Category: Moms

Research on glycogen storage disease

Research on glycogen storage disease

Article CAS PubMed Central PubMed Google Scholar Moses, S. Despite gross glycogenn, the Electrolyte balance benefits may be largely asymptomatic without hypoglycemia. Ai JHe W, Huang X, Wu Y, Lei Y, Yu C, Görgülü K, Diakopoulos KN, Lu N, Zhu Y.

Video

Glycogen Storage Disorders -Center for Rare Disease Therapy -UPMC Children's Hospital of Pittsburgh Stotage storage diseases Energy drinks for immune system encompass glycogeb group of rare Reseaech diseases due dysfunction of glycogen metabolism. Hypoglycemia is Diabetes complications coma most Researdh primary manifestation of GSD, and Electrolyte balance benefits Rdsearch glucose metabolism can Sgorage neurological damage. The aims of this study Electrolyte balance benefits to first investigate the metabolic, genetic, and neurological profiles of children with GSD, and to test the hypothesis whether GSD type I would have greater neurological impact than GSD type IX. Genetic testing was conducted for the following genes using multigene panel analysis. The biochemical data and magnetic resonance imaging of the brain presented by the patients were evaluated. The criteria of adequate metabolic control were adopted based on the European Study on Glycogen Storage Disease type I consensus.

Research on glycogen storage disease -

Indeed, a metabolic imbalance results in overnight hypoglycemia and ketosis, that are associated to short stature, osteopenia, and neurologic complications [ 43 ]. GSDs types 0, VI and particularly type IX would benefit from a strict glycemia monitoring.

A minority of patients with mutations of PHKA2 and PHKG2 associated to a severe phenotype often require overnight feeding to maintain euglycemia [ 85 ].

Since gluconeogenesis is preserved, protein supplementation provides gluconeogenic precursors that can be used for repletion of Krebs cycle intermediates and endogenous glucose production in GSD types 0, IV, VI and IX. By improving glucose homeostasis, hepatic glycogen accumulation and secondary complications might be restrained.

High protein intake is especially needed in GSD type VI to improve muscle function [ 44 ]. In Ross and co-workers [ 85 ] described the efficacy of an extended-release cornstarch Glycosade in GSD types 0, III, VI and IX to achieve a longer time of euglycemia during the night, with stable values of other markers of metabolic control and hepatic function.

In the United States, the extended-release cornstarch preparation has been approved for nocturnal use in GSD patients above 5 years of age. However, the administration of Glycosade in patient between 2 and 5 years of age resulted safe and effective as well [ 86 ].

Adverse effects such as abdominal distension, diarrhea and flatulence have been reported, but to date they were not recorded in patients with GSD types 0, VI and IX [ 61 ].

Patients with GSD type 0 are treated with frequent feeds of hyperglucidic diet plus cornstarch and protein supplementation. Patients with GSD type IV are managed with hyperglucidic diet plus cornstarch, nocturnal enteral feeding, protein enrichment with the aim to limit the accumulation of glycogen, to prevent catabolism and to improve growth and fasting tolerance.

The more severe forms are treated with liver transplantation [ 26 ]. For GSD type XI, Pennisi and co-workers [ 63 ] proposed the nocturnal enteral nutrition in younger children and in patients with a severe growth delay in order to prevent fasting hypoglycemia.

Frequent, small meals, restricted in glucose and galactose, and raw cornstarch administration at night are used to prevent metabolic acidosis, which may occur at times of surgery or other stresses.

Hypercholesterolemia may require a medical treatment with statins after five years of age; bicarbonate supplementation may be required to balance the urinary bicarbonate loss [ 63 ]. According to the available data, universally accepted guidelines for the management of these types of GSDs have not been defined.

Nevertheless, an appropriate follow-up should be provided, in order to establish a good metabolic control and monitor the possible complications. Medical and nutritional evaluations and blood assessment, including complete liver and renal function, lipid profile, calcium-phosphate metabolism, serum electrolytes, blood gas analysis and urinalysis, should be fulfilled every 6 months on average; a higher frequency is recommended in younger patients and in those who have not achieved a metabolic balance.

A continuous glucose monitoring may be helpful to survey the glycemic fluctuations, especially in younger patients. Alpha-fetoprotein levels along with abdomen ultrasound can be used to screen for hepatocellular carcinoma, even though there are no validated surveillance protocols to date [ 37 ].

GSD type IV patients require a complete cardiac function evaluation, including electrocardiogram and echocardiography.

For patients with GSD types VI and IX after 5 years of age a cardiac evaluation is recommended every 1—2 years [ 44 ]. Regarding the bone metabolism, a careful assessment of calcium and vitamin D intake and monitoring of OH vitamin D level is recommended.

Calcium, phosphate and vitamin D supplementations, along with annual DXA scan evaluation, are required to prevent osteopenia and fractures, particularly in GSD type XI, along with a surveillance of renal function [ 61 ].

Skeletal X-Rays are required in GSD type XI to evaluate rickets evolution [ 55 , 56 ]. GSDs type 0, IV, VI, IX and XI with liver involvement may have a similar clinical presentation.

However, these diseases exhibit a phenotypic continuum, and even in the mildest forms, regular monitoring and dietary adjustments are necessary to restrain disease progression and complications. Some cases may exhibit a clinical burden with severe organ complications.

Building a proper knowledge among physicians about these rare conditions is crucial to improve prognosis and quality of life of patients, especially those affected by the most severe forms.

Further studies are needed to outline the genotype—phenotype correlation and define personalized therapies and management. Ozen H. Glycogen storage diseases: new perspectives. World J Gastroenterol. Article CAS PubMed PubMed Central Google Scholar. Roach PJ, Depaoli-Roach AA, Hurley TD, Tagliabracci VS.

Glycogen and its metabolism: some new developments and old themes. Biochem J. Article CAS PubMed Google Scholar. Beyzaei Z, Geramizadeh B, Karimzadeh S.

Diagnosis of hepatic glycogen storage disease patients with overlapping clinical symptoms by massively parallel sequencing: a systematic review of literature. Orphanet J Rare Dis. Article PubMed PubMed Central Google Scholar.

Fernandes SA, Cooper GE, Gibson RA, Kishnani PS. Benign or not benign? deep phenotyping of liver glycogen storage disease IX. Mol Genet Metab. Weinstein DA, Correia CE, Saunders AC, Wolfsdorf JI.

Hepatic glycogen synthase deficiency: an infrequently recognized cause of ketotic hypoglycemia. Brown LM, Corrado MM, van der Ende RM, Derks TG, Chen MA, Siegel S, et al. Evaluation of glycogen storage disease as a cause of ketotic hypoglycemia in children.

J Inherit Metab Dis. Grünert SC, Schwab KO, Pohl M, Sass JO, Santer R. Fanconi-Bickel syndrome: GLUT2 mutations associated with a mild phenotype. Cameron JM, Levandovskiy V, MacKay N, Utgikar R, Ackerley C, Chiasson D, et al. Identification of a novel mutation in GYS1 muscle-specific glycogen synthase resulting in sudden cardiac death, that is diagnosable from skin fibroblasts.

Arko JJ, Debeljak M, Tansek MZ, Battelino T, Groselj U. A patient with glycogen storage disease type 0 and a novel sequence variant in GYS2: a case report and literature review. J Int Med Res. Article PubMed Google Scholar.

Spiegel R, Mahamid J, Orho-Melander M, Miron D, Horovitz Y. The variable clinical phenotype of liver glycogen synthase deficiency. J Pediatr Endocrinol Metab. Soggia AP, Correa-Giannella ML, Fortes MA, Luna AM, Pereira MA. A novel mutation in the glycogen synthase 2 gene in a child with glycogen storage disease type 0.

BMC Med Genet. Orho M, Bosshard NU, Buist NR, Gitzelmann R, Aynsley-Green A, Blümel P, et al. Mutations in the liver glycogen synthase gene in children with hypoglycemia due to glycogen storage disease type 0.

J Clin Invest. Kamenets EA, Gusarova EA, Milovanova NV, Itkis YS, Strokova TV, Melikyan MA, et al. Hepatic glycogen synthase GYS2 deficiency: seven novel patients and seven novel variants.

JIMD Rep. Bachrach BE, Weinstein DA, Orho-Melander M, Burgess A, Wolfsdorf JI. Glycogen synthase deficiency glycogen storage disease type 0 presenting with hyperglycemia and glucosuria: report of three new mutations.

J Pediatr. Gitzelmann R, Spycher MA, Feil G, Müller J, Seilnacht B, Stahl M, et al. Liver glycogen synthase deficiency: a rarely diagnosed entity. Eur J Pediatr. Matei L, Teodorescu MI, Kozma A, Iordan Dumitru AD, Stoicescu SM, Carniciu S.

Persistent asymptomatic severe hypoglycaemia due to type 0a Glycogenosis—general and oro-dental aspects. Acta Endocrinol Buchar.

Article CAS Google Scholar. Kasapkara ÇS, Aycan Z, Açoğlu E, Senel S, Oguz MM, Ceylaner S. The variable clinical phenotype of three patients with hepatic glycogen synthase deficiency. Ponzi E, Maiorana A, Lepri FR, Mucciolo M, Semeraro M, Taurisano R, et al.

Persistent hypoglycemia in children: targeted gene panel improves the diagnosis of hypoglycemia due to inborn errors of metabolism. Sandhu T, Polan M, Yu Z, Lu R, Makkar A. Case of neonatal fatality from neuromuscular variant of glycogen storage disease type IV.

Magoulas PL, El-Hattab AW. Glycogen Storage Disease Type IV. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Mirzaa GM, Amemiya A, editors.

GeneReviews® [Internet]. Seattle WA : University of Washington, Seattle; [updated ]. Szymańska E, Szymańska S, Truszkowska G, Ciara E, Pronicki M, Shin YS. Variable clinical presentation of glycogen storage disease type IV: from severe hepatosplenomegaly to cardiac insufficiency.

Some discrepancies in genetic and biochemical abnormalities. Arch Med Sci. Zellweger H, Mueller S, Ionasescu V, Schochet SS, McCormick WF. A new cause of infantile hypotonia. Greene HL, Ghishan FK, Brown B, McClenathan DT, Freese D. Hypoglycemia in type IV glycogenosis: hepatic improvement in two patients with nutritional management.

Moses SW, Parvari R. The variable presentations of glycogen storage disease type IV: a review of clinical, enzymatic and molecular studies. Curr Mol Med. Sreekantam S, Rizvi H, Brown R, Santra S, Raiman J, Vijay S, et al.

An uncommon cause of early infantile liver disease and raised chitotriosidase. Derks TGJ, Peeks F, de Boer F, Fokkert-Wilts M, van der Doef HPJ, van den Heuvel MC, et al. The potential of dietary treatment in patients with glycogen storage disease type IV.

Sokal EM, Van Hoof F, Alberti D, de Ville de Goyet J, de Barsy T, Otte JB. Progressive cardiac failure following orthotopic liver transplantation for type IV glycogenosis.

Rosenthal P, Podesta L, Grier R, Said JW, Sher L, Cocjin J, et al. Failure of liver transplantation to diminish cardiac deposits of amylopectin and leukocyte inclusions in type IV glycogen storage disease. Liver Transplant Surg. Liu M, Sun LY.

Liver transplantation for glycogen storage disease type IV. Front Pediatr. McConkie-Rosell A, Wilson C, Piccoli DA, Boyle J, DeClue T, Kishnani P, et al. Clinical and laboratory findings in four patients with the non-progressive hepatic form of type IV glycogen storage disease.

Dhawan A, Tan KC, Portmann B, Mowat AP. Glycogenosis type IV: liver transplant at 12 years. Arch Dis Child. Lamperti C, Salani S, Lucchiari S, Bordoni A, Ripolone M, Fagiolari G, et al. Neuropathological study of skeletal muscle, heart, liver, and brain in a neonatal form of glycogen storage disease type IV associated with a new mutation in GBE1 gene.

Tarnopolsky MA. Myopathies related to glycogen metabolism disorders. Labrador E, Weinstein DA. Glycogen Storage Disease Type VI. P — Burwinkel B, Bakker HD, Herschkovitz E, Moses SW, Shin YS, Kilimann MW. Mutations in the liver glycogen phosphorylase gene PYGL underlying glycogenosis type VI.

Am J Hum Genet. Grünert SC, Hannibal L, Spiekerkoetter U. the phenotypic and genetic spectrum of glycogen storage disease type VI. Genes Basel. Kishnani PS, Goldstein J, Austin SL, Arn P, Bachrach B, Bali DS, et al. ACMG Work group on diagnosis and management of glycogen storage diseases type VI and IX.

Diagnosis and management of glycogen storage diseases type VI and IX: a clinical practice resource of the American College of Medical Genetics and Genomics ACMG.

Genet Med. Lu SQ, Feng JY, Liu J, Xie XB, Lu Y, Abuduxikuer K. Glycogen storage disease type VI can progress to cirrhosis: ten Chinese patients with GSD VI and a literature review.

Burda P, Hochuli M. Hepatic glycogen storage disorders: what have we learned in recent years? Curr Opin Clin Nutr Metab Care. Beyzaei Z, Ezgu F, Geramizadeh B, Alborzi A, Shojazadeh A. Novel mutations in the PHKB gene in an iranian girl with severe liver involvement and glycogen storage disease type IX: a case report and review of literature.

BMC Pediatr. Hendrickx J, Coucke P, Hors-Cayla MC, Smit GP, Shin YS, Deutsch J, et al. Localization of a new type of X-linked liver glycogenosis to the chromosomal region Xp22 containing the liver alpha-subunit of phosphorylase kinase PHKA2. Fu J, Wang T, Xiao X. A novel PHKA2 mutation in a Chinese child with glycogen storage disease type IXa: a case report and literature review.

Tsilianidis LA, Fiske LM, Siegel S, Lumpkin C, Hoyt K, Wasserstein M, et al. Aggressive therapy improves cirrhosis in glycogen storage disease type IX. Roscher A, Patel J, Hewson S, Nagy L, Feigenbaum A, Kronick J, et al.

The natural history of glycogen storage disease types VI and IX: long-term outcome from the largest metabolic center in Canada. Johnson AO, Goldstein JL, Bali D. Glycogen storage disease type IX: novel PHKA2 missense mutation and cirrhosis.

J Pediatr Gastroenterol Nutr. Rodríguez-Jiménez C, Santos-Simarro F, Campos-Barros Á, Camarena C, Lledín D, Vallespín E, et al. A new variant in PHKA2 is associated with glycogen storage disease type IXa. Mol Genet Metab Rep. Burwinkel B, Amat L, Gray RG, Matsuo N, Muroya K, Narisawa K, et al.

Variability of biochemical and clinical phenotype in X-linked liver glycogenosis with mutations in the phosphorylase kinase PHKA2 gene. Hum Genet. Burwinkel B, Maichele AJ, Aagenaes O, Bakker HD, Lerner A, Shin YS, et al.

Autosomal glycogenosis of liver and muscle due to phosphorylase kinase deficiency is caused by mutations in the phosphorylase kinase beta subunit PHKB. Hum Mol Genet. Burwinkel B, Rootwelt T, Kvittingen EA, Chakraborty PK, Kilimann MW.

Severe phenotype of phosphorylase kinase-deficient liver glycogenosis with mutations in the PHKG2 gene. Pediatr Res. Chen MA, Weinstein DA. Glycogen storage diseases: diagnosis, treatment and outcome. Transl Sci Rare Dis. Article Google Scholar. Waheed N, Saeed A, Ijaz S, Fayyaz Z, Anjum MN, Zahoor Y, et al.

Variability of clinical and biochemical phenotype in liver phosphorylase kinase deficiency with variants in the phosphorylase kinase PHKG2 gene. Al-Haggar M. Fanconi—Bickel syndrome as an example of marked allelic heterogeneity. World J Nephrol. Sharari S, Abou-Alloul M, Hussain K, Ahmad KF.

Fanconi—Bickel syndrome: a review of the mechanisms that lead to dysglycaemia. Int J Mol Sci. Article CAS PubMed Central Google Scholar. Mohandas Nair K, Sakamoto O, Jagadeesh S, Nampoothiri S.

Fanconi—Bickel syndrome. Indian J Pediatr. Afroze B, Chen M. Fanconi—Bickel syndrome: two pakistani patients presenting with hypophosphatemic rickets. J Pediatr Genet. Sahin F, Sipahi T, Doğan H, Oksal A, Ertan U. Special feature: pathological case of the month.

Glycogen storage disease with renal tubular dysfunction type XI, Fanconi-Bickel syndrome. Arch Pediatr Adolesc Med. Seyer P, Vallois D, Poitry-Yamate C, Schütz F, Metref S, Tarussio D, et al.

Hepatic glucose sensing is required to preserve β cell glucose competence. Ozer EA, Aksu N, Uclar E, Erdogan H, Bakiler AR, Tsuda M, et al. No mutation in the SLC2A2 GLUT2 gene in a Turkish infant with Fanconi—Bickel syndrome.

Pediatr Nephrol. Stanescu DE, Hughes N, Kaplan B, Stanley CA, De León DD. Novel presentations of congenital hyperinsulinism due to mutations in the MODY genes: HNF1A and HNF4A.

J Clin Endocrinol Metab. Stanik J, Skopkova M, Brennerova K, Danis D, Rosolankova M, Salingova A, et al. Congenital hyperinsulinism and glycogenosis-like phenotype due to a novel HNF4A mutation. Diabetes Res Clin Pract. Ross KM, Ferrecchia IA, Dahlberg KR, Dambska M, Ryan PT, Weinstein DA.

Dietary management of the glycogen storage diseases: evolution of treatment and ongoing controversies. Adv Nutr. Hoogeveen IJ, van der Ende RM, van Spronsen FJ, de Boer F, Heiner-Fokkema MR, Derks TG.

Normoglycemic ketonemia as biochemical presentation in ketotic glycogen storage disease. Pennisi A, Maranda B, Benoist JF, Baudouin V, Rigal O, Pichard S, et al. Nocturnal enteral nutrition is therapeutic for growth failure in Fanconi—Bickel syndrome.

Szymańska E, Lipiński P, Rokicki D, Książyk J, Tylki-Szymańska A. Over year follow-up of patients with hepatic glycogen storage diseases: single-center experience. Diagnostics Basel. Willems PJ, Gerver WJ, Berger R, Fernandes J.

JM conceptualized the research, gathered and analyzed the data, and wrote the initial manuscript. DV helped with the data analyses and draft of the initial manuscript. BT reviewed the neuroimaging scans. MC coordinated the study, and revised and conducted critical reviews of the manuscript for key intellectual content.

MS helped with the data analyses. All authors contributed to the article and approved the submitted version. This study was funded in part by the Coordenação de Aperfeiçoamento de Pessoal de Nivel Superior-Brazil CAPES conferred to JM and DV Finance Code The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The authors are grateful to the families and children who participated in the study. Kanungo S, Wells K, Tribett T, El-Gharbawy A. Glycogen Metabolism and Glycogen Storage Disorders.

Ann Transl Med —4. doi: PubMed Abstract CrossRef Full Text Google Scholar. Rich LR, Harris W, Brown AM. The Role of Brain Glycogen in Supporting Physiological Function. Front Neurosci Wolfsdorf JI, Weinstein DA. Glycogen Storage Diseases. Rev Endocr Metab Disord 4 1 — Aydemir Y, Gürakan F, Saltık Temizel İN, Demir H, Oğuz KK, Yalnızoğlu D, et al.

Evaluation of Central Nervous System in Patients With Glycogen Storage Disease Type 1a. Turk J Pediatr —8. Beyzaei Z, Geramizadeh B, Karimzadeh S.

Diagnosis of Hepatic Glycogen Storage Disease Patients With Overlapping Clinical Symptoms by Massively Parallel Sequencing: A Systematic Review of Literature. Orphanet J Rare Dis — Rake JP, Visser G, Labrune P, Leonard JV, Ullrich K, Smit GPA.

Glycogen Storage Disease Type I: Diagnosis, Management, Clinical Course and Outcome. Results of the European Study on Glycogen Storage Disease Type I ESGSD I. Eur J Pediatr Suppl — CrossRef Full Text Google Scholar. Ellingwood SS, Cheng A. Biochemical and Clinical Aspects of Glycogen Storage Diseases.

J Endocrinol R— Bali GS, Chen YT, Austin S. Glycogen Storage Disease: Type I. In: Adam MP, Ardinger HH, Pagon RA, editors. Seattle, Washington: University of Washington — Google Scholar. Beauchamp NJ, Dalton A, Ramaswami U, Niinikoski H, Mention K, Kenny P, et al.

Glycogen Storage Disease Type IX: High Variability in Clinical Phenotype. Mol Genet Metab — Herbert M, Goldstein JL, Rehder C, Austin S, Kishnani PS, Bali DS. Phosphorylase Kinase Deficiency Summary Genetic Counseling. Achouitar S, Goldstein JL, Mohamed M, Austin S, Boyette K, Blanpain FM, et al.

Common Mutation in the PHKA2 Gene With Variable Phenotype in Patients With Liver Phosphorylase B Kinase Deficiency.

Mol Genet Metab —4. Neil WP, Hemmen TM. Neurologic Manifestations of Hypoglycemia. In: Rigobelo E, editor. Diabetes - Damages and Treatments. Rijeka: InTech McNay EC, Williamson A, McCrimmon RJ, Sherwin RS. Cognitive and Neural Hippocampal Effects of Long-Term Moderate Recurrent Hypoglycemia.

Diabetes — Ho MS, Weller NJ, Ives FJ, Carne CL, Murray K, vanden Driesen RI, et al. Prevalence of Structural Central Nervous System Abnormalities in Early-Onset Type 1 Diabetes Mellitus. J Pediatr — Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, et al.

Standards and Guidelines for the Interpretation of Sequence Variants: A Joint Consensus Recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med — Froissart R, Piraud M, Boudjemline AM, Vianey-saban C, Petit F, Hubert-buron A, et al.

Glucosephosphatase Deficiency. Orphanet J Rare Dis Rake JP, ten Berge AM, Visser G, Verlind E, Niezen-Koning KE, Buys CHCM, et al. Glycogen Storage Disease Type Ia: Recent Experience With Mutation Analysis, a Summary of Mutations Reported in the Literature and a Newly Developed Diagnostic Flowchart.

Eur J Pediatr — Lei KJ, Chen YT, Chen H, Wong LJC, Liu JL, McConkie-Rosell A, et al. Genetic Basis of Glycogen Storage Disease Type 1a: Prevalent Mutations At the glucosephosphatase Locus. Am J Hum Genet — PubMed Abstract Google Scholar.

Chevalier-Porst F, Bozon D, Bonardot AM, Bruni N, Mithieux G, Mathieu M, et al. Mutation Analysis in 24 French Patients With Glycogen Storage Disease Type La. J Med Genet — Reis F de C, Caldas HC, Norato DYJ, Schwartz IVD, Giugliani R, Burin MG, et al.

Glycogen Storage Disease Type Ia: Molecular Study in Brazilian Patients. J Hum Genet —9. Carlin MP, Scherrer DZ, De Tommaso AMA, Bertuzzo CS, Steiner CE.

Determining Mutations in G6PC and SLC37A4 Genes in a Sample of Brazilian Patients With Glycogen Storage Disease Types Ia and Ib. Genet Mol Biol —6. Akanuma J, Nishigaki T, Fujii K, Matsubara Y, Inui K, Takahashi K, et al.

Glycogen Storage Disease Type Ia: Molecular Diagnosis of 51 Japanese Patients and Characterization of Splicing Mutations by Analysis of Ectopically Transcribed mRNA From Lymphoblastoid Cells.

Am J Med Genet — Burwinkel B, Maichele AJ, Aagenaes Ø, Bakker HD, Lerner A, Shin YS, et al. Autosomal Glycogenosis of Liver and Muscle Due to Phosphorylase Kinase Deficiency is Caused by Mutations in the Phosphorylase Kinase β Subunit PHKB.

Hum Mol Genet — De Angelis LC, Brigati G, Polleri G, Malova M, Parodi A, Minghetti D, et al. GSD9c results from impaired gamma unit of phosphorylase kinase enzyme function in liver and testis, with early childhood presentations of recurrent hypoglycemia, hepatomegaly progressing to liver cirrhosis and end stage liver disease; apart from motor delay, hepatosplenomegaly, renal tubular damage and muscle weakness Because the gamma subunit contains the catalytic site of the enzyme, GSD9C typically has a more severe phenotype.

In personal experience SK unpublished data , a teenager presented with seizure disorder, microcephaly, intellectual disability, short stature apart from clinical presentations noted above. Apart from clinical presentation noted above, elevated liver enzymes and lactate with severe fasting ketosis in setting of normal triglycerides creatine kinase CK and uric acid can be seen.

Measurement of enzyme activity in liver may help but diagnostic confirmation includes molecular analysis of PHKG2 gene Nutritional approaches to prevent recurrent hypoglycemia with high-protein and complex carbohydrates and symptomatic treatment and management for any hypoglycemia related complications may be helpful.

In personal experience SK unpublished data , teenage presentation of a GSD9c case with liver failure was treated successfully with liver transplantation and at 3-year post transplantation, showed improvement in cognitive abilities in late adolescence, to secure successful vocational training and employment, improved muscle strength, resolution of hepatosplenomegaly and seizures.

GSD9C is an autosomal recessive disorder caused by mutations of the PHKG2 gene which encodes the gamma subunit of phosphorylase kinase on chromosome 16p GSD3, also known as Cori Disease or Forbes disease results from glycogen debrancher enzyme GDE deficiency with impaired glycogen breakdown and abnormal glycogen accumulation, affecting liver, skeletal and cardiac muscles 35 , Muscular symptoms become apparent during and after adolescence though hypertrophic cardiomyopathy seen in younger childhood 43 , As noted above, characteristic findings include fasting hypoglycemia with ketosis, hyperlipidemia, elevated CK, an inverse relationship between a patients age and liver enzymes, lack of lactic acidosis and hyperuricemia 35 , 43 - A diagnosis can be made by mutation analysis of the AGL gene 46 or liver biopsy to detect the enzymatic defect.

Best approaches are nutritional with frequent meals, with high protein content and lesser amounts of UCCS than in GSD1 , or bedtime glycosade helps growth during adolescence Though there are suggestions that a modified Atkins diet improves myopathy symptoms GSD3 has autosomal recessive inheritance, with 58 different reported mutations in the AGL gene on chromosome 1p GSD4, also known as Andersen disease or Brancher deficiency, is a glycogenolysis defect with impaired and few α-1,6-glycosidic bonds along glycogen chain, resulting in abnormal glycogen with limited branch points limited dextran similar to amylopectin or polyglucosan Clinical presentation is variable and historically classified as two hepatic and four neuromuscular forms based upon age of onset and severity More recent studies suggest that GSD4 phenotypes should be considered a continuum of disease as opposed to discreet subtypes 49 , The classical hepatic GSD4 typically presents within 18 months of birth with patients having a failure to thrive, hepatosplenomegaly, and liver cirrhosis As the disease progresses, liver failure ultimately results, leading to death by the age of 5 unless a liver transplant is performed A non-progressive hepatic form with a similar presentation has also been described 52 , Neuromuscular variants range in onset from in utero presenting perinatally as fetal akinesia deformation sequence FADS to adulthood as adult polyglucosan body disease APBD with wide severity range from perinatal death to mild symptoms Commonly seen features of the neuromuscular variant of disease includes: hypotonia, muscle atrophy, myopathy, cardiomyopathy, central nervous system CNS , and peripheral nerve system PNS dysfunction Liver dysfunction with abnormal coagulation can be non-specific findings and amylopectin like material deposition can be seen in liver, heart, muscle, brain, spinal cord or reduced glycogen branching enzyme GBE activity seen in liver, muscle or leukocyte; but confirmation made by molecular analysis of GBE1 gene Unlike other liver GSDs, GSD4 has no specific treatment.

Early liver transplant is indicated in patients with the classical hepatic form but only in absence of cardiac or CNS disease GSD4 is rare and has autosomal recessive inheritance with mutations in the GBE1 gene on chromosome 3p GSD9B, also known as phosphorylase kinase deficiency of liver and muscle, have predominant hepatomegaly, short stature seen in early childhood and, sometimes in addition, muscle weakness and hypotonia Can be asymptomatic, but hypoglycemia and reduced enzyme activity can be seen.

Diagnosis is mainly confirmed by mutation analysis of the PHKB gene. Symptomatic with prevention of hypoglycemia with hi-protein and complex carbohydrate diet; though there is no specific treatment for muscle disease GSD9B is an autosomal recessive disorder caused by mutations of β subunit of PHKB gene on chromosome 16q GSD0b, also known as muscle glycogen synthase deficiency, is rare and seems to affect muscle mitochondrial structure and function apart from depleted glycogen Known symptoms include muscle fatigue, exercise intolerance, recurrent exertional syncope, hypertrophic cardiomyopathy, sudden cardiac death without cardiomyopathy 55 - Clinical suspicion with molecular analysis of GYS1 gene provides diagnostic confirmation.

Muscle biopsy can show depleted glycogen; oxidative fibers and abnormal mitochondria 55 - No specific treatment, preventive measures, supportive therapy with high protein complex carbohydrates diet may help. GSD0b is an autosomal recessive disorder caused by mutations of GSY1 gene on chromosome 19q GSD2, also known as Pompe disease or acid maltase deficiency results from impaired lysosomal acid-α-glucosidase GAA function and accumulation of lysosomal glycogen in skeletal, respiratory and cardiac muscle and often considered as lysosomal storage disorder LSD than GSD.

A non-classical infantile form shows slower symptom progression, is less severe with no cardiomyopathy Late-onset Pompe disease childhood, juvenile, and adult forms is often used to describe patients who present after the first year of life with muscle weakness, and hypotonia Clinical suspicion as noted above with characteristic evidence of hypertrophic cardiomyopathy with EKG findings of shortened PR interval and high QRS complexes and elevated CK is seen in the infantile-onset form.

While, proximal myopathy with diaphragmatic weakness is seen in late-onset disease. Elevated blood aminotransferases and CK are common but diagnostic confirmation noted with deficient GAA enzyme activity in lymphocytes, fibroblasts, and muscle or molecular analysis of biallelic GAA gene GAA activity is usually absent in infantile-onset disease or decreased in late-onset disease.

Some genotype—phenotype correlations exist and determined by the type of the mutation 62 , Dried blood spot testing measuring GAA enzyme activity has helped GSD2 to be included in Newborn Screening Evaluation of the CRIM status is important, since CRIM negative status is associated with poor response to ERT and poor prognosis, if immunomodulation is not started early Results of newborn screening in Taiwan demonstrated significant long-term benefits from the early identification and treatment of patients with infantile Pompe disease before symptoms appeared making an argument for its inclusion in newborn screening panels in many states in the U.

and trialed in several countries 65 , Enzyme replacement therapy ERT using human recombinant acid α-glucosidase, the only approved treatment in the US and Europe since , is based on its ability to degrade accumulated lysosomal glycogen and improve cardiac and skeletal muscle function Though, a negative cross reacting immune material CRIM -negative status has high anti-rhGAA IgG antibodies development and resultant reduced ERT therapeutic effect with poor outcomes if not treated early with immunosuppression 65 , If early diagnosis of late-onset disease is made via newborn screening, the question of when to start treatment in an asymptomatic patient is debated.

Improvement in pulmonary function is seen in symptomatic patients with late-onset disease GSD2 is a pan-ethnic autosomal recessive disorder caused by mutations of the GAA gene on chromosome 17q The increasing list of GAA gene pathogenic mutations can be found at www.

The estimated prevalence is considered to be 1 in 5, GSD5, also known as McArdle disease results from deficient muscle phosphorylase activity and results in impaired glycogenolysis leading to exercise intolerance, muscle weakness and cramping alleviated by rest, and exercise induced rhabdomyolysis.

A common history of childhood onset exercise intolerance and a wide range of severity and age of onset reported with most serious complication being renal failure from myoglobinuria and rhabdomyolysis. Apart from clinical suspicion, elevated CK, myoglobinuria and renal dysfunction as common biochemical markers with additional non-invasive diagnostic confirmation with molecular analysis of PYGM gene is indicated.

Invasive muscle biopsy with negative muscle phosphorylase activity can help diagnosis too. Oral sucrose loading 30—40 minutes before exercise helps exercise tolerance as exogenous fuel source to help energy gap with lack of endogenous glucose from glycogenolysis and free fatty acids availability until ~10 minutes into exercising Regular exercise of moderate intensity helps maximize circulatory capacity and increase fuel delivery to muscles GSD5 is an autosomal recessive disorder caused by mutations of PYGM gene on chromosome 11q GSD7, also known as Tarui disease results from deficient muscle subunit of phosphofructokinase PFK enzyme as a rate limiting factor, with resultant impaired glycogenolysis and glycolysis.

The classical form is characterized by exercise intolerance, often with rhabdomyolysis , muscle cramps and pain. In some cases jaundice accompanied by increased serum bilirubin, exercise related elevated CK levels, myoglobinuria and myogenic hyperuricemia may also be seen 72 ,

Depending Electrolyte balance benefits the type of GSD a glycohen has, glycogen may build up in the liver, Successful weight loss the diseass, or both. GSD can eRsearch affect blood Appetite control planner, the Research on glycogen storage disease, kidneys, and other organs. Normally, glycogen is stored in the liver until the body needs energy. Then, enzymes convert glycogen into glucose so that it can travel through the bloodstream to cells that need fuel. Every cell in the body contains enzymes, but children with GSD lack one of the enzymes responsible for making glycogen or converting glycogen to glucose. GSD is a rare condition. Research on glycogen storage disease

Author: Yotaxe

1 thoughts on “Research on glycogen storage disease

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com