibitory concentrations of fructose-1,6-bisphosphate. Conversely, the association constant of the complex increased in the presence of dihydroxyacetone phosphate (DHAP) and non-inhibitory concentrations of fructose-1,6-bisphosphate. Notably, in vivo FRET studies confirmed the interaction between aldolase B and FBPase-1. Also, the co-expression of aldolase B and FBPase-1 in cultured cells suggested that FBPase-1 guides the cellular localization of aldolase B. Our results provide further evidence that metabolic conditions modulate aldolase B and FBPase-1 activity at the cellular level through the regulation of their interaction, suggesting that their association confers a catalytic advantage for both enzymes.


Assuntos
Metabolismo Energético , Frutose-Bifosfatase/metabolismo , Frutose-Bifosfato Aldolase/metabolismo , Gluconeogênese , Glicólise , Hepatócitos/metabolismo , Modelos Biológicos , Animais , Células Cultivadas , Transferência Ressonante de Energia de Fluorescência , Imunofluorescência , Frutose-Bifosfatase/química , Frutose-Bifosfatase/genética , Frutose-Bifosfato Aldolase/química , Frutose-Bifosfato Aldolase/genética , Células HeLa , Hepatócitos/citologia , Hepatócitos/enzimologia , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Masculino , Microscopia Confocal , Transporte Proteico , Ratos Wistar , Proteínas Recombinantes de Fusão/metabolismo
7.
J Cell Biochem ; 114(7): 1653-64, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23386391

RESUMO

Glycogen is the main source of glucose for many biological events. However, this molecule may have other functions, including those that have deleterious effects on cells. The rate-limiting enzyme in glycogen synthesis is glycogen synthase (GS). It is encoded by two genes, GYS1, expressed in muscle (muscle glycogen synthase, MGS) and other tissues, and GYS2, primarily expressed in liver (liver glycogen synthase, LGS). Expression of GS and its activity have been widely studied in many tissues. To date, it is not clear which GS isoform is responsible for glycogen synthesis and the role of glycogen in testis. Using RT-PCR, Western blot and immunofluorescence, we have detected expression of MGS but not LGS in mice testis during development. We have also evaluated GS activity and glycogen storage at different days after birth and we show that both GS activity and levels of glycogen are higher during the first days of development. Using RT-PCR, we have also shown that malin and laforin are expressed in testis, key enzymes for regulation of GS activity. These proteins form an active complex that regulates MGS by poly-ubiquitination in both Sertoli cell and male germ cell lines. In addition, PTG overexpression in male germ cell line triggered apoptosis by caspase3 activation, proposing a proapoptotic role of glycogen in testis. These findings suggest that GS activity and glycogen synthesis in testis could be regulated and a disruption of this process may be responsible for the apoptosis and degeneration of seminiferous tubules and possible cause of infertility.


Assuntos
Células Germinativas/citologia , Células Germinativas/metabolismo , Glicogênio Sintase/metabolismo , Glicogênio/metabolismo , Isoformas de Proteínas/metabolismo , Testículo/citologia , Testículo/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Glicogênio Sintase/genética , Immunoblotting , Masculino , Camundongos , Camundongos Transgênicos , Isoformas de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Túbulos Seminíferos/citologia , Túbulos Seminíferos/metabolismo , Testículo/enzimologia
8.
J Biol Chem ; 285(48): 37170-7, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20841354

RESUMO

In this study, we tested the efficacy of increasing liver glycogen synthase to improve blood glucose homeostasis. The overexpression of wild-type liver glycogen synthase in rats had no effect on blood glucose homeostasis in either the fed or the fasted state. In contrast, the expression of a constitutively active mutant form of the enzyme caused a significant lowering of blood glucose in the former but not the latter state. Moreover, it markedly enhanced the clearance of blood glucose when fasted rats were challenged with a glucose load. Hepatic glycogen stores in rats overexpressing the activated mutant form of liver glycogen synthase were enhanced in the fed state and in response to an oral glucose load but showed a net decline during fasting. In order to test whether these effects were maintained during long term activation of liver glycogen synthase, we generated liver-specific transgenic mice expressing the constitutively active LGS form. These mice also showed an enhanced capacity to store glycogen in the fed state and an improved glucose tolerance when challenged with a glucose load. Thus, we conclude that the activation of liver glycogen synthase improves glucose tolerance in the fed state without compromising glycogenolysis in the postabsorptive state. On the basis of these findings, we propose that the activation of liver glycogen synthase may provide a potential strategy for improvement of glucose tolerance in the postprandial state.


Assuntos
Glicemia , Expressão Gênica , Glicogênio Sintase/genética , Glicogênio Sintase/metabolismo , Glicogênio Hepático/metabolismo , Fígado/metabolismo , Animais , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ratos , Ratos Transgênicos , Ratos Wistar
9.
Nat Neurosci ; 10(11): 1407-13, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17952067

RESUMO

Glycogen synthesis is normally absent in neurons. However, inclusion bodies resembling abnormal glycogen accumulate in several neurological diseases, particularly in progressive myoclonus epilepsy or Lafora disease. We show here that mouse neurons have the enzymatic machinery for synthesizing glycogen, but that it is suppressed by retention of muscle glycogen synthase (MGS) in the phosphorylated, inactive state. This suppression was further ensured by a complex of laforin and malin, which are the two proteins whose mutations cause Lafora disease. The laforin-malin complex caused proteasome-dependent degradation both of the adaptor protein targeting to glycogen, PTG, which brings protein phosphatase 1 to MGS for activation, and of MGS itself. Enforced expression of PTG led to glycogen deposition in neurons and caused apoptosis. Therefore, the malin-laforin complex ensures a blockade of neuronal glycogen synthesis even under intense glycogenic conditions. Here we explain the formation of polyglucosan inclusions in Lafora disease by demonstrating a crucial role for laforin and malin in glycogen synthesis.


Assuntos
Apoptose/fisiologia , Regulação da Expressão Gênica/fisiologia , Glicogênio/metabolismo , Neurônios/metabolismo , Animais , Astrócitos/fisiologia , Proteínas de Transporte/farmacologia , Células Cultivadas , Córtex Cerebral/citologia , Embrião de Mamíferos , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Camundongos , Mutação/fisiologia , Proteínas Tirosina Fosfatases não Receptoras/farmacologia , Interferência de RNA/fisiologia , Transfecção , Tubulina (Proteína)/metabolismo , Ubiquitina-Proteína Ligases
10.
Hum Mol Genet ; 17(5): 667-78, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18029386

RESUMO

Lafora progressive myoclonus epilepsy (LD) is a fatal autosomal recessive neurodegenerative disorder characterized by the presence of glycogen-like intracellular inclusions called Lafora bodies. LD is caused by mutations in two genes, EPM2A and EPM2B, encoding respectively laforin, a dual-specificity protein phosphatase, and malin, an E3 ubiquitin ligase. Previously, we and others have suggested that the interactions between laforin and PTG (a regulatory subunit of type 1 protein phosphatase) and between laforin and malin are critical in the pathogenesis of LD. Here, we show that the laforin-malin complex downregulates PTG-induced glycogen synthesis in FTO2B hepatoma cells through a mechanism involving ubiquitination and degradation of PTG. Furthermore, we demonstrate that the interaction between laforin and malin is a regulated process that is modulated by the AMP-activated protein kinase (AMPK). These findings provide further insights into the critical role of the laforin-malin complex in the control of glycogen metabolism and unravel a novel link between the energy sensor AMPK and glycogen metabolism. These data advance our understanding of the functional role of laforin and malin, which hopefully will facilitate the development of appropriate LD therapies.


Assuntos
Proteínas de Transporte/genética , Glicogênio/biossíntese , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Quinases Ativadas por AMP , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Fenômenos Fisiológicos Celulares , Escherichia coli/genética , Glicogênio/análise , Proteínas de Fluorescência Verde/metabolismo , Humanos , Rim/citologia , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases não Receptoras/química , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Estatística como Assunto , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina-Proteína Ligases , Ubiquitinação
11.
Nat Cell Biol ; 19(2): 94-105, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28092655

RESUMO

The cytoplasmic polyadenylation element-binding (CPEB) proteins regulate pre-mRNA processing and translation of CPE-containing mRNAs in early embryonic development and synaptic activity. However, specific functions in adult organisms are poorly understood. Here we show that CPEB4 is required for adaptation to high-fat-diet- and ageing-induced endoplasmic reticulum (ER) stress, and subsequent hepatosteatosis. Stress-activated liver CPEB4 expression is dual-mode regulated. First, Cpeb4 mRNA transcription is controlled by the circadian clock, and then its translation is regulated by the unfolded protein response (UPR) through upstream open reading frames within the 5'UTR. Thus, the CPEB4 protein is synthesized only following ER stress but the induction amplitude is circadian. In turn, CPEB4 activates a second wave of UPR translation required to maintain ER and mitochondrial homeostasis. Our results suggest that combined transcriptional and translational Cpeb4 regulation generates a 'circadian mediator', which coordinates hepatic UPR activity with periods of high ER-protein-folding demand. Accordingly, CPEB4 deficiency results in non-alcoholic fatty liver disease.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Retículo Endoplasmático/metabolismo , Fígado Gorduroso/genética , Regulação da Expressão Gênica/genética , Proteínas de Ligação a RNA/genética , Animais , Estresse do Retículo Endoplasmático/genética , Homeostase/fisiologia , Camundongos , Biossíntese de Proteínas , Precursores de RNA/metabolismo , Transcrição Gênica/genética , Resposta a Proteínas não Dobradas/fisiologia
12.
Cell Metab ; 26(1): 256-266.e4, 2017 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-28683291

RESUMO

Glycogenin is considered essential for glycogen synthesis, as it acts as a primer for the initiation of the polysaccharide chain. Against expectations, glycogenin-deficient mice (Gyg KO) accumulate high amounts of glycogen in striated muscle. Furthermore, this glycogen contains no covalently bound protein, thereby demonstrating that a protein primer is not strictly necessary for the synthesis of the polysaccharide in vivo. Strikingly, in spite of the higher glycogen content, Gyg KO mice showed lower resting energy expenditure and less resistance than control animals when subjected to endurance exercise. These observations can be attributed to a switch of oxidative myofibers toward glycolytic metabolism. Mice overexpressing glycogen synthase in the muscle showed similar alterations, thus indicating that this switch is caused by the excess of glycogen. These results may explain the muscular defects of GSD XV patients, who lack glycogenin-1 and show high glycogen accumulation in muscle.


Assuntos
Glucosiltransferases/metabolismo , Glicogênio/metabolismo , Glicoproteínas/metabolismo , Músculo Esquelético/fisiologia , Animais , Metabolismo Energético , Glucosiltransferases/genética , Glicogênio Sintase/metabolismo , Glicoproteínas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/metabolismo , Consumo de Oxigênio
13.
Nat Commun ; 7: 11199, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27045898

RESUMO

The mechanisms that allow breast cancer (BCa) cells to metabolically sustain rapid growth are poorly understood. Here we report that BCa cells are dependent on a mechanism to supply precursors for intracellular lipid production derived from extracellular sources and that the endothelial lipase (LIPG) fulfils this function. LIPG expression allows the import of lipid precursors, thereby contributing to BCa proliferation. LIPG stands out as an essential component of the lipid metabolic adaptations that BCa cells, and not normal tissue, must undergo to support high proliferation rates. LIPG is ubiquitously and highly expressed under the control of FoxA1 or FoxA2 in all BCa subtypes. The downregulation of either LIPG or FoxA in transformed cells results in decreased proliferation and impaired synthesis of intracellular lipids.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Lipase/metabolismo , Metabolismo dos Lipídeos/genética , 4-Butirolactona/análogos & derivados , 4-Butirolactona/farmacologia , Animais , Transporte Biológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Doxiciclina/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Fator 3-alfa Nuclear de Hepatócito/antagonistas & inibidores , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/antagonistas & inibidores , Fator 3-beta Nuclear de Hepatócito/genética , Humanos , Lactonas/farmacologia , Lipase/antagonistas & inibidores , Lipase/genética , Células MCF-7 , Camundongos , Camundongos Nus , Invasividade Neoplásica , Orlistate , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Biochem J ; 378(Pt 3): 1073-7, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-14651477

RESUMO

Pharmacological inhibition of liver GP (glycogen phosphorylase), which is currently being studied as a treatment for Type II (non-insulin-dependent) diabetes, may affect muscle glycogen metabolism. In the present study, we analysed the effects of the GP inhibitor CP-91149 on non-engineered or GP-overexpressing cultured human muscle cells. We found that CP-91149 treatment decreased muscle GP activity by (1) converting the phosphorylated AMP-independent a form into the dephosphorylated AMP-dependent b form and (2) inhibiting GP a activity and AMP-mediated GP b activation. Dephosphorylation of GP was exerted, irrespective of incubation of the cells with glucose, whereas inhibition of its activity was synergic with glucose. As expected, CP-91149 impaired the glycogenolysis induced by glucose deprivation. CP-91149 also promoted the dephosphorylation and activation of GS (glycogen synthase) in non-engineered or GP-overexpressing cultured human muscle cells, but exclusively in glucose-deprived cells. However, this inhibitor did not activate GS in glucose-deprived but glycogen-replete cells overexpressing PTG (protein targeting to glycogen), thus suggesting that glycogen inhibits the CP-91149-mediated activation of GS. Consistently, CP-91149 promoted glycogen resynthesis, but not its overaccumulation. Hence, treatment with CP-91149 impairs muscle glycogen breakdown, but enhances its recovery, which may be useful for the treatment of Type II (insulin-dependent) diabetes.


Assuntos
Amidas/farmacologia , Inibidores Enzimáticos/farmacologia , Glicogênio Fosforilase/antagonistas & inibidores , Glicogênio/metabolismo , Indóis/farmacologia , Fibras Musculares Esqueléticas/metabolismo , Células Cultivadas , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/metabolismo , Humanos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/enzimologia
15.
FEBS Lett ; 577(1-2): 154-8, 2004 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-15527777

RESUMO

In primary cultured hepatocytes, fructose-1,6-bisphosphatase (FBPase) localization is modulated by glucose, dihydroxyacetone (DHA) and insulin. In the absence of these substrates, FBPase was present in the cytoplasm, but the addition of glucose or DHA induced its translocation to the nucleus. As expected, we observed the opposite effect of glucose on glucokinase localization. The addition of insulin in the absence of glucose largely increased the amount of nuclear FBPase. Moreover, at high concentrations of glucose or DHA, FBPase shifted from the cytosol to the cell periphery and co-localized with GS. Interestingly, the synthesis of Glu-6-P and glycogen induced by DHA was not inhibited by insulin. These results indicate that FBPase is involved in glycogen synthesis from gluconeogenic precursors. Overall, these findings show that translocation may be a new integrative mechanism for gluconeogenesis and glyconeogenesis.


Assuntos
Frutose-Bifosfatase/metabolismo , Hepatócitos/enzimologia , Frações Subcelulares/enzimologia , Animais , Di-Hidroxiacetona/fisiologia , Imunofluorescência , Glucose/fisiologia , Insulina/fisiologia , Masculino , Ratos
16.
FEBS Lett ; 546(1): 127-32, 2003 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-12829248

RESUMO

Traditionally, glycogen synthase (GS) has been considered to catalyze the key step of glycogen synthesis and to exercise most of the control over this metabolic pathway. However, recent advances have shown that other factors must be considered. Moreover, the control of glycogen deposition does not follow identical mechanisms in muscle and liver. Glucose must be phosphorylated to promote activation of GS. Glucose-6-phosphate (Glc-6-P) binds to GS, causing the allosteric activation of the enzyme probably through a conformational rearrangement that simultaneously converts it into a better substrate for protein phosphatases, which can then lead to the covalent activation of GS. The potency of Glc-6-P for activation of liver GS is determined by its source, since Glc-6-P arising from the catalytic action of glucokinase (GK) is much more effective in mediating the activation of the enzyme than the same metabolite produced by hexokinase I (HK I). As a result, hepatic glycogen deposition from glucose is subject to a system of control in which the 'controller', GS, is in turn controlled by GK. In contrast, in skeletal muscle, the control of glycogen synthesis is shared between glucose transport and GS. The characteristics of the two pairs of isoenzymes, liver GS/GK and muscle GS/HK I, and the relationships that they establish are tailored to suit specific metabolic roles of the tissues in which they are expressed. The key enzymes in glycogen metabolism change their intracellular localization in response to glucose. The changes in the intracellular distribution of liver GS and GK triggered by glucose correlate with stimulation of glycogen synthesis. The translocation of GS, which constitutes an additional mechanism of control, causes the orderly deposition of hepatic glycogen and probably represents a functional advantage in the metabolism of the polysaccharide.


Assuntos
Glicogênio Hepático/metabolismo , Animais , Células Cultivadas , Ativação Enzimática , Glucoquinase/metabolismo , Glucose/metabolismo , Glucose-6-Fosfato/metabolismo , Glicogênio Sintase/química , Glicogênio Sintase/metabolismo , Hepatócitos/enzimologia , Hexoquinase/metabolismo , Humanos , Isoenzimas/metabolismo , Modelos Biológicos , Músculo Esquelético/enzimologia
17.
Acta Diabetol ; 51(4): 543-52, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24458975

RESUMO

Liver and muscle glycogen content is reduced in diabetic patients but there is no information on the effect of diabetes on the glycogen content in the retinal pigment epithelium (RPE). The main aim of the study was to compare the glycogen content in the RPE between diabetic and non-diabetic human donors. Glycogen synthase (GS) and glycogen phosphorylase (GP), the key enzymes of glycogen metabolism, as well as their isoforms, were also assessed. For this purpose, 44 human postmortem eye cups were included (22 from 11 type 2 diabetic and 22 from 11 non-diabetic donors matched by age). Human RPE cells cultured in normoglycemic and hyperglycemic conditions were also analyzed. Glycogen content as well as the mRNA, protein content and enzyme activity of GS and GP were determined. In addition, GS and GP isoforms were characterized. In the RPE from diabetic donors, as well as in RPE cells grown in hyperglycemic conditions, the glycogen content was increased. The increase in glycogen content was associated with an increase in GS without changes in GP levels. In RPE form human donors, the muscle GS isoform but not the liver GS isoform was detected. Regarding GP, the muscle and brain isoform of GP but not the liver GP isoform were detected. We conclude that glycogen storage is increased in the RPE of diabetic patients, and it is associated with an increase in GS activity. Further studies aimed at determining the role of glycogen deposits in the pathogenesis of diabetic retinopathy are warranted.


Assuntos
Diabetes Mellitus/metabolismo , Glicogênio/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Diabetes Mellitus/enzimologia , Diabetes Mellitus/genética , Feminino , Glicogênio Fosforilase/genética , Glicogênio Fosforilase/metabolismo , Glicogênio Sintase/genética , Glicogênio Sintase/metabolismo , Humanos , Fígado/enzimologia , Fígado/metabolismo , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/enzimologia , Músculo Esquelético/metabolismo , Epitélio Pigmentado da Retina/enzimologia , Doadores de Tecidos
18.
Diabetes ; 62(12): 4070-82, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23990365

RESUMO

The liver responds to an increase in blood glucose levels in the postprandial state by uptake of glucose and conversion to glycogen. Liver glycogen synthase (GYS2), a key enzyme in glycogen synthesis, is controlled by a complex interplay between the allosteric activator glucose-6-phosphate (G6P) and reversible phosphorylation through glycogen synthase kinase-3 and the glycogen-associated form of protein phosphatase 1. Here, we initially performed mutagenesis analysis and identified a key residue (Arg(582)) required for activation of GYS2 by G6P. We then used GYS2 Arg(582)Ala knockin (+/R582A) mice in which G6P-mediated GYS2 activation had been profoundly impaired (60-70%), while sparing regulation through reversible phosphorylation. R582A mutant-expressing hepatocytes showed significantly reduced glycogen synthesis with glucose and insulin or glucokinase activator, which resulted in channeling glucose/G6P toward glycolysis and lipid synthesis. GYS2(+/R582A) mice were modestly glucose intolerant and displayed significantly reduced glycogen accumulation with feeding or glucose load in vivo. These data show that G6P-mediated activation of GYS2 plays a key role in controlling glycogen synthesis and hepatic glucose-G6P flux control and thus whole-body glucose homeostasis.


Assuntos
Glucose-6-Fosfato/metabolismo , Glicogênio Sintase/metabolismo , Hepatócitos/metabolismo , Glicogênio Hepático/biossíntese , Fígado/metabolismo , Animais , Glicemia/metabolismo , Glucose/farmacologia , Glicogênio Sintase/genética , Hepatócitos/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Insulina/farmacologia , Fígado/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Fosforilação
19.
EMBO Mol Med ; 4(8): 719-29, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22549942

RESUMO

Under physiological conditions, most neurons keep glycogen synthase (GS) in an inactive form and do not show detectable levels of glycogen. Nevertheless, aberrant glycogen accumulation in neurons is a hallmark of patients suffering from Lafora disease or other polyglucosan disorders. Although these diseases are associated with mutations in genes involved in glycogen metabolism, the role of glycogen accumulation remains elusive. Here, we generated mouse and fly models expressing an active form of GS to force neuronal accumulation of glycogen. We present evidence that the progressive accumulation of glycogen in mouse and Drosophila neurons leads to neuronal loss, locomotion defects and reduced lifespan. Our results highlight glycogen accumulation in neurons as a direct cause of neurodegeneration.


Assuntos
Doença de Depósito de Glicogênio/genética , Glicogênio Sintase/metabolismo , Glicogênio/metabolismo , Doenças Neurodegenerativas/etiologia , Neurônios/enzimologia , Neurônios/patologia , Animais , Modelos Animais de Doenças , Drosophila , Doença de Depósito de Glicogênio/patologia , Doença de Depósito de Glicogênio/fisiopatologia , Glicogênio Sintase/genética , Locomoção , Longevidade , Camundongos , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/fisiopatologia
20.
EMBO Mol Med ; 3(11): 667-81, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21882344

RESUMO

Lafora disease (LD) is caused by mutations in either the laforin or malin gene. The hallmark of the disease is the accumulation of polyglucosan inclusions called Lafora Bodies (LBs). Malin knockout (KO) mice present polyglucosan accumulations in several brain areas, as do patients of LD. These structures are abundant in the cerebellum and hippocampus. Here, we report a large increase in glycogen synthase (GS) in these mice, in which the enzyme accumulates in LBs. Our study focused on the hippocampus where, under physiological conditions, astrocytes and parvalbumin-positive (PV(+)) interneurons expressed GS and malin. Although LBs have been described only in neurons, we found this polyglucosan accumulation in the astrocytes of the KO mice. They also had LBs in the soma and some processes of PV(+) interneurons. This phenomenon was accompanied by the progressive loss of these neuronal cells and, importantly, neurophysiological alterations potentially related to impairment of hippocampal function. Our results emphasize the relevance of the laforin-malin complex in the control of glycogen metabolism and highlight altered glycogen accumulation as a key contributor to neurodegeneration in LD.


Assuntos
Modelos Animais de Doenças , Glicogênio Sintase/metabolismo , Doença de Lafora/enzimologia , Doença de Lafora/fisiopatologia , Camundongos , Degeneração Neural/enzimologia , Animais , Astrócitos/enzimologia , Feminino , Glicogênio/metabolismo , Glicogênio Sintase/genética , Hipocampo/enzimologia , Humanos , Corpos de Inclusão/enzimologia , Corpos de Inclusão/genética , Doença de Lafora/genética , Doença de Lafora/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Degeneração Neural/genética , Degeneração Neural/patologia , Degeneração Neural/fisiopatologia , Neurônios/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA