Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
J Inherit Metab Dis ; 47(1): 93-118, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37421310

ABSTRACT

Glycogen storage disorders (GSDs) are inherited disorders of metabolism resulting from the deficiency of individual enzymes involved in the synthesis, transport, and degradation of glycogen. This literature review summarizes the development of gene therapy for the GSDs. The abnormal accumulation of glycogen and deficiency of glucose production in GSDs lead to unique symptoms based upon the enzyme step and tissues involved, such as liver and kidney involvement associated with severe hypoglycemia during fasting and the risk of long-term complications including hepatic adenoma/carcinoma and end stage kidney disease in GSD Ia from glucose-6-phosphatase deficiency, and cardiac/skeletal/smooth muscle involvement associated with myopathy +/- cardiomyopathy and the risk for cardiorespiratory failure in Pompe disease. These symptoms are present to a variable degree in animal models for the GSDs, which have been utilized to evaluate new therapies including gene therapy and genome editing. Gene therapy for Pompe disease and GSD Ia has progressed to Phase I and Phase III clinical trials, respectively, and are evaluating the safety and bioactivity of adeno-associated virus vectors. Clinical research to understand the natural history and progression of the GSDs provides invaluable outcome measures that serve as endpoints to evaluate benefits in clinical trials. While promising, gene therapy and genome editing face challenges with regard to clinical implementation, including immune responses and toxicities that have been revealed during clinical trials of gene therapy that are underway. Gene therapy for the glycogen storage diseases is under development, addressing an unmet need for specific, stable therapy for these conditions.


Subject(s)
Carcinoma, Hepatocellular , Glycogen Storage Disease Type II , Glycogen Storage Disease Type I , Glycogen Storage Disease , Liver Neoplasms , Animals , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Glycogen Storage Disease/metabolism , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , Glycogen Storage Disease Type I/complications , Liver/metabolism , Glycogen/metabolism , Genetic Therapy/methods , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology
2.
Mol Genet Metab ; 138(3): 107525, 2023 03.
Article in English | MEDLINE | ID: mdl-36796138

ABSTRACT

Glycogen storage disease type IV (GSD IV) is an ultra-rare autosomal recessive disorder caused by pathogenic variants in GBE1 which results in reduced or deficient glycogen branching enzyme activity. Consequently, glycogen synthesis is impaired and leads to accumulation of poorly branched glycogen known as polyglucosan. GSD IV is characterized by a remarkable degree of phenotypic heterogeneity with presentations in utero, during infancy, early childhood, adolescence, or middle to late adulthood. The clinical continuum encompasses hepatic, cardiac, muscular, and neurologic manifestations that range in severity. The adult-onset form of GSD IV, referred to as adult polyglucosan body disease (APBD), is a neurodegenerative disease characterized by neurogenic bladder, spastic paraparesis, and peripheral neuropathy. There are currently no consensus guidelines for the diagnosis and management of these patients, resulting in high rates of misdiagnosis, delayed diagnosis, and lack of standardized clinical care. To address this, a group of experts from the United States developed a set of recommendations for the diagnosis and management of all clinical phenotypes of GSD IV, including APBD, to support clinicians and caregivers who provide long-term care for individuals with GSD IV. The educational resource includes practical steps to confirm a GSD IV diagnosis and best practices for medical management, including (a) imaging of the liver, heart, skeletal muscle, brain, and spine, (b) functional and neuromusculoskeletal assessments, (c) laboratory investigations, (d) liver and heart transplantation, and (e) long-term follow-up care. Remaining knowledge gaps are detailed to emphasize areas for improvement and future research.


Subject(s)
Glycogen Storage Disease Type IV , Glycogen Storage Disease , Neurodegenerative Diseases , Child, Preschool , Humans , Glycogen Storage Disease Type IV/diagnosis , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/therapy , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Glycogen
3.
J Inherit Metab Dis ; 45(6): 1018-1027, 2022 11.
Article in English | MEDLINE | ID: mdl-36088581

ABSTRACT

Value-based healthcare (VBHC) intends to achieve better outcomes for patients, to improve quality of patient care, with reduced costs. Four dimensions define a model of intimately related value-pillars: personal value, allocative value, technical value, and societal value. VBHC is mostly applied in common diseases, and there are fundamental challenges in applying VBHC strategies to low volume, high complex healthcare situations, such as rare diseases, including inherited metabolic disorders. This article summarizes current practices at various academical domains (i.e., research, healthcare, education, and training) that (aim to) increase values at various value-pillars for persons with liver glycogen storage diseases or fatty acid oxidation disorders and their families. Future perspectives may include facilitating virtual networks to function as integrated practice units, improving measurement of outcomes, and creating information technology platforms to overcome the ethical, legal, societal, and technical challenges of data sharing for healthcare and research purposes.


Subject(s)
Acidosis , Glycogen Storage Disease , Lipid Metabolism, Inborn Errors , Muscular Diseases , Humans , Delivery of Health Care/methods , Fatty Acids , Glycogen Storage Disease/therapy , Lipid Metabolism, Inborn Errors/therapy , Liver
4.
Curr Opin Pediatr ; 34(5): 496-502, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35942643

ABSTRACT

PURPOSE OF REVIEW: Glycogen storage disease is a group of disorders primarily characterized by hepatomegaly and fasting hypoglycemia. This group of disorders may also affect the muscle, kidneys, and neurodevelopment. With an overall prevalence of 1 : 20 000, GSDs are disorders that clinicians should diagnose in a timely manner because adequate management can prevent complications, such as neurodevelopmental delay and liver disease [1] . As there are numerous types of GSDs, being able to distinguish one type from another can be overwhelming. In this review, we focus on hepatic GSDs to provide a concise review of clinical presentation, diagnosis, and current management. RECENT FINDINGS: GSDs are considered rare disorders, and one of the main challenges is the delay in diagnosis, misdiagnosis, or under diagnosis. However, with molecular genetic testing now readily available, confirming the diagnosis is no longer as difficult or invasive as it was in the past. SUMMARY: Current therapy for this group of disorders requires maintaining stable glucose levels. Avoiding hypoglycemia, as well as hyperglycemia, is critical in managing these patients. Being able to distinguish the types of GSDs and understanding the specific treatments for each enzymatic defect will optimize patient care.


Subject(s)
Glycogen Storage Disease , Hypoglycemia , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Humans , Hypoglycemia/diagnosis , Hypoglycemia/etiology , Hypoglycemia/therapy , Liver
5.
Transfus Apher Sci ; 61(1): 103289, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34690074

ABSTRACT

Herein, we aimed to present a child with extremely severe hypertriglyceridemia (ESHTG) secondary to diabetic ketoacidosis concomitant with type IX glycogen storage disease (GSD). Extremely severe hypertriglyceridemia (10 700 mg/dL) was detected through the apparent lipemic appearance of the sampled blood in a 17-year-old male patient with severe diabetic ketoacidosis. In spite of insulin infusion, the patient's clinical condition deteriorated to acute pancreatitis. Single sessions of therapeutic plasma exchange (TPE) along with insulin treatment have successfully intercepted the progression of the state of acute pancreatitis. The patient was also diagnosed with type IX GSD on the basis of the genetic analyses performed for the potential underlying metabolic diseases. In conclusion, underlying metabolic diseases, such as glycogen storage disease, should be investigated in patients with diabetic ketoacidosis accompanied by severe hypertriglyceridemia. If ESHTG does not relieve despite insulin infusion, and/or acute pancreatitis occurs as a complication, TPE should be kept in mind.


Subject(s)
Diabetes Complications/complications , Diabetic Ketoacidosis/etiology , Diabetic Ketoacidosis/therapy , Glycogen Storage Disease/complications , Glycogen Storage Disease/therapy , Hypertriglyceridemia/therapy , Plasma Exchange/methods , Adolescent , Diabetic Ketoacidosis/physiopathology , Glycogen Storage Disease/pathology , Humans , Male
6.
Hum Mol Genet ; 28(R1): R31-R41, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31227835

ABSTRACT

The focus of this review is the development of gene therapy for glycogen storage diseases (GSDs). GSD results from the deficiency of specific enzymes involved in the storage and retrieval of glucose in the body. Broadly, GSDs can be divided into types that affect liver or muscle or both tissues. For example, glucose-6-phosphatase (G6Pase) deficiency in GSD type Ia (GSD Ia) affects primarily the liver and kidney, while acid α-glucosidase (GAA) deficiency in GSD II causes primarily muscle disease. The lack of specific therapy for the GSDs has driven efforts to develop new therapies for these conditions. Gene therapy needs to replace deficient enzymes in target tissues, which has guided the planning of gene therapy experiments. Gene therapy with adeno-associated virus (AAV) vectors has demonstrated appropriate tropism for target tissues, including the liver, heart and skeletal muscle in animal models for GSD. AAV vectors transduced liver and kidney in GSD Ia and striated muscle in GSD II mice to replace the deficient enzyme in each disease. Gene therapy has been advanced to early phase clinical trials for the replacement of G6Pase in GSD Ia and GAA in GSD II (Pompe disease). Other GSDs have been treated in proof-of-concept studies, including GSD III, IV and V. The future of gene therapy appears promising for the GSDs, promising to provide more efficacious therapy for these disorders in the foreseeable future.


Subject(s)
Genetic Therapy , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Animals , Biomarkers , Clinical Trials as Topic , Combined Modality Therapy , Gene Editing , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/genetics , Glycogen Storage Disease/metabolism , Humans , Immunomodulation , Liver/metabolism , Organ Specificity , Standard of Care , Transduction, Genetic , Transgenes , Treatment Outcome
7.
Genet Med ; 21(4): 772-789, 2019 04.
Article in English | MEDLINE | ID: mdl-30659246

ABSTRACT

PURPOSE: Glycogen storage disease (GSD) types VI and IX are rare diseases of variable clinical severity affecting primarily the liver. GSD VI is caused by deficient activity of hepatic glycogen phosphorylase, an enzyme encoded by the PYGL gene. GSD IX is caused by deficient activity of phosphorylase kinase (PhK), the enzyme subunits of which are encoded by various genes: ɑ (PHKA1, PHKA2), ß (PHKB), É£ (PHKG1, PHKG2), and δ (CALM1, CALM2, CALM3). Glycogen storage disease types VI and IX have a wide spectrum of clinical manifestations and often cannot be distinguished from each other, or from other liver GSDs, on clinical presentation alone. Individuals with GSDs VI and IX can present with hepatomegaly with elevated serum transaminases, ketotic hypoglycemia, hyperlipidemia, and poor growth. This guideline for the management of GSDs VI and IX was developed as an educational resource for health-care providers to facilitate prompt and accurate diagnosis and appropriate management of patients. METHODS: A national group of experts in various aspects of GSDs VI and IX met to review the limited evidence base from the scientific literature and provided their expert opinions. Consensus was developed in each area of diagnosis, treatment, and management. Evidence bases for these rare disorders are largely based on expert opinion, particularly when targeted therapeutics that have to clear the US Food and Drug Administration (FDA) remain unavailable. RESULTS: This management guideline specifically addresses evaluation and diagnosis across multiple organ systems involved in GSDs VI and IX. Conditions to consider in a differential diagnosis stemming from presenting features and diagnostic algorithms are discussed. Aspects of diagnostic evaluation and nutritional and medical management, including care coordination, genetic counseling, and prenatal diagnosis are addressed. CONCLUSION: A guideline that will facilitate the accurate diagnosis and optimal management of patients with GSDs VI and IX was developed. This guideline will help health-care providers recognize patients with GSDs VI and IX, expedite diagnosis, and minimize adverse sequelae from delayed diagnosis and inappropriate management. It will also help identify gaps in scientific knowledge that exist today and suggest future studies.


Subject(s)
Genomics , Glycogen Storage Disease/genetics , Hypoglycemia/genetics , Phosphorylase Kinase/genetics , Disease Management , Genetics, Medical/trends , Glycogen/genetics , Glycogen/metabolism , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/epidemiology , Glycogen Storage Disease/therapy , Guidelines as Topic , Humans , Hypoglycemia/metabolism , Hypoglycemia/therapy , Liver/metabolism , Liver/pathology , Mutation , Phosphorylase Kinase/chemistry , United States/epidemiology
8.
Mol Ther ; 26(3): 814-821, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29428299

ABSTRACT

Glycogen storage disease type Ia (GSD1a) is an inherited metabolic disorder caused by the deficiency of glucose-6-phosphatase (G6Pase). GSD1a is associated with life-threatening hypoglycemia and long-term liver and renal complications. We examined the efficacy of mRNA-encoding human G6Pase in a liver-specific G6Pase-/- mouse model (L-G6PC-/-) that exhibits the same hepatic biomarkers associated with GSD1a patients, such as fasting hypoglycemia, and elevated levels of hepatic glucose-6-phosphate (G6P), glycogen, and triglycerides. We show that a single systemic injection of wild-type or native human G6PC mRNA results in significant improvements in fasting blood glucose levels for up to 7 days post-dose. These changes were associated with significant reductions in liver mass, hepatic G6P, glycogen, and triglycerides. In addition, an engineered protein variant of human G6Pase, designed for increased duration of expression, showed superior efficacy to the wild-type sequence by maintaining improved fasting blood glucose levels and reductions in liver mass for up to 12 days post-dose. Our results demonstrate for the first time the effectiveness of mRNA therapy as a potential treatment in reversing the hepatic abnormalities associated with GSD1a.


Subject(s)
Blood Glucose , Genetic Therapy , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease/genetics , Glycogen Storage Disease/metabolism , Liver/metabolism , RNA, Messenger/genetics , Animals , Biomarkers , Disease Models, Animal , Fasting , Gene Expression , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease/pathology , Glycogen Storage Disease/therapy , Immunohistochemistry , Liver/pathology , Male , Metabolic Networks and Pathways , Mice , Mice, Knockout , Protein Engineering
9.
Curr Opin Neurol ; 30(5): 449-456, 2017 10.
Article in English | MEDLINE | ID: mdl-28737584

ABSTRACT

PURPOSE OF REVIEW: The field of muscle glycogenoses has progressed in recent years by the identification of new disorders, and by reaching a better understanding of pathophysiology of the disorders and the physiology of glycogen metabolism. RECENT FINDINGS: In this review, we describe the clinical and pathological features of the three most recently described muscle glycogenoses caused by recessive mutations in GYG1, RBCK1 and PGM1. The three involved enzymes play different roles in glycogen metabolism. Glycogenin-1 (GYG1) is involved in the initial steps of glycogen synthesis, whereas phosphoglucomutase catalyzes two metabolic pathways; the connection between galactose and glycogen on one side, and glucose metabolism on the other side. The metabolic consequences of mutations in the ubiquitin ligase gene RBCK1 are still poorly understood. GYG1 deficiency has been associated with cardiomyopathies with abnormal storage material in the heart, but most cases present with a polyglucosan body myopathy without cardiac involvement. SUMMARY: The recent identification of new glycogenosis not only allows to improve the knowledge of glycogen metabolism, but also builds bridges with protein glycosylation and immune system.


Subject(s)
Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Muscular Diseases/genetics , Muscular Diseases/therapy , Glucosyltransferases/genetics , Glycogen/metabolism , Glycoproteins/genetics , Humans , Mutation , Phosphoglucomutase/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics
10.
Biochim Biophys Acta ; 1852(4): 615-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24997454

ABSTRACT

Metabolic myopathies are disorders of utilization of carbohydrates or fat in muscles. The acute nature of energy failure is manifested either by a metabolic crisis with weakness, sometimes associated with respiratory failure, or by myoglobinuria. A typical disorder where permanent weakness occurs is glycogenosis type II (GSDII or Pompe disease) both in infantile and late-onset forms, where respiratory insufficiency is manifested by a large number of cases. In GSDII the pathogenetic mechanism is still poorly understood, and has to be attributed more to structural muscle alterations, possibly in correlation to macro-autophagy, rather than to energetic failure. This review is focused on recent advances about GSDII and its treatment, and the most recent notions about the management and treatment of other metabolic myopathies will be briefly reviewed, including glycogenosis type V (McArdle disease), glycogenosis type III (debrancher enzyme deficiency or Cori disease), CPT-II deficiency, and ETF-dehydrogenase deficiency (also known as riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency or RR-MADD). The discovery of the genetic defect in ETF dehydrogenase confirms the etiology of this syndrome. Other metabolic myopathies with massive lipid storage and weakness are carnitine deficiency, neutral lipid storage-myopathy (NLSD-M), besides RR-MADD. Enzyme replacement therapy is presented with critical consideration and for each of the lipid storage disorders, representative cases and their response to therapy is included. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.


Subject(s)
Electron-Transferring Flavoproteins/deficiency , Enzyme Replacement Therapy , Glycogen Storage Disease , Iron-Sulfur Proteins/deficiency , Lipid Metabolism, Inborn Errors , Muscular Diseases , Oxidoreductases Acting on CH-NH Group Donors/deficiency , Animals , Glycogen Storage Disease/enzymology , Glycogen Storage Disease/genetics , Glycogen Storage Disease/pathology , Glycogen Storage Disease/therapy , Humans , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/therapy , Muscular Diseases/enzymology , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Diseases/therapy
11.
Rev Neurol (Paris) ; 172(10): 541-545, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27663060

ABSTRACT

The field of glycogenosis has been greatly expanded over the past few years with the discovery of new metabolic diseases that have allowed new metabolic pathways to be deciphered. Described here are the clinical and pathological features of four recently described muscle glycogenoses caused by GYS1, GYG1, RBCK1 and PGM1 gene mutations. The initial steps of glycogen synthesis are involved in deficiencies of glycogenin-1 (GYG1) and muscle glycogen synthase (GYS1). Phosphoglucomutase deficiency disrupts two metabolic pathways: the connection between galactose and glycogen on the one hand, and glucose metabolism on the other. However, the metabolic consequences of mutations in the ubiquitin ligase gene RBCK1 are still poorly understood.


Subject(s)
Glycogen Storage Disease/therapy , Glycogen Storage Disease/genetics , Glycogen Storage Disease/pathology , Humans , Muscle, Skeletal/pathology
12.
J Inherit Metab Dis ; 38(3): 505-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25224826

ABSTRACT

Glycogen storage diseases (GSD), a unique category of inherited metabolic disorders, were first described early in the twentieth century. Since then, the biochemical and genetic bases of these disorders have been determined, and an increasing number of animal models for GSD have become available. At least seven large mammalian models have been developed for laboratory research on GSDs. These models have facilitated the development of new therapies, including gene therapy, which are undergoing clinical translation. For example, gene therapy prolonged survival and prevented hypoglycemia during fasting for greater than one year in dogs with GSD type Ia, and the need for periodic re-administration to maintain efficacy was demonstrated in that dog model. The further development of gene therapy could provide curative therapy for patients with GSD and other inherited metabolic disorders.


Subject(s)
Dependovirus/genetics , Glycogen Storage Disease/classification , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Hypoglycemia/genetics , Animals , Cats , Disease Models, Animal , Dogs , Genetic Therapy , Genetic Vectors , Sheep
13.
Mol Genet Metab ; 113(3): 171-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25266922

ABSTRACT

OBJECTIVES: Glycogen storage disease (GSD) types VI and IX are caused by phosphorylase system deficiencies. To evaluate the natural history and long-term treatment outcome of the patients with GSD-VI and -IX, we performed an observational retrospective case study of 21 patients with confirmed diagnosis of GSD-VI or -IX. METHODS: All patients with GSD-VI or -IX, diagnosed at The Hospital for Sick Children, were included. Electronic and paper charts were reviewed for clinical features, biochemical investigations, molecular genetic testing, diagnostic imaging, long-term outcome and treatment by two independent research team members. All information was entered into an Excel database. RESULTS: We report on the natural history and treatment outcomes of the 21 patients with GSD-VI and -IX and 16 novel pathogenic mutations in the PHKA2, PHKB, PHKG2 and PYGL genes. We report for the first time likely liver adenoma on liver ultrasound and liver fibrosis on liver biopsy specimens in patients with GSD-VI and mild cardiomyopathy on echocardiography in patients with GSD-VI and -IXb. CONCLUSION: We recommend close monitoring in all patients with GSD-VI and -IX for the long-term liver and cardiac complications. There is a need for future studies if uncooked cornstarch and high protein diet would be able to prevent long-term complications of GSD-VI and -IX.


Subject(s)
Glycogen Storage Disease Type VI/genetics , Adolescent , Canada , Child , Child, Preschool , DNA Mutational Analysis , Female , Glycogen Storage Disease/complications , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Glycogen Storage Disease Type VI/complications , Glycogen Storage Disease Type VI/therapy , Humans , Liver Cirrhosis/genetics , Male , Retrospective Studies , Treatment Outcome
14.
Cells ; 13(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38920680

ABSTRACT

Human induced pluripotent stem cell (iPSC) and CRISPR-Cas9 gene-editing technologies have become powerful tools in disease modeling and treatment. By harnessing recent biotechnological advancements, this review aims to equip researchers and clinicians with a comprehensive and updated understanding of the evolving treatment landscape for metabolic and genetic disorders, highlighting how iPSCs provide a unique platform for detailed pathological modeling and pharmacological testing, driving forward precision medicine and drug discovery. Concurrently, CRISPR-Cas9 offers unprecedented precision in gene correction, presenting potential curative therapies that move beyond symptomatic treatment. Therefore, this review examines the transformative role of iPSC technology and CRISPR-Cas9 gene editing in addressing metabolic and genetic disorders such as alpha-1 antitrypsin deficiency (A1AD) and glycogen storage disease (GSD), which significantly impact liver and pulmonary health and pose substantial challenges in clinical management. In addition, this review discusses significant achievements alongside persistent challenges such as technical limitations, ethical concerns, and regulatory hurdles. Future directions, including innovations in gene-editing accuracy and therapeutic delivery systems, are emphasized for next-generation therapies that leverage the full potential of iPSC and CRISPR-Cas9 technologies.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Glycogen Storage Disease , Induced Pluripotent Stem Cells , alpha 1-Antitrypsin Deficiency , Humans , alpha 1-Antitrypsin Deficiency/therapy , alpha 1-Antitrypsin Deficiency/genetics , Induced Pluripotent Stem Cells/metabolism , CRISPR-Cas Systems/genetics , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Glycogen Storage Disease/metabolism , Gene Editing/methods , Genetic Therapy/methods , Animals
15.
Neurotherapeutics ; 21(5): e00446, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39277505

ABSTRACT

Glycogen storage diseases (GSDs) comprise a group of inherited metabolic disorders characterized by defects in glycogen metabolism, leading to abnormal glycogen accumulation in multiple tissues, most notably affecting the liver, skeletal muscle, and heart. Recent findings have uncovered the importance of glycogen metabolism in the brain, sustaining a myriad of physiological functions and linking its perturbation to central nervous system (CNS) pathology. This link resulted in classification of neurological-GSDs (n-GSDs), a group of diseases with shared deficits in neurological glycogen metabolism. The n-GSD patients exhibit a spectrum of clinical presentations with common etiology while requiring tailored therapeutic approaches from the traditional GSDs. Recent research has elucidated the genetic and biochemical mechanisms and pathophysiological basis underlying different n-GSDs. Further, the last decade has witnessed some promising developments in novel therapeutic approaches, including enzyme replacement therapy (ERT), substrate reduction therapy (SRT), small molecule drugs, and gene therapy targeting key aspects of glycogen metabolism in specific n-GSDs. This preclinical progress has generated noticeable success in potentially modifying disease course and improving clinical outcomes in patients. Herein, we provide an overview of current perspectives on n-GSDs, emphasizing recent advances in understanding their molecular basis, therapeutic developments, underscore key challenges and the need to deepen our understanding of n-GSDs pathogenesis to develop better therapeutic strategies that could offer improved treatment and sustainable benefits to the patients.


Subject(s)
Genetic Therapy , Glycogen Storage Disease , Humans , Glycogen Storage Disease/therapy , Glycogen Storage Disease/metabolism , Glycogen Storage Disease/genetics , Animals , Genetic Therapy/methods , Genetic Therapy/trends , Glycogen/metabolism , Enzyme Replacement Therapy/methods , Nervous System Diseases/therapy , Nervous System Diseases/metabolism
16.
Curr Opin Neurol ; 26(5): 544-53, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23995275

ABSTRACT

PURPOSE OF REVIEW: This review highlights recent contributions regarding clinical heterogeneity, pathogenic mechanisms, therapeutic trials, and animal models of the muscle glycogenoses. RECENT FINDINGS: Most recent publications have dealt with the clinical effects of enzyme replacement therapy (ERT) in glycogenosis type II (Pompe disease), including the cognitive development of children with the infantile form who have reached school age. Standardized exercise testing has shown the similarity between McArdle disease and one of the most recently described muscle glycogenoses, phosphoglucomutase deficiency. Cycle ergometry in patients with glycogenosis type III (debrancher deficiency) without overt weakness has documented exercise intolerance relieved by glucose infusion, consistent with the glycogenolytic block. A mouse model of McArdle disease faithfully recapitulates most features of the human disease and will prove valuable for a better understanding of pathogenesis and therapeutic modalities. Polyglucosan body myopathy with cardiomyopathy has been associated with mutations in RBCK1, a ubiquitin ligase, which have also been reported in children with early-onset immune disorder. The role of polyglucosan storage in muscle and in both central and peripheral nervous systems has been confirmed in the infantile and late-onset forms of glycogenosis type IV (brancher enzyme deficiency). Additional novel findings include the involvement of the heart in one patient with phosphofructokinase (PFK) deficiency and the presence of tubular aggregates in a manifesting heterozygote with phosphoglycerate mutase deficiency. SUMMARY: Important recent developments in the field of muscle glycogenoses include a new disease entity, a new animal model of McArdle disease, and better knowledge of the pathogenesis in some glycogenoses and of the long-term effects of enzyme replacement therapy in Pompe disease.


Subject(s)
Glycogen Storage Disease/therapy , Animals , Disease Models, Animal , Enzyme Replacement Therapy/methods , Exercise/physiology , Glycogen Storage Disease/etiology , Glycogen Storage Disease/genetics , Glycogen Storage Disease/pathology , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/congenital , Muscular Diseases/pathology , Muscular Diseases/therapy
17.
J Hum Genet ; 58(5): 285-92, 2013 May.
Article in English | MEDLINE | ID: mdl-23486339

ABSTRACT

Many reports have been published on the long-term outcome and treatment of hepatic glycogen storage diseases (GSDs) overseas; however, none have been published from Japan. We investigated the clinical manifestations, treatment, and prognosis of 127 hepatic GSD patients who were evaluated and treated between January 1999 and December 2009. A characteristic genetic pattern was noted in the Japanese GSD patients: most GSD Ia patients had the g727t mutation, and many GSD Ib patients had the W118R mutation. Forty-one percent (14/34) of GSD Ia patients and 18% (2/11) of GSD Ib patients of ages 13 years 4 months had liver adenoma. Among subjects aged 10 years, 19% (7/36) of the GSD Ia patients and none of the GSD Ib patients had renal dysfunction. The mean height of male GSD Ia patients aged 18 years was 160.8±10.6 cm (n=14), and that of their female counterparts was 147.8±3.80 cm (n=9). Patients with hepatic GSDs develop a variety of symptoms but can survive in the long term by diet therapy, corn starch treatment and supportive care. Liver transplantation for hepatic GSDs is an important treatment strategy and can help improve the patients'quality of life.


Subject(s)
Glycogen Storage Disease/epidemiology , Adolescent , Adult , Age Factors , Age of Onset , Body Height , Child , Child, Preschool , Female , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/therapy , Humans , Infant , Infant, Newborn , Japan/epidemiology , Liver Transplantation , Male , Prognosis , Young Adult
19.
Paediatr Anaesth ; 23(9): 817-23, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23565573

ABSTRACT

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.


Subject(s)
Anesthesia , Glycogen Storage Disease Type V/therapy , Adolescent , Anesthesia, General , Child , Child, Preschool , Glycogen Storage Disease/physiopathology , Glycogen Storage Disease/therapy , Glycogen Storage Disease Type V/complications , Humans , Infant , Infant, Newborn , Male , Malignant Hyperthermia/genetics , Malignant Hyperthermia/physiopathology , Patient Care Planning , Young Adult
20.
World J Gastroenterol ; 29(25): 3932-3963, 2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37476587

ABSTRACT

Glycogen storage diseases (GSDs), also referred to as glycogenoses, are inherited metabolic disorders of glycogen metabolism caused by deficiency of enzymes or transporters involved in the synthesis or degradation of glycogen leading to aberrant storage and/or utilization. The overall estimated GSD incidence is 1 case per 20000-43000 live births. There are over 20 types of GSD including the subtypes. This heterogeneous group of rare diseases represents inborn errors of carbohydrate metabolism and are classified based on the deficient enzyme and affected tissues. GSDs primarily affect liver or muscle or both as glycogen is particularly abundant in these tissues. However, besides liver and skeletal muscle, depending on the affected enzyme and its expression in various tissues, multiorgan involvement including heart, kidney and/or brain may be seen. Although GSDs share similar clinical features to some extent, there is a wide spectrum of clinical phenotypes. Currently, the goal of treatment is to maintain glucose homeostasis by dietary management and the use of uncooked cornstarch. In addition to nutritional interventions, pharmacological treatment, physical and supportive therapies, enzyme replacement therapy (ERT) and organ transplantation are other treatment approaches for both disease manifestations and long-term complications. The lack of a specific therapy for GSDs has prompted efforts to develop new treatment strategies like gene therapy. Since early diagnosis and aggressive treatment are related to better prognosis, physicians should be aware of these conditions and include GSDs in the differential diagnosis of patients with relevant manifestations including fasting hypoglycemia, hepatomegaly, hypertransaminasemia, hyperlipidemia, exercise intolerance, muscle cramps/pain, rhabdomyolysis, and muscle weakness. Here, we aim to provide a comprehensive review of GSDs. This review provides general characteristics of all types of GSDs with a focus on those with liver involvement.


Subject(s)
Glycogen Storage Disease , Humans , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Liver/metabolism , Glycogen/metabolism , Muscle, Skeletal/metabolism , Genetic Therapy
SELECTION OF CITATIONS
SEARCH DETAIL