Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Rev Endocr Metab Disord ; 25(4): 707-725, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38556561

RESUMO

Hepatic glycogen storage diseases constitute a group of disorders due to defects in the enzymes and transporters involved in glycogen breakdown and synthesis in the liver. Although hypoglycemia and hepatomegaly are the primary manifestations of (most of) hepatic GSDs, involvement of the endocrine system has been reported at multiple levels in individuals with hepatic GSDs. While some endocrine abnormalities (e.g., hypothalamic­pituitary axis dysfunction in GSD I) can be direct consequence of the genetic defect itself, others (e.g., osteopenia in GSD Ib, insulin-resistance in GSD I and GSD III) may be triggered by the (dietary/medical) treatment. Being aware of the endocrine abnormalities occurring in hepatic GSDs is essential (1) to provide optimized medical care to this group of individuals and (2) to drive research aiming at understanding the disease pathophysiology. In this review, a thorough description of the endocrine manifestations in individuals with hepatic GSDs is presented, including pathophysiological and clinical implications.


Assuntos
Doença de Depósito de Glicogênio , Humanos , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/fisiopatologia , Hepatopatias/metabolismo , Hepatopatias/fisiopatologia , Hepatopatias/etiologia , Doenças do Sistema Endócrino/metabolismo , Doenças do Sistema Endócrino/fisiopatologia , Fígado/metabolismo , Fígado/fisiopatologia
2.
Mol Genet Metab ; 133(3): 269-276, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34083142

RESUMO

INTRODUCTION: Liver Glycogen Storage Disease IX is a rare metabolic disorder of glycogen metabolism caused by deficiency of the phosphorylase kinase enzyme (PhK). Variants in the PHKG2 gene, encoding the liver-specific catalytic γ2 subunit of PhK, are associated with a liver GSD IX subtype known as PHKG2 GSD IX or GSD IX γ2. There is emerging evidence that patients with GSD IX γ2 can develop severe and progressive liver disease, yet research regarding the disease has been minimal to date. Here we characterize the first mouse model of liver GSD IX γ2. METHODS: A Phkg2-/- mouse model was generated via targeted removal of the Phkg2 gene. Knockout (Phkg2-/-, KO) and wild type (Phkg2+/+, WT) mice up to 3 months of age were compared for morphology, Phkg2 transcription, PhK enzyme activity, glycogen content, histology, serum liver markers, and urinary glucose tetrasaccharide Glcα1-6Glcα1-4Glcα1-4Glc (Glc4). RESULTS: When compared to WT controls, KO mice demonstrated significantly decreased liver PhK enzyme activity, increased liver: body weight ratio, and increased glycogen in the liver, with no glycogen accumulation observed in the brain, quadricep, kidney, and heart. KO mice demonstrated elevated liver blood markers as well as elevated urine Glc4, a commonly used biomarker for glycogen storage disease. KO mice demonstrated features of liver structural damage. Hematoxylin & Eosin and Masson's Trichrome stained KO mice liver histology slides revealed characteristic GSD hepatocyte architectural changes and early liver fibrosis, as have been reported in liver GSD patients. DISCUSSION: This study provides the first evidence of a mouse model that recapitulates the liver-specific pathology of patients with GSD IX γ2. The model will provide the first platform for further study of disease progression in GSD IX γ2 as well as for the evaluation of novel therapeutics.


Assuntos
Modelos Animais de Doenças , Doença de Depósito de Glicogênio/fisiopatologia , Glicogênio/metabolismo , Hepatopatias/fisiopatologia , Fígado/fisiopatologia , Camundongos , Fosforilase Quinase/genética , Animais , Feminino , Doença de Depósito de Glicogênio/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilase Quinase/deficiência
3.
J Inherit Metab Dis ; 44(3): 534-543, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33141444

RESUMO

Adult polyglucosan body disease (APBD) represents a complex autosomal recessive inherited neurometabolic disorder due to homozygous or compound heterozygous pathogenic variants in GBE1 gene, resulting in deficiency of glycogen-branching enzyme and secondary storage of glycogen in the form of polyglucosan bodies, involving the skeletal muscle, diaphragm, peripheral nerve (including autonomic fibers), brain white matter, spinal cord, nerve roots, cerebellum, brainstem and to a lesser extent heart, lung, kidney, and liver cells. The diversity of new clinical presentations regarding neuromuscular involvement is astonishing and transformed APBD in a key differential diagnosis of completely different clinical conditions, including axonal and demyelinating sensorimotor polyneuropathy, progressive spastic paraparesis, motor neuronopathy presentations, autonomic disturbances, leukodystrophies or even pure myopathic involvement with limb-girdle pattern of weakness. This review article aims to summarize the main clinical, biochemical, genetic, and diagnostic aspects regarding APBD with special focus on neuromuscular presentations.


Assuntos
Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/fisiopatologia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/fisiopatologia , Adulto , Encéfalo/patologia , Doença de Depósito de Glicogênio/patologia , Humanos , Músculo Esquelético/patologia , Doenças do Sistema Nervoso/patologia , Nervos Periféricos/patologia , Fenótipo , Medula Espinal/patologia
4.
Can J Neurol Sci ; 47(3): 400-403, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31987065

RESUMO

Glycogen storage diseases (GSDs) result from the deficiency of enzymes involved in glycogen synthesis and breakdown into glucose. Mutations in the gene PHKA2 encoding phosphorylase kinase regulatory subunit alpha 2 have been linked to GSD type IXa. We describe a family with two adult brothers with neonatal hepatosplenomegaly and later onset of hearing loss, cognitive impairment, and cerebellar involvement. Whole-exome sequencing was performed on both subjects and revealed a shared hemizygous missense variant (c.A1561G; p.T521A) in exon 15 of PHKA2. The phenotype broadens the clinical and magnetic resonance imaging spectrum of GSD type IXa to include later onset neurological manifestations.


Assuntos
Ataxia Cerebelar/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Epilepsia/fisiopatologia , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Doença de Depósito de Glicogênio/fisiopatologia , Perda Auditiva Neurossensorial/fisiopatologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Fosforilase Quinase/genética , Adulto , Encéfalo/diagnóstico por imagem , Incontinência Fecal/fisiopatologia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doença de Depósito de Glicogênio/genética , Hepatomegalia/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Irmãos , Esplenomegalia/fisiopatologia , Sequenciamento do Exoma
5.
Rev Gastroenterol Peru ; 40(1): 73-76, 2020.
Artigo em Espanhol | MEDLINE | ID: mdl-32369470

RESUMO

The glycogen storage diseases contain a range of diseases that are characterized by the abnormal storage or utilization of glycogen, the organs most affected being muscle and / or liver. Hepatomegaly may be a clinical sign that could guide to the diagnosis. We describe a 15-year-old patient with hepatomegaly, hypertransaminasemia and growth retardation. He was diagnosed with a glycogen storage disease by liver biopsy.


Assuntos
Doença de Depósito de Glicogênio/diagnóstico , Hepatopatias/diagnóstico , Adolescente , Doença de Depósito de Glicogênio/fisiopatologia , Hepatomegalia/diagnóstico , Hepatomegalia/etiologia , Humanos , Hepatopatias/fisiopatologia , Masculino
6.
Mol Genet Metab ; 125(1-2): 79-85, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30037503

RESUMO

INTRODUCTION: Hepatic glycogen storage diseases (GSDs) are a group of inherited disorders of carbohydrate metabolism for which dietary management is the cornerstone. Safety and acute complications associated with dietary management have been poorly documented. We hypothesized that safety issues and complications associated with dietary management are prevalent amongst patients with these ultra-rare disorders. METHODS: A questionnaire was developed consisting of 40 questions and was distributed via eight GSD patient organizations from multiple countries. Respondents were (caregivers of) patients with self-reported hepatic GSD. RESULTS: 249 GSD patients from 26 countries responded with a median age of 14.8 years (range: 0.5-66.1). Although management was considered safe by 71% of patients, 51% reported at least one acute complication associated with dietary management, with a total number of 425 reported complications. Most frequently reported causes were: not waking up by an alarm clock (n = 70), forgetting a meal (n = 57) and infections (n = 43). Most frequently reported complications were: hypoglycemia (n = 112), hospital admissions (n = 79) and drowsiness (n = 74). Most complications occurred before the age of 12 years (82%; 637/774 total number of reported events) and during night time (63%; 340/536). Only 61% (152/249) of the GSD patients reported using a written emergency protocol. CONCLUSIONS: Safety issues and complications associated with dietary management are prevalently reported by (caregivers of) 249 GSD patients. A discrepancy has been observed between the patient's perspective on safety of dietary management and occurrence of complications as a result of dietary management.


Assuntos
Dieta/efeitos adversos , Doença de Depósito de Glicogênio/dietoterapia , Doença de Depósito de Glicogênio/epidemiologia , Fígado/fisiopatologia , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Doença de Depósito de Glicogênio/fisiopatologia , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Inquéritos e Questionários , Adulto Jovem
7.
Mol Genet Metab ; 123(3): 326-330, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29223626

RESUMO

BACKGROUND: Glycemic control in hepatic glycogen storage diseases (GSDs) relies on specific nutritional recommendations, including strict avoidance of a fasting period. Uncooked cornstarch (UCCS) is an important therapeutic component. A new modified UCCS, Glycosade™, was created with the objective of prolonging euglycemia. We aimed to determine the length of euglycemia on Glycosade™ using a continuous glucose monitor (CGM) and to evaluate whether longer euglycemia and thus less nighttime interruptions would improve sleep and quality of life (QoL) after the introduction of the modified cornstarch. METHODS: We conducted a prospective cohort study to assess quality and quantity of sleep and quality of life (QoL) in patients with GSDs on standard UCCS and after the introduction of Glycosade™. Sleep and QoL evaluation was done for patients using validated questionnaires, a standardized sleep diary and actigraphy. Length of fast and glucose variability were determined with CGM. RESULTS: Nine adults with GSD Ia took part in the study. Glycosade™ introduction was done under close supervision during a hospital admission. Comparison of sleep in 9 patients showed sleep disturbances on standard UCCS that were improved with Glycosade™. QoL was normal both pre and post Glycosade™. The CGM confirmed maintenance of a longer fasting period with Glycosade™ at home. CONCLUSION: Glycosade™ represents an alternative option for GSD patients. We showed possible benefits in terms of sleep quality. We also confirmed the longer length of fast on Glycosade™. SYNOPSIS: A new modified form of uncooked starch for patients with glycogen storage disease represents an alternative option as it showed a longer length of fast and improvements in sleep quality.


Assuntos
Jejum/fisiologia , Doença de Depósito de Glicogênio/fisiopatologia , Hipoglicemia/dietoterapia , Qualidade de Vida , Sono/fisiologia , Amido , Actigrafia , Adulto , Glicemia/fisiologia , Feminino , Glucose/administração & dosagem , Doença de Depósito de Glicogênio/sangue , Humanos , Hipoglicemia/sangue , Hipoglicemia/tratamento farmacológico , Hipoglicemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Resultado do Tratamento , Adulto Jovem
8.
Hum Mol Genet ; 24(23): 6801-10, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26385640

RESUMO

Glycogen storage disease type IV (GSD IV) is a rare autosomal recessive disorder caused by deficiency of the glycogen-branching enzyme (GBE). The diagnostic hallmark of the disease is the accumulation of a poorly branched form of glycogen known as polyglucosan (PG). The disease is clinically heterogeneous, with variable tissue involvement and age at onset. Complete loss of enzyme activity is lethal in utero or in infancy and affects primarily the muscle and the liver. However, residual enzyme activity as low as 5-20% leads to juvenile or adult onset of a disorder that primarily affects the central and peripheral nervous system and muscles and in the latter is termed adult polyglucosan body disease (APBD). Here, we describe a mouse model of GSD IV that reflects this spectrum of disease. Homologous recombination was used to knock in the most common GBE1 mutation p.Y329S c.986A > C found in APBD patients of Ashkenazi Jewish decent. Mice homozygous for this allele (Gbe1(ys/ys)) exhibit a phenotype similar to APBD, with widespread accumulation of PG. Adult mice exhibit progressive neuromuscular dysfunction and die prematurely. While the onset of symptoms is limited to adult mice, PG accumulates in tissues of newborn mice but is initially absent from the cerebral cortex and heart muscle. Thus, PG is well tolerated in most tissues, but the eventual accumulation in neurons and their axons causes neuropathy that leads to hind limb spasticity and premature death. This mouse model mimics the pathology and pathophysiologic features of human adult-onset branching enzyme deficiency.


Assuntos
Modelos Animais de Doenças , Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio Tipo IV/metabolismo , Mutação , Animais , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Técnicas de Introdução de Genes , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/fisiopatologia , Doença de Depósito de Glicogênio Tipo IV/genética , Doença de Depósito de Glicogênio Tipo IV/fisiopatologia , Camundongos , Músculo Estriado/metabolismo , Músculo Estriado/fisiopatologia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/fisiopatologia , Sistema Nervoso Periférico/metabolismo , Sistema Nervoso Periférico/fisiopatologia , Fenótipo
9.
Mol Genet Metab ; 122(3): 117-121, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28882528

RESUMO

OBJECTIVE: We investigated metabolism and physiological responses to exercise in an 18-year-old woman with multiple congenital abnormalities and exertional muscle fatigue, tightness, and rhabdomyolysis. METHODS: We studied biochemistry in muscle and fibroblasts, performed mutation analysis, assessed physiological responses to forearm and cycle-ergometer exercise combined with stable-isotope techniques and indirect calorimetry, and evaluated the effect of IV glucose infusion and oral sucrose ingestion on the exercise response. RESULTS: Phosphoglucomutase type 1 (PGM1) activity in muscle and fibroblasts was severely deficient and PGM1 in muscle was undetectable by Western blot. The patient was compound heterozygous for missense (R422W) and nonsense (Q530X) mutations in PGM1. Forearm exercise elicited no increase in lactate, but an exaggerated increase in ammonia, and provoked a forearm contracture. Comparable to patients with McArdle disease, the patient developed a 'second wind' with a spontaneous fall in exercise heart rate and perceived exertion. Like in McArdle disease, this was attributable to an increase in muscle oxidative capacity. Carbohydrate oxidation was blocked during exercise, and the patient had exaggerated oxidation of fat to fuel exercise. Exercise heart rate and perceived exertion were lower after IV glucose and oral sucrose. Muscle glycogen level was low normal. CONCLUSIONS: The second wind phenomenon has been considered to be pathognomonic for McArdle disease, but we demonstrate that it can also be present in PGM1 deficiency. We show that severe loss of PGM1 activity causes blocked muscle glycogenolysis that mimics McArdle disease, but may also limit glycogen synthesis, which broadens the phenotypic spectrum of this disorder.


Assuntos
Exercício Físico/fisiologia , Doença de Depósito de Glicogênio/fisiopatologia , Glicogênio/metabolismo , Doenças Musculares/fisiopatologia , Adolescente , Biópsia , Feminino , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio Tipo V/fisiopatologia , Glicogenólise , Frequência Cardíaca , Humanos , Lactatos/metabolismo , Masculino , Fadiga Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Oxirredução , Consumo de Oxigênio , Esforço Físico , Rabdomiólise , Pele/patologia
10.
Genet Med ; 18(10): 1037-43, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26913919

RESUMO

PURPOSE: Glycogen storage disease (GSD) is an umbrella term for a group of genetic disorders that involve the abnormal metabolism of glycogen; to date, 23 types of GSD have been identified. The nonspecific clinical presentation of GSD and the lack of specific biomarkers mean that Sanger sequencing is now widely relied on for making a diagnosis. However, this gene-by-gene sequencing technique is both laborious and costly, which is a consequence of the number of genes to be sequenced and the large size of some genes. METHODS: This work reports the use of massive parallel sequencing to diagnose patients at our laboratory in Spain using either a customized gene panel (targeted exome sequencing) or the Illumina Clinical-Exome TruSight One Gene Panel (clinical exome sequencing (CES)). Sequence variants were matched against biochemical and clinical hallmarks. RESULTS: Pathogenic mutations were detected in 23 patients. Twenty-two mutations were recognized (mostly loss-of-function mutations), including 11 that were novel in GSD-associated genes. In addition, CES detected five patients with mutations in ALDOB, LIPA, NKX2-5, CPT2, or ANO5. Although these genes are not involved in GSD, they are associated with overlapping phenotypic characteristics such as hepatic, muscular, and cardiac dysfunction. CONCLUSIONS: These results show that next-generation sequencing, in combination with the detection of biochemical and clinical hallmarks, provides an accurate, high-throughput means of making genetic diagnoses of GSD and related diseases.Genet Med 18 10, 1037-1043.


Assuntos
Doença de Depósito de Glicogênio/diagnóstico , Doença de Depósito de Glicogênio/genética , Glicogênio/genética , Patologia Molecular , Adolescente , Adulto , Anoctaminas , Criança , Pré-Escolar , Canais de Cloreto/genética , Exoma/genética , Feminino , Frutose-Bifosfato Aldolase/genética , Glicogênio/metabolismo , Doença de Depósito de Glicogênio/fisiopatologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Masculino , Mutação , Proteínas Nucleares/genética , Esterol Esterase/genética , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética , Adulto Jovem
11.
Muscle Nerve ; 53(6): 976-81, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26789422

RESUMO

INTRODUCTION: Adult polyglucosan body disease (APBD) usually presents with progressive spastic paraparesis, neurogenic bladder, and distal lower limb sensory abnormalities. It is caused by mutations in the glycogen branching enzyme gene (GBE1). METHODS: We describe a woman with an unusual phenotype manifesting as progressive left brachial more than lumbosacral plexopathies, with central sensory and corticospinal tract involvement. RESULTS: Magnetic resonance imaging of the brain and cervical spine showed abnormal T2 signal within the ventral pons and medulla bilaterally, involving the pyramidal tracts and the medial leminisci. There was also medullary and cervical spine atrophy. On nerve biopsy, large polyglucosan bodies were noted in the endoneurium. The patient was found to be compound heterozygous for 2 novel mutations in GBE1. Peripheral blood leukocyte GBE activity was markedly reduced to 7% of normal, confirming the diagnosis of APBD. CONCLUSIONS: In this report we describe a new phenotype of APBD associated with 2 novel mutations. Muscle Nerve 53: 976-981, 2016.


Assuntos
Progressão da Doença , Lateralidade Funcional/fisiologia , Doença de Depósito de Glicogênio/fisiopatologia , Doenças do Sistema Nervoso/fisiopatologia , Condução Nervosa/fisiologia , Análise Mutacional de DNA , Feminino , Sistema da Enzima Desramificadora do Glicogênio/genética , Doença de Depósito de Glicogênio/diagnóstico por imagem , Doença de Depósito de Glicogênio/genética , Humanos , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Doenças do Sistema Nervoso/diagnóstico por imagem , Doenças do Sistema Nervoso/genética , RNA Mensageiro/metabolismo , Tempo de Reação/fisiologia , Nervo Sural/patologia
12.
Circ Res ; 114(6): 966-75, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24503893

RESUMO

RATIONALE: AMP-activated protein kinase is a master regulator of cell metabolism and an attractive drug target for cancer and metabolic and cardiovascular diseases. Point mutations in the regulatory γ2-subunit of AMP-activated protein kinase (encoded by Prkag2 gene) caused a unique form of human cardiomyopathy characterized by cardiac hypertrophy, ventricular preexcitation, and glycogen storage. Understanding the disease mechanisms of Prkag2 cardiomyopathy is not only beneficial for the patients but also critical to the use of AMP-activated protein kinase as a drug target. OBJECTIVE: We sought to identify the pro-growth-signaling pathway(s) triggered by Prkag2 mutation and to distinguish it from the secondary response to glycogen storage. METHODS AND RESULTS: In a mouse model of N488I mutation of the Prkag2 gene (R2M), we rescued the glycogen storage phenotype by genetic inhibition of glucose-6-phosphate-stimulated glycogen synthase activity. Ablation of glycogen storage eliminated the ventricular preexcitation but did not affect the excessive cardiac growth in R2M mice. The progrowth effect in R2M hearts was mediated via increased insulin sensitivity and hyperactivity of Akt, resulting in activation of mammalian target of rapamycin and inactivation of forkhead box O transcription factor-signaling pathways. Consequently, cardiac myocyte proliferation during the postnatal period was enhanced in R2M hearts followed by hypertrophic growth in adult hearts. Inhibition of mammalian target of rapamycin activity by rapamycin or restoration of forkhead box O transcription factor activity by overexpressing forkhead box O transcription factor 1 rescued the abnormal cardiac growth. CONCLUSIONS: Our study reveals a novel mechanism for Prkag2 cardiomyopathy, independent of glycogen storage. The role of γ2-AMP-activated protein kinase in cell growth also has broad implications in cardiac development, growth, and regeneration.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Cardiomiopatia Hipertrófica Familiar/genética , Doença de Depósito de Glicogênio/genética , Glicogênio/biossíntese , Miocárdio/metabolismo , Miócitos Cardíacos/patologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Cardiomiopatia Hipertrófica Familiar/enzimologia , Cardiomiopatia Hipertrófica Familiar/metabolismo , Cardiomiopatia Hipertrófica Familiar/fisiopatologia , Divisão Celular , Crescimento Celular , Modelos Animais de Doenças , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Técnicas de Introdução de Genes , Teste de Complementação Genética , Glucose-6-Fosfato/metabolismo , Glucose-6-Fosfato/farmacologia , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/fisiopatologia , Glicogênio Sintase/genética , Glicogênio Sintase/fisiologia , Resistência à Insulina/genética , Camundongos , Miócitos Cardíacos/metabolismo , Síndromes de Pré-Excitação/genética , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/fisiologia
13.
Clin Exp Pharmacol Physiol ; 42(4): 415-25, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25676548

RESUMO

Cardiac glycogen regulation involves a complex interplay between multiple signalling pathways, allosteric activation of enzymes, and sequestration for autophagic degradation. Signalling pathways appear to converge on glycogen regulatory enzymes via insulin (glycogen synthase kinase 3ß, protein phosphatase 1, allosteric action of glucose-6-phosphate), ß-adrenergic (phosphorylase kinase protein phosphatase 1 inhibitor), and 5' adenosine monophosphate-activated protein kinase (allosteric action of glucose-6-phosphate, direct glycogen binding, insulin receptor). While cytosolic glycogen synthesis and breakdown are relatively well understood, recent findings relating to phagic glycogen degradation highlight a new area of investigation in the heart. It has been recently demonstrated that a specific glycophagy pathway is operational in the myocardium. Proteins involved in recruiting glycogen to the forming phagosome have been identified. Starch-binding domain-containing protein 1 is involved in binding glycogen and mediating membrane anchorage via interaction with a homologue of the phagosomal protein light-chain 3. Specifically, it has been shown that starch-binding domain-containing protein 1 and light-chain 3 have discrete phagosomal immunolocalization patterns in cardiomyocytes, indicating that autophagic trafficking of glycogen and protein cargo in cardiomyocytes can occur via distinct pathways. There is strong evidence from glycogen storage diseases that phagic/lysosomal glycogen breakdown is important for maintaining normal cardiac glycogen levels and does not simply constitute a redundant 'alternative' breakdown route for glycogen. Advancing understanding of glycogen handling in the heart is an important priority with relevance not only to genetic glycogen storage diseases but also to cardiac metabolic stress disorders such as diabetes and ischaemia.


Assuntos
Doença de Depósito de Glicogênio/metabolismo , Glicogênio/metabolismo , Cardiopatias/metabolismo , Miocárdio/metabolismo , Animais , Metabolismo Energético , Doença de Depósito de Glicogênio/patologia , Doença de Depósito de Glicogênio/fisiopatologia , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Cinética , Lisossomos/metabolismo , Miocárdio/patologia , Fagossomos/metabolismo , Transdução de Sinais
14.
Curr Opin Clin Nutr Metab Care ; 17(4): 329-37, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24848530

RESUMO

PURPOSE OF REVIEW: Autophagy is an evolutionarily conserved cellular programme for the turnover of organelles, proteins, and other macromolecules, involving the lysosomal degradation pathway. Emerging evidence suggests that autophagy can play a central role in human metabolism as well as impact diverse cellular processes including organelle homeostasis, cell death and proliferation, lipid and glycogen metabolism, and the regulation of inflammation and immune responses. The purpose of this review is to examine recent evidence for the role of autophagy in cellular metabolism, and its relevance to select human diseases that involve disorders of metabolism. RECENT FINDINGS: Recent studies suggest that autophagy may play multiple roles in metabolic diseases, including diabetes and its complications, metabolic syndrome and obesity, myopathies and other inborn errors of metabolism, as well as other diseases that may involve altered mitochondrial function. SUMMARY: Strategies aimed at modulating autophagy may lead to therapies for diseases in which altered cellular and tissue metabolism play a key role.


Assuntos
Autofagia , Inflamação/fisiopatologia , Síndrome Metabólica/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Doença de Depósito de Glicogênio/fisiopatologia , Homeostase , Humanos , Mitocôndrias/metabolismo , Doenças Musculares/fisiopatologia , Obesidade/fisiopatologia
15.
IUBMB Life ; 65(8): 657-64, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23846999

RESUMO

Glycogen is an energy storage depot for the mammalian species. This review focuses on recent developments that have identified the role of nuclear hormone receptor (NR) signaling and epigenomic control in the regulation of important genes that modulate glycogen metabolism. Specifically, new studies have revealed that the NR4A subgroup (of the NR superfamily) are strikingly sensitive to beta-adrenergic stimulation in skeletal muscle, and transgenic studies in mice have revealed the expression of these NRs affects endurance and glycogen levels in muscle. Furthermore, other studies have demonstrated that one of the NR coregulator class of enzymes that mediate chromatin remodeling, the histone methyltransferases (for example, protein arginine methyltransferase 4) regulates the expression of several genes involved in glycogen metabolism and glycogen storage diseases in skeletal muscle. Importantly, NRs and histone methyltransferases, have the potential to be pharmacologically exploited and may provide novel targets in the quest to treat disorders of glycogen storage.


Assuntos
Epigenômica , Glicogênio/metabolismo , Músculo Esquelético/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Doença de Depósito de Glicogênio/fisiopatologia , Camundongos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/fisiologia , Proteína-Arginina N-Metiltransferases/metabolismo
16.
Ann Neurol ; 72(3): 433-41, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23034915

RESUMO

OBJECTIVE: Adult polyglucosan body disease (APBD) is an autosomal recessive leukodystrophy characterized by neurogenic bladder, progressive spastic gait, and peripheral neuropathy. Polyglucosan bodies accumulate in the central and peripheral nervous systems and are often associated with glycogen branching enzyme (GBE) deficiency. To improve clinical diagnosis and enable future evaluation of therapeutic strategies, we conducted a multinational study of the natural history and imaging features of APBD. METHODS: We gathered clinical, biochemical, and molecular findings in 50 APBD patients with GBE deficiency from Israel, the United States, France, and the Netherlands. Brain and spine magnetic resonance images were reviewed in 44 patients. RESULTS: The most common clinical findings were neurogenic bladder (100%), spastic paraplegia with vibration loss (90%), and axonal neuropathy (90%). The median age was 51 years for the onset of neurogenic bladder symptoms, 63 years for wheelchair dependence, and 70 years for death. As the disease progressed, mild cognitive decline may have affected up to half of the patients. Neuroimaging showed hyperintense white matter abnormalities on T2 and fluid attenuated inversion recovery sequences predominantly in the periventricular regions, the posterior limb of the internal capsule, the external capsule, and the pyramidal tracts and medial lemniscus of the pons and medulla. Atrophy of the medulla and spine was universal. p.Y329S was the most common GBE1 mutation, present as a single heterozygous (28%) or homozygous (48%) mutation. INTERPRETATION: APBD with GBE deficiency, with occasional exceptions, is a clinically homogenous disorder that should be suspected in patients with adult onset leukodystrophy or spastic paraplegia with early onset of urinary symptoms and spinal atrophy.


Assuntos
Doença de Depósito de Glicogênio , Imageamento por Ressonância Magnética , Doenças do Sistema Nervoso , Enzima Ramificadora de 1,4-alfa-Glucana/genética , Enzima Ramificadora de 1,4-alfa-Glucana/metabolismo , Adulto , Idoso , Córtex Cerebral/patologia , Feminino , França , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/patologia , Doença de Depósito de Glicogênio/fisiopatologia , Humanos , Israel , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Mutação/genética , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/fisiopatologia , Países Baixos , Exame Neurológico , Medula Espinal/patologia , Estados Unidos
17.
Curr Neurol Neurosci Rep ; 13(3): 333, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23335027

RESUMO

Disorders of glycogen metabolism are inborn errors of energy homeostasis affecting primarily skeletal muscle, heart, liver, and, less frequently, the central nervous system. These rare diseases are quite variable in age of onset, symptoms, morbidity, and mortality. This review provides an update on disorders of glycogen metabolism affecting skeletal muscle exclusively or predominantly. From a pathogenetic perspective, we classify these diseases as primary, if the defective enzyme is directly involved in glycogen/glucose metabolism, or secondary, if the genetic mutation affects proteins which indirectly regulate glycogen or glucose processing. In addition to summarizing the most recent clinical reports in this field, we briefly describe animal models of human glycogen disorders. These experimental models are greatly improving the understanding of the pathogenetic mechanisms underlying the muscle degenerative process associated to these diseases and provide in vivo platforms to test new therapeutic strategies.


Assuntos
Modelos Animais de Doenças , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/fisiopatologia , Doenças Neuromusculares/metabolismo , Doenças Neuromusculares/fisiopatologia , Animais , Glicogênio/metabolismo , Humanos , Doenças por Armazenamento dos Lisossomos/metabolismo , Doenças por Armazenamento dos Lisossomos/fisiopatologia , Doenças Musculares/metabolismo , Doenças Musculares/fisiopatologia
18.
Paediatr Anaesth ; 23(9): 817-23, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23565573

RESUMO

McArdles disease (glycogen storage disease type v) is a rare condition in which energy-metabolism in the muscle is hampered. A case report is presented and the possible risk for perioperative complications including malignant hyperthermia is discussed. A checklist for the anesthesiological management of patients with McArdles disease is provided. A short overview of anesthesiological challenges and perioperative complications of other glycogen storage diseases is given.


Assuntos
Anestesia , Doença de Depósito de Glicogênio Tipo V/terapia , Adolescente , Anestesia Geral , Criança , Pré-Escolar , Doença de Depósito de Glicogênio/fisiopatologia , Doença de Depósito de Glicogênio/terapia , Doença de Depósito de Glicogênio Tipo V/complicações , Humanos , Lactente , Recém-Nascido , Masculino , Hipertermia Maligna/genética , Hipertermia Maligna/fisiopatologia , Planejamento de Assistência ao Paciente , Adulto Jovem
19.
Acta Myol ; 30(2): 96-102, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22106711

RESUMO

In this selective review, we consider a number of unsolved questions regarding the glycogen storage diseases (GSD). Thus, the pathogenesis of Pompe disease (GSD II) is not simply explained by excessive intralysosomal glycogen storage and may relate to a more general dysfunction of autophagy. It is not clear why debrancher deficiency (GSD III) causes fixed myopathy rather than exercise intolerance, unless this is due to the frequent accompanying neuropathy. The infantile neuromuscular presentation of branching enzyme deficiency (GSD IV) is underdiagnosed and is finally getting the attention it deserves. On the other hand, the late-onset variant of GSD IV (adult polyglucosan body disease APBD) is one of several polyglucosan disorders (including Lafora disease) due to different etiologies. We still do not understand the clinical heterogeneity of McArdle disease (GSD V) or the molecular basis of the rare fatal infantile form. Similarly, the multisystemic infantile presentation of phosphofructokinase deficiency (GSD VII) is a conundrum. We observed an interesting association between phosphoglycerate kinase deficiency (GSD IX) and juvenile Parkinsonism, which is probably causal rather than casual. Also unexplained is the frequent and apparently specific association of phosphoglycerate mutase deficiency (GSD X) and tubular aggregates. By paying more attention to problems than to progress, we aimed to look to the future rather than to the past.


Assuntos
Metabolismo dos Carboidratos/genética , Doença de Depósito de Glicogênio , Músculo Esquelético/metabolismo , Músculo Liso/metabolismo , Adulto , Idade de Início , Biópsia , Criança , Progressão da Doença , Eletrodiagnóstico , Pesquisa Empírica , Pesquisa em Genética , Doença de Depósito de Glicogênio/classificação , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/patologia , Doença de Depósito de Glicogênio/fisiopatologia , Doença de Depósito de Glicogênio/terapia , Humanos , Lactente , Padrões de Herança , Músculo Esquelético/patologia , Músculo Liso/patologia , Terapias em Estudo
20.
J Cell Biol ; 170(1): 127-39, 2005 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-15998804

RESUMO

Lysosomal storage is the most common cause of neurodegenerative brain disease in preadulthood. However, the underlying cellular mechanisms that lead to neuronal dysfunction are unknown. Here, we report that loss of Drosophila benchwarmer (bnch), a predicted lysosomal sugar carrier, leads to carbohydrate storage in yolk spheres during oogenesis and results in widespread accumulation of enlarged lysosomal and late endosomal inclusions. At the bnch larval neuromuscular junction, we observe similar inclusions and find defects in synaptic vesicle recycling at the level of endocytosis. In addition, loss of bnch slows endosome-to-lysosome trafficking in larval garland cells. In adult bnch flies, we observe age-dependent synaptic dysfunction and neuronal degeneration. Finally, we find that loss of bnch strongly enhances tau neurotoxicity in a dose-dependent manner. We hypothesize that, in bnch, defective lysosomal carbohydrate efflux leads to endocytic defects with functional consequences in synaptic strength, neuronal viability, and tau neurotoxicity.


Assuntos
Metabolismo dos Carboidratos , Proteínas de Transporte/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Lisossomos/metabolismo , Degeneração Neural/metabolismo , Malformações do Sistema Nervoso/metabolismo , Animais , Sobrevivência Celular/genética , Modelos Animais de Doenças , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestrutura , Endocitose/genética , Endossomos/metabolismo , Endossomos/patologia , Endossomos/ultraestrutura , Feminino , Doença de Depósito de Glicogênio/genética , Doença de Depósito de Glicogênio/metabolismo , Doença de Depósito de Glicogênio/fisiopatologia , Larva/crescimento & desenvolvimento , Larva/metabolismo , Larva/ultraestrutura , Lisossomos/patologia , Lisossomos/ultraestrutura , Masculino , Proteínas de Membrana , Microscopia Eletrônica de Transmissão , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/fisiopatologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Junção Neuromuscular/ultraestrutura , Oogênese/genética , Transporte Proteico/genética , Saco Vitelino/metabolismo , Saco Vitelino/ultraestrutura , Proteínas tau/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA