Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
J Inherit Metab Dis ; 44(1): 226-239, 2021 01.
Article in English | MEDLINE | ID: mdl-33448466

ABSTRACT

Glycogen storage disease type IIIa (GSDIIIa) is an inborn error of carbohydrate metabolism caused by a debranching enzyme deficiency. A subgroup of GSDIIIa patients develops severe myopathy. The purpose of this study was to investigate whether acute nutritional ketosis (ANK) in response to ketone-ester (KE) ingestion is effective to deliver oxidative substrate to exercising muscle in GSDIIIa patients. This was an investigator-initiated, researcher-blinded, randomized, crossover study in six adult GSDIIIa patients. Prior to exercise subjects ingested a carbohydrate drink (~66 g, CHO) or a ketone-ester (395 mg/kg, KE) + carbohydrate drink (30 g, KE + CHO). Subjects performed 15-minute cycling exercise on an upright ergometer followed by 10-minute supine cycling in a magnetic resonance (MR) scanner at two submaximal workloads (30% and 60% of individual maximum, respectively). Blood metabolites, indirect calorimetry data, and in vivo 31 P-MR spectra from quadriceps muscle were collected during exercise. KE + CHO induced ANK in all six subjects with median peak ßHB concentration of 2.6 mmol/L (range: 1.6-3.1). Subjects remained normoglycemic in both study arms, but delta glucose concentration was 2-fold lower in the KE + CHO arm. The respiratory exchange ratio did not increase in the KE + CHO arm when workload was doubled in subjects with overt myopathy. In vivo 31 P MR spectra showed a favorable change in quadriceps energetic state during exercise in the KE + CHO arm compared to CHO in subjects with overt myopathy. Effects of ANK during exercise are phenotype-specific in adult GSDIIIa patients. ANK presents a promising therapy in GSDIIIa patients with a severe myopathic phenotype. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov identifier: NCT03011203.


Subject(s)
Beverages , Exercise , Glycogen Storage Disease Type III/diet therapy , Ketosis/chemically induced , Muscular Diseases/diet therapy , Adult , Blood Glucose/analysis , Carbohydrate Metabolism , Cross-Over Studies , Diet, Ketogenic , Dietary Carbohydrates , Esters/administration & dosage , Female , Glycogen Storage Disease Type III/metabolism , Humans , Ketones/administration & dosage , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Netherlands , Phenotype
2.
Ann Nutr Metab ; 76(4): 233-241, 2020.
Article in English | MEDLINE | ID: mdl-32712609

ABSTRACT

INTRODUCTION: Glycogen storage disease Type III (GSD III) is an autosomal recessive disease caused by the deficiency of glycogen debranching enzyme, encoded by the AGL gene. Two clinical types of the disease are most prevalent: GSD IIIa involves the liver and muscle, whereas IIIb affects only the liver. The classical dietetic management of GSD IIIa involves prevention of fasting, frequent feeds with high complex carbohydrates in small children, and a low-carb-high-protein diet in older children and adults. Recently, diets containing high amount of fat, including ketogenic and modified Atkins diet (MAD), have been suggested to have favorable outcome in GSD IIIa. METHODS: Six patients, aged 3-31 years, with GSD IIIa received MAD for a duration of 3-7 months. Serum glucose, transaminases, creatine kinase (CK) levels, capillary ketone levels, and cardiac parameters were followed-up. RESULTS: In all patients, transaminase levels dropped in response to MAD. Decrease in CK levels were detected in 5 out of 6 patients. Hypoglycemia was evident in 2 patients but was resolved by adding uncooked cornstarch to diet. CONCLUSION: Our study demonstrates that GSD IIIa may benefit from MAD both clinically and biochemically.


Subject(s)
Diet, High-Protein Low-Carbohydrate/methods , Glycogen Storage Disease Type III/diet therapy , Adolescent , Adult , Blood Glucose/analysis , Child , Child, Preschool , Female , Glycogen Storage Disease Type III/blood , Humans , Male , Transaminases/blood , Treatment Outcome , Young Adult
3.
J Inherit Metab Dis ; 43(4): 770-777, 2020 07.
Article in English | MEDLINE | ID: mdl-32064649

ABSTRACT

A potential role of dietary lipids in the management of hepatic glycogen storage diseases (GSDs) has been proposed, but no consensus on management guidelines exists. The aim of this study was to describe current experiences with dietary lipid manipulations in hepatic GSD patients. An international study was set up to identify published and unpublished cases describing hepatic GSD patients with a dietary lipid manipulation. A literature search was performed according to the Cochrane Collaboration methodology through PubMed and EMBASE (up to December 2018). All delegates who attended the dietetics session at the IGSD2017, Groningen were invited to share unpublished cases. Due to multiple biases, only data on GSDIII were presented. A total of 28 cases with GSDIII and a dietary lipid manipulation were identified. Main indications were cardiomyopathy and/or myopathy. A high fat diet was the most common dietary lipid manipulation. A decline in creatine kinase concentrations (n = 19, P < .001) and a decrease in cardiac hypertrophy in paediatric GSDIIIa patients (n = 7, P < .01) were observed after the introduction with a high fat diet. This study presents an international cohort of GSDIII patients with different dietary lipid manipulations. High fat diet may be beneficial in paediatric GSDIIIa patients with cardiac hypertrophy, but careful long-term monitoring for potential complications is warranted, such as growth restriction, liver inflammation, and hepatocellular carcinoma development.


Subject(s)
Cardiomyopathies/etiology , Dietary Fats , Glycogen Storage Disease Type III/diet therapy , Cardiomyopathies/physiopathology , Child , Glycogen Storage Disease Type III/complications , Humans , Liver/pathology , Monitoring, Physiologic , Triglycerides/blood
4.
Acta Myol ; 38(1): 17-20, 2019 03.
Article in English | MEDLINE | ID: mdl-31309177

ABSTRACT

Glycogen storage disease type III (GSDIII) management in adult patients includes a high-protein diet with cornstarch supplementation to maintain a normal level of glucose in the blood. This regimen can prevent hypoglycaemia but does not seem to improve skeletal muscle and heart function. A 34 years-old patient with GSD IIIa with hypertrophic cardiomyopathy was then treated with a modified Atkins ketogenic diet. After 12 months of treatment ejection fraction raised from 30 to 45%, liver enzymes were reduced and CK plasma level dropped from 568 to 327 U/l. Physical activity increased from about 1300 to 2800 steps per day and health-related quality of life assessment ameliorated. An increase in uric acid triglycerides plasma level was observed. This data obtained in an adult patient confirm previous reports evidencing the effectiveness of ketogenic diets in improving cardiac and muscular manifestations in children with GSDIII.


Subject(s)
Diet, High-Protein Low-Carbohydrate , Diet, Ketogenic , Glycogen Storage Disease Type III/diet therapy , Adult , Humans , Male , Quality of Life
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3407-3417, 2018 10.
Article in English | MEDLINE | ID: mdl-30076962

ABSTRACT

Glycogen disease type III (GSDIII), a rare incurable autosomal recessive disorder due to glycogen debranching enzyme deficiency, presents with liver, heart and skeletal muscle impairment, hepatomegaly and ketotic hypoglycemia. Muscle weakness usually worsens to fixed myopathy and cardiac involvement may present in about half of the patients during disease. Management relies on careful follow-up of symptoms and diet. No common agreement was reached on sugar restriction and treatment in adulthood. We administered two dietary regimens differing in their protein and carbohydrate content, high-protein (HPD) and high-protein/glucose-free (GFD), to our mouse model of GSDIII, starting at one month of age. Mice were monitored, either by histological, biochemical and molecular analysis and motor functional tests, until 10 months of age. GFD ameliorated muscle performance up to 10 months of age, while HPD showed little improvement only in young mice. In GFD mice, a decreased muscle glycogen content and fiber vacuolization was observed, even in aged animals indicating a protective role of proteins against skeletal muscle degeneration, at least in some districts. Hepatomegaly was reduced by about 20%. Moreover, the long-term administration of GFD did not worsen serum parameters even after eight months of high-protein diet. A decreased phosphofructokinase and pyruvate kinase activities and an increased expression of Krebs cycle and gluconeogenesis genes were seen in the liver of GFD fed mice. Our data show that the concurrent use of proteins and a strictly controlled glucose supply could reduce muscle wasting, and indicate a better metabolic control in mice with a glucose-free/high-protein diet.


Subject(s)
Diet, High-Protein/methods , Glycogen Storage Disease Type III/diet therapy , Hepatomegaly/diet therapy , Muscle, Skeletal/physiopathology , Animals , Citric Acid Cycle , Diet, High-Protein Low-Carbohydrate/methods , Disease Models, Animal , Female , Glycogen Storage Disease Type III/metabolism , Glycogen Storage Disease Type III/physiopathology , Hepatomegaly/metabolism , Male , Mice , Mice, Knockout , Muscle, Skeletal/drug effects , Phosphofructokinases/metabolism , Physical Conditioning, Animal , Pyruvate Kinase/metabolism , Treatment Outcome
6.
Curr Opin Clin Nutr Metab Care ; 18(4): 415-21, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26001652

ABSTRACT

PURPOSE OF REVIEW: Glycogen storage disorders (GSDs) are inborn errors of metabolism with abnormal storage or utilization of glycogen. The present review focuses on recent advances in hepatic GSD types I, III and VI/IX, with emphasis on clinical aspects and treatment. RECENT FINDINGS: Evidence accumulates that poor metabolic control is a risk factor for the development of long-term complications, such as liver adenomas, low bone density/osteoporosis, and kidney disease in GSD I. However, mechanisms leading to these complications remain poorly understood and are being investigated. Molecular causes underlying neutropenia and neutrophil dysfunction in GSD I have been elucidated. Case series provide new insights into the natural course and outcome of GSD types VI and IX. For GSD III, a high protein/fat diet has been reported to improve (cardio)myopathy, but the beneficial effect of this dietary concept on muscle and liver disease manifestations needs to be further established in prospective studies. SUMMARY: Although further knowledge has been gained regarding pathophysiology, disease course, treatment, and complications of hepatic GSDs, more controlled prospective studies are needed to assess effects of different dietary and medical treatment options on long-term outcome and quality of life.


Subject(s)
Glycogen Storage Disease Type III/physiopathology , Glycogen Storage Disease Type I/physiopathology , Glycogen Storage Disease Type VI/physiopathology , Liver/physiopathology , Animals , Cardiomyopathies/complications , Cardiomyopathies/diet therapy , Cardiomyopathies/physiopathology , Diet, Carbohydrate-Restricted , Diet, High-Fat , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Dietary Proteins/administration & dosage , Disease Models, Animal , Glycogen/metabolism , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/diagnosis , Glycogen Storage Disease Type I/diet therapy , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/diagnosis , Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type VI/complications , Glycogen Storage Disease Type VI/diagnosis , Glycogen Storage Disease Type VI/diet therapy , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/diet therapy , Liver Cirrhosis/physiopathology
7.
Neurology ; 84(17): 1767-71, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25832663

ABSTRACT

OBJECTIVE: Glycogen storage disease type IIIa (GSDIIIa) is classically regarded as a glycogenosis with fixed weakness, but we hypothesized that exercise intolerance in GSDIIIa is related to muscle energy failure and that oral fructose ingestion could improve exercise tolerance in this metabolic myopathy. METHODS: We challenged metabolism with cycle-ergometer exercise and measured substrate turnover and oxidation rates using stable isotope methodology and indirect calorimetry in 3 patients and 6 age-matched controls on 1 day, and examined the effect of fructose ingestion on exercise tolerance in the patients on another day. RESULTS: Total fatty acid oxidation rates during exercise were higher in patients than controls, 32.1 (SE 1.2) vs 20.7 (SE 0.5; range 15.8-29.3) µmol/kg/min (p = 0.048), and oxidation of carbohydrates was lower in patients, 1.0 (SE 5.4) vs 38.4 (SE 8.0; range 23.0-77.1) µmol/kg/min (p = 0.024). Fructose ingestion improved exercise tolerance in the patients. CONCLUSION: Similar to patients with McArdle disease, in whom muscle glycogenolysis is also impaired, GSDIIIa is associated with a reduced skeletal muscle oxidation of carbohydrates and a compensatory increase in fatty acid oxidation, and fructose ingestion improves exercise tolerance. Our results indicate that GSDIIIa should not only be viewed as a glycogenosis with fixed skeletal muscle weakness, but should also be considered among the glycogenoses presenting with exercise-related dynamic symptoms caused by muscular energy deficiency. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that ingestion of fructose improves exercise tolerance in patients with GSDIIIa.


Subject(s)
Energy Metabolism/physiology , Exercise , Fructose/pharmacology , Glycogen Storage Disease Type III/metabolism , Muscle Weakness/metabolism , Muscle, Skeletal/metabolism , Adolescent , Adult , Fructose/administration & dosage , Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type III/physiopathology , Humans , Muscle Weakness/diet therapy , Muscle, Skeletal/physiopathology , Young Adult
8.
J Inherit Metab Dis ; 38(3): 545-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25164784

ABSTRACT

In childhood, GSD type III causes relatively severe fasting intolerance, classically associated with ketotic hypoglycaemia. During follow up, history of (documented) hypoglycaemia, clinical parameters (growth, liver size, motor development, neuromuscular parameters), laboratory parameters (glucose, lactate, ALAT, cholesterol, triglycerides, creatine kinase and ketones) and cardiac parameters all need to be integrated in order to titrate dietary management, for which age-dependent requirements need to be taken into account. Evidence from case studies and small cohort studies in both children and adults with GSD III demonstrate that prevention of hypoglycaemia and maintenance of euglycemia is not sufficient to prevent complications. Moreover, over-treatment with carbohydrates may even be harmful. The ageing cohort of GSD III patients, including the non-traditional clinical presentations in adulthood, raises ‬‬‬new questions.


Subject(s)
Dietary Carbohydrates/therapeutic use , Dietary Proteins/therapeutic use , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/diet therapy , Hypoglycemia/prevention & control , Fasting , Humans
9.
J Pediatr Endocrinol Metab ; 28(1-2): 195-200, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25153581

ABSTRACT

AIM: To determine the individual fasting tolerance for patients with glycogen storage disease type III (GSD III) and to assess their linear growth velocity after tailoring of dose intervals of oral uncooked cornstarch. PATIENTS AND METHODS: A prospective cohort study included 32 patients with GSD III aged 6 months-11.5 years (median: 3.3 years). The fasting tolerance of each patient was determined as the time interval between starch administration until the drop in blood glucose level was below 60 mg/dL. RESULTS: Some 27 patients (84.4%) developed hypoglycemia. The intervals between oral cornstarch administration were tailored for each child according to his/her individual fasting tolerance. After a 6-month follow up there was a significant reduction in seizure attacks (p<0.01) and liver size (p<0.01), but there was no statistically significant difference in liver transaminase and serum lactate levels. There was a significant improvement in height (p<0.01) and linear growth velocity (p<0.05) of these patients after at least a 12-month follow up. CONCLUSION: Adjusting the intervals between the cornstarch doses for each patient with GSD III, according to individual fasting tolerance test was very beneficial and resulted in improvement of the linear growth velocity and reduction in the frequency of hypoglycemic seizures as well as the size of the liver. Individual scheduling of cornstarch doses prevents complications in those who develop hypoglycemia at short intervals; it also allows some relaxation in schedule for those who can tolerate longer fasting hours to improve their appetite and prolong their uninterrupted sleep hours.


Subject(s)
Body Height/drug effects , Glycogen Storage Disease Type III/blood , Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type III/diagnosis , Starch/administration & dosage , Blood Glucose/drug effects , Blood Glucose/metabolism , Child , Child Development/drug effects , Child, Preschool , Fasting/blood , Female , Follow-Up Studies , Glycogen Storage Disease Type III/physiopathology , Humans , Hypoglycemia/blood , Hypoglycemia/epidemiology , Infant , Male , Starch/pharmacology
10.
Orphanet J Rare Dis ; 9: 196, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25431232

ABSTRACT

BACKGROUND: Frequent feeds with carbohydrate-rich meals or continuous enteral feeding has been the therapy of choice in glycogen storage disease (Glycogenosis) type III. Recent guidelines on diagnosis and management recommend frequent feedings with high complex carbohydrates or cornstarch avoiding fasting in children, while in adults a low-carb-high-protein-diet is recommended. While this regimen can prevent hypoglycaemia in children it does not improve skeletal and heart muscle function, which are compromised in patients with glycogenosis IIIa. Administration of carbohydrates may elicit reactive hyperinsulinism, resulting in suppression of lipolysis, ketogenesis, gluconeogenesis, and activation of glycogen synthesis. Thus, heart and skeletal muscle are depleted of energy substrates. Modified Atkins diet leads to increased blood levels of ketone bodies and fatty acids. We hypothesize that this health care intervention improves the energetic balance of muscles. METHODS: We treated 2 boys with glycogenosis IIIa aged 9 and 11 years with a modified Atkins diet (10 g carbohydrate per day, protein and fatty acids ad libitum) over a period of 32 and 26 months, respectively. RESULTS: In both patients, creatine kinase levels in blood dropped in response to Atkins diet. When diet was withdrawn in one of the patients he complained of chest pain, reduced physical strength and creatine kinase levels rapidly increased. This was reversed when Atkins diet was reintroduced. One patient suffered from severe cardiomyopathy which significantly improved under diet. Patients with glycogenosis IIIa benefit from an improved energetic state of heart and skeletal muscle by introduction of Atkins diet both on a biochemical and clinical level. Apart from transient hypoglycaemia no serious adverse effects were observed.


Subject(s)
Diet, Carbohydrate-Restricted/methods , Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type III/diagnosis , Muscular Diseases/diet therapy , Muscular Diseases/diagnosis , Blood Glucose/metabolism , Child , Dietary Proteins/administration & dosage , Glycogen Storage Disease Type III/blood , Humans , Male , Muscular Diseases/blood
13.
J Inherit Metab Dis ; 32 Suppl 1: S103-6, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19322675

ABSTRACT

Glycogen storage disease type III (GSD III) is caused by a deficiency in debranching enzyme, which leads to an accumulation of abnormal glycogen called limit dextrin in affected tissues. Muscle and liver involvement is present in GSD type IIIa, while the defect is limited to the liver only in GSD type IIIb. Besides skeletal muscle involvement, a cardiomyopathy resembling idiopathic hypertrophic cardiomyopathy is seen. Management consists of maintaining normoglycaemia by supplementation with cornstarch therapy and/or protein. While studies are lacking regarding the best treatment for skeletal muscle disease, a high-protein diet was previously reported to be beneficial. No cases of improvement in cardiomyopathy have been reported. Our patient presented in infancy with hypoglycaemia and hepatomegaly. His prescribed management consisted of cornstarch supplementation and a high-protein diet providing 20% of his total energy needs. At 16 years of age, he developed a severe cardiomyopathy with a left ventricular mass index of 209 g/m(2). The cardiomyopathy remained stable on a protein intake of 20-25% of total energy. At age 22 years, the diet was changed to increase his protein intake to 30% of total energy and minimize his cornstarch therapy to only what was required to maintain normoglycaemia. Dramatic improvement in the cardiomyopathy occurred. Over one year, his left ventricular mass index decreased from 159.7 g/m(2) to 78 g/m(2) (normal 50-86 g/m(2)) and the creatine kinase levels decreased from 455 U/L to 282 U/L. Avoidance of overtreatment with carbohydrate and a high-protein diet can reverse and may prevent cardiomyopathy.


Subject(s)
Cardiomyopathies/diet therapy , Cardiomyopathies/etiology , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/diet therapy , Cardiomyopathies/physiopathology , Dietary Proteins/administration & dosage , Glycogen Storage Disease Type III/physiopathology , Humans , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/pathology , Liver/pathology , Male , Starch/administration & dosage , Young Adult
14.
J Inherit Metab Dis ; 24(8): 873-4, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11916322

ABSTRACT

A 10-month-old girl with glycogen storage disease type III developed recurrent severe hypoglycaemia induced by infectious mononucleosis. Severe metabolic damage probably reflected a rapid breakdown of liver cells induced by the viral infection.


Subject(s)
Glycogen Storage Disease Type III/complications , Hypoglycemia/etiology , Infectious Mononucleosis/complications , Female , Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type III/physiopathology , Humans , Hypoglycemia/physiopathology , Infant , Infectious Mononucleosis/physiopathology , Lactic Acid/blood , Liver/physiopathology , Recurrence , Starch/administration & dosage
15.
Eur J Pediatr ; 158 Suppl 2: S43-8, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10603098

ABSTRACT

UNLABELLED: Glycogen storage disease (GSD) types I, III, and IV can be associated with severe liver disease. The possible development of hepatocellular carcinoma and/or hepatic failure make these GSDs potential candidates for liver transplantation. Early diagnosis and initiation of effective dietary therapy have dramatically improved the outcome of GSD type I by reducing the incidence of liver adenoma and renal insufficiency. Nine type I and 3 type III patients have received liver transplants because of poor metabolic control, multiple liver adenomas, or progressive liver failure. Metabolic abnormalities were corrected in all GSD type I and type III patients, while catch-up growth was reported only in two patients. Whether liver transplantation results in reversal and/or prevention of renal disease remains unclear. Neutropenia persisted in both GSDIb patients post liver transplantation necessitating continuous granulocyte colony stimulating factor treatment. Thirteen GSD type IV patients were liver transplanted because of progressive liver cirrhosis and failure. All but one patient have not had neuromuscular or cardiac complications during follow-up periods for as long as 13 years. Four have died within a week and 5 years after transplantation. Caution should be taken in selecting GSD type IV candidates for liver transplantation because of the variable phenotype, which may include life-limiting extrahepatic manifestations. It remains to be evaluated, whether a genotype-phenotype correlation exists for GSD type IV, which may aid in the decision making. CONCLUSION: Liver transplantation should be considered for patients with glycogen storage disease who have developed liver malignancy or hepatic failure, and for type IV patients with the classical and progressive hepatic form.


Subject(s)
Glycogen Storage Disease Type III/surgery , Glycogen Storage Disease Type IV/surgery , Glycogen Storage Disease Type I/surgery , Liver Diseases/surgery , Liver Transplantation , Adolescent , Adult , Child , Female , Glycogen Storage Disease Type I/diet therapy , Glycogen Storage Disease Type III/diet therapy , Humans , Liver Diseases/etiology , Male , Neutropenia/drug therapy , Prognosis
16.
Nutr Hosp ; 13(5): 228-32, 1998.
Article in Spanish | MEDLINE | ID: mdl-9830843

ABSTRACT

RATIONALE: Administering raw corn starch can maintain normoglycemia for long periods after being ingested, thus facilitating control in patients with type I and III glycogenosis. METHODS: The metabolic effects and the effects on the nutritional status of a treatment with fractionated administrations of raw starch are assessed in two patients with type I glycogenosis (ages 18 and 12 years) and one patient with type III glycogenosis (aged 13 years). In the first two cases the response was previously studied after administering a load of raw corn starch in a water suspension, in an amount similar to the estimated rate of endogenous glucose production during the fasting period (5 mg/kg/minute). RESULTS: The results of the overload of starch showed a normoglycemia and an absence of lactoacidosis between 4 and 6 hours after its ingestion. The three patients were given two doses of raw corn starch (2 g/kg/dose) at 1.00 and 5.00 hours during the night. After one year of treatment, all patients showed glycemia levels at 9.00 AM that were greater than 90 mg/dl and lactic acid levels that were lower than 2.4 mmol/l. Moreover, in two of the cases there was an increase in the growth rate. In all cases the amount of the hepatomegaly decreased as did the size of the hepatic adenomas that were present in two of the cases. CONCLUSIONS: In patients with type I and III glycogenosis, raw corn starch can balance the results of the nightly gastric glucose infusion, both with regard to the metabolic control and with regard to the growth.


Subject(s)
Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type I/diet therapy , Phytotherapy , Starch/therapeutic use , Zea mays/therapeutic use , Humans
18.
J Am Diet Assoc ; 93(12): 1423-30, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8245377

ABSTRACT

Hepatic glycogen storage diseases (GSD) are a group of rare genetic disorders in which glycogen cannot be metabolized to glucose in the liver because of one of a number of possible enzyme deficiencies along the glycogenolytic pathway. Patients with GSD are usually diagnosed in infancy or early childhood with hypoglycemia, hepatomegaly, poor physical growth, and a deranged biochemical profile. Dietary therapies have been devised to use the available alternative metabolic pathways to compensate for disturbed glycogenolysis in GSD I (glucose-6-phosphatase deficiency), GSD III (debrancher enzyme deficiency), GSD VI (phosphorylase deficiency, which is less common), GSD IX (phosphorylase kinase deficiency), and GSD IV (brancher enzyme deficiency). In GSD I, glucose-6-phosphate cannot be dephosphorylated to free glucose. Managing this condition entails overnight continuous gastric high-carbohydrate feedings; frequent daytime feedings with energy distributed as 65% carbohydrate, 10% to 15% protein, and 25% fat; and supplements of uncooked cornstarch. In GSD III, though glycogenolysis is impeded, gluconeogenesis is enhanced to help maintain endogenous glucose production. In contrast to treatment for GSD I, advocated treatment for GSD III comprises frequent high-protein feedings during the day and a high-protein snack at night; energy is distributed as 45% carbohydrate, 25% protein, and 30% fat. Patients with GSD IV, VI, and IX have benefited from high-protein diets similar to that recommended for patients with GSD III.


Subject(s)
Glycogen Storage Disease Type III/diet therapy , Glycogen Storage Disease Type I/diet therapy , Glycogen Storage Disease/diet therapy , Adolescent , Age Factors , Child , Child, Preschool , Dietary Proteins/administration & dosage , Dietary Proteins/therapeutic use , Enteral Nutrition , Glycogen Storage Disease/therapy , Glycogen Storage Disease Type I/therapy , Glycogen Storage Disease Type IV/diet therapy , Glycogen Storage Disease Type VI/diet therapy , Humans , Hypoglycemia/prevention & control , Infant , Infant, Newborn , Phosphorylase Kinase/deficiency , Starch/therapeutic use , Time Factors
20.
Arch Invest Med (Mex) ; 22(3-4): 285-8, 1991.
Article in English | MEDLINE | ID: mdl-1844114

ABSTRACT

An eleven year old boy was referred because of sudden loss of consciousness, muscular weakness, poor general health, severe hypoglycemia with seizures and hepatomegaly. Response to oral glucose and galactose increased blood lactic acid and glucose at different times. Fasting values of blood lactic was normal, but glucose was found at 33 mg/dl. Similar test made up two hours after feeding revealed hyperlactatemia (35-50 mg/dL) and hyperglycemia (129 mg/dL). Glucagon did not result in a rise of glucose at fasting or feeding. Hepatic glycogen content was found 15 gm/100 mg of tissue. The enzyme activities revealed a deficiency of the liver debranching enzyme while leukocytes had normal enzyme activity. Hepatic biopsy showed liver fibrosis. The present case had the clinical characteristics of severe form of glycogen storage disease. A low carbohydrate and high protein diet was indicated in order to increase the gluconeogenic precursors. Although debranching enzyme deficiency is almost always benign a high carbohydrate diet induced a more severe expression of the disease.


Subject(s)
Dietary Carbohydrates/administration & dosage , Dietary Proteins/administration & dosage , Glycogen Storage Disease Type III/diet therapy , Child , Dietary Carbohydrates/adverse effects , Fibrosis , Glycogen Debranching Enzyme System/analysis , Glycogen Debranching Enzyme System/deficiency , Glycogen Storage Disease Type III/complications , Glycogen Storage Disease Type III/enzymology , Growth Disorders/etiology , Humans , Hypoglycemia/etiology , Leukocytes/enzymology , Liver/enzymology , Liver/pathology , Liver Glycogen/metabolism , Male , Muscles/pathology , Seizures/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...