Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Publication year range
1.
Mol Genet Metab ; 129(1): 3-12, 2020 01.
Article in English | MEDLINE | ID: mdl-31787497

ABSTRACT

The glycogen storage diseases are a group of inherited metabolic disorders that are characterized by specific enzymatic defects involving the synthesis or degradation of glycogen. Each disorder presents with a set of symptoms that are due to the underlying enzyme deficiency and the particular tissues that are affected. Autophagy is a process by which cells degrade and recycle unneeded or damaged intracellular components such as lipids, glycogen, and damaged mitochondria. Recent studies showed that several of the glycogen storage disorders have abnormal autophagy which can disturb normal cellular metabolism and/or mitochondrial function. Here, we provide a clinical overview of the glycogen storage disorders, a brief description of autophagy, and the known links between specific glycogen storage disorders and autophagy.


Subject(s)
Autophagy , Glycogen Storage Disease/drug therapy , Glycogen Storage Disease/etiology , Glycogen/metabolism , Animals , Glycogen Storage Disease/pathology , Glycogen Storage Disease Type I/drug therapy , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/etiology , Glycogenolysis , Humans , Muscle, Skeletal/physiopathology
2.
J Inherit Metab Dis ; 41(6): 1015-1025, 2018 11.
Article in English | MEDLINE | ID: mdl-29967951

ABSTRACT

BACKGROUND AND AIMS: Glycogen storage disease type Ib (GSD1b) is a rare metabolic and immune disorder caused by a deficiency in the glucose-6-phosphate transporter (G6PT) and characterized by impaired glucose homeostasis, myeloid dysfunction, and long-term risk of hepatocellular adenomas. Despite maximal therapy, based on a strict diet and on granulocyte colony-stimulating factor treatment, long-term severe complications still develop. Understanding the pathophysiology of GSD1b is a prerequisite to develop new therapeutic strategies and depends on the availability of animal models. The G6PT-KO mouse mimics the human disease but is very fragile and rarely survives weaning. We generated a conditional G6PT-deficient mouse as an alternative model for studying the long-term pathophysiology of the disease. We utilized this conditional mouse to develop an inducible G6PT-KO model to allow temporally regulated G6PT deletion by the administration of tamoxifen (TM). METHODS: We generated a conditional G6PT-deficient mouse utilizing the CRElox strategy. Histology, histochemistry, and phenotype analyses were performed at different times after TM-induced G6PT inactivation. Neutrophils and monocytes were isolated and analyzed for functional activity with standard techniques. RESULTS: The G6PT-inducible KO mice display the expected disturbances of G6P metabolism and myeloid dysfunctions of the human disorder, even though with a milder intensity. CONCLUSIONS: TM-induced inactivation of G6PT in these mice leads to a phenotype which mimics that of human GSD1b patients. The conditional mice we have generated represent an excellent tool to study the tissue-specific role of the G6PT gene and the mechanism of long-term complications in GSD1b.


Subject(s)
Antiporters/deficiency , Disease Models, Animal , Glucose/metabolism , Glycogen Storage Disease Type I/genetics , Homeostasis , Monosaccharide Transport Proteins/deficiency , Animals , Antiporters/genetics , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/pathology , Mice , Mice, Knockout , Monosaccharide Transport Proteins/genetics , Neutropenia/etiology , Tamoxifen/administration & dosage
4.
Vestn Otorinolaringol ; (2): 93-4, 2014.
Article in Russian | MEDLINE | ID: mdl-24781183

ABSTRACT

This paper reports a clinical case that gives evidence of the possibility of cochlear implantation after liver transplantation. Patient K. aged 3 years 10 months was admitted to the Russian Research and Practical Centre of Audiology and Hearing Rehabilitation with the diagnosis of type IB glycogenosis after maternal liver transplantation associated with chronic neutropenia, chronic cutaneous and mucosal infection, partial symptomatic partial epilepsy, retarded psycho-motor development, and complaints of the absence of auditory response. The audiological examination provided materials for the diagnosis of grade IV bilateral sensorineural hearing loss tending toward deafness. Cochlear implantation recommended to the patient was performed on February 20, 2013 using the HiRes 90 K implant with the HiFocus Helix electrode (Advanced Bionics, USA). The surgical intervention and the postoperative period passed without complications. The speech processor was activated one month after surgery. The results of surdopedagogical testing gave evidence of successful rehabilitation promising the further improvement. It is concluded that immunosuppressive therapy is not an absolute contraindication for cochlear implantation, but this procedure requires detailed examination and thorough preparation for the forthcoming surgery.


Subject(s)
Cochlear Implantation/methods , Glycogen Storage Disease Type I/complications , Hearing Loss, Sensorineural/surgery , Immunocompromised Host , Child, Preschool , Cochlear Implantation/standards , Female , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/immunology , Hearing Loss, Sensorineural/immunology , Hearing Loss, Sensorineural/rehabilitation , Humans , Treatment Outcome
5.
Nat Genet ; 13(2): 203-9, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8640227

ABSTRACT

Glycogen storage disease type 1a (GSD-1a) is caused by a deficiency in microsomal glucose-6-phosphatase (G6Pase), the key enzyme in glucose homeostasis. A G6Pase knockout mouse which mimics the pathophysiology of human GSD-1a patients was created to understand the pathogenesis of this disorder, to delineate the mechanisms of G6Pase catalysis, and to develop future therapeutic approaches. By examining G6Pase in the liver and kidney, the primary gluconeogenic tissues, we demonstrate that glucose-6-P transport and hydrolysis are performed by separate proteins which are tightly coupled. We propose a modified translocase catalytic unit model for G6Pase catalysis.


Subject(s)
Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/enzymology , Glycogen Storage Disease Type I/etiology , Animals , Animals, Newborn , Base Sequence , Biological Transport , Blood Glucose/analysis , Glucose-6-Phosphate , Glucosephosphates/genetics , Glucosephosphates/metabolism , Glycogen Storage Disease Type I/genetics , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Models, Biological , Molecular Sequence Data , Phenotype
6.
J Hepatol ; 54(3): 529-37, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21109326

ABSTRACT

BACKGROUND AND AIMS: Glycogen storage disease type 1a (GSD1a) is an inherited disease caused by a deficiency in the catalytic subunit of the glucose-6 phosphatase enzyme (G6Pase). GSD1a is characterized by hypoglycaemia, hyperlipidemia, and lactic acidosis with associated hepatic (including hepatocellular adenomas), renal, and intestinal disorders. A total G6pc (catalytic subunit of G6Pase) knock-out mouse model has been generated that mimics the human pathology. However, these mice rarely live longer than 3 months and long-term liver pathogenesis cannot be evaluated. Herein, we report the long-term characterization of a liver-specific G6pc knock-out mouse model (L-G6pc(-/-)). METHODS: We generated L-G6pc(-/-) mice using an inducible CRE-lox strategy and followed up the development of hepatic tumours using magnetic resonance imaging. RESULTS: L-G6pc(-/-) mice are viable and exhibit normoglycemia in the fed state. They develop hyperlipidemia, lactic acidosis, and uricemia during the first month after gene deletion. However, these plasmatic parameters improved after 6 months. L-G6pc(-/-) mice develop hepatomegaly with glycogen accumulation and hepatic steatosis. Using an MRI approach, we could detect hepatic nodules with diameters of less than 1 mm, 9 months after induction of deficiency. Hepatic nodules (1 mm) were detected in 30-40% of L-G6pc(-/-) mice at 12 months. After 18 months, all L-G6pc(-/-) mice developed multiple hepatocellular adenomas of 1-10 mm diameter. CONCLUSIONS: This is the first report of a viable animal model of the hepatic pathology of GSD1a, including the late development of hepatocellular adenomas.


Subject(s)
Adenoma, Liver Cell/etiology , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/genetics , Liver Neoplasms, Experimental/etiology , Liver/enzymology , Adenoma, Liver Cell/enzymology , Adenoma, Liver Cell/pathology , Animals , Base Sequence , DNA Primers , Disease Models, Animal , Fatty Liver/enzymology , Fatty Liver/etiology , Fatty Liver/pathology , Female , Gene Knockout Techniques , Gene Targeting , Glycogen Storage Disease Type I/enzymology , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/genetics , Hepatomegaly/enzymology , Hepatomegaly/etiology , Hepatomegaly/pathology , Humans , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease
7.
Hum Mutat ; 29(7): 921-30, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18449899

ABSTRACT

Glucose-6-phosphatase-alpha (G6PC) is a key enzyme in glucose homeostasis that catalyzes the hydrolysis of glucose-6-phosphate to glucose and phosphate in the terminal step of gluconeogenesis and glycogenolysis. Mutations in the G6PC gene, located on chromosome 17q21, result in glycogen storage disease type Ia (GSD-Ia), an autosomal recessive metabolic disorder. GSD-Ia patients manifest a disturbed glucose homeostasis, characterized by fasting hypoglycemia, hepatomegaly, nephromegaly, hyperlipidemia, hyperuricemia, lactic acidemia, and growth retardation. G6PC is a highly hydrophobic glycoprotein, anchored in the membrane of the endoplasmic reticulum with the active center facing into the lumen. To date, 54 missense, 10 nonsense, 17 insertion/deletion, and three splicing mutations in the G6PC gene have been identified in more than 550 patients. Of these, 50 missense, two nonsense, and two insertion/deletion mutations have been functionally characterized for their effects on enzymatic activity and stability. While GSD-Ia is not more prevalent in any ethnic group, mutations unique to Caucasian, Oriental, and Jewish populations have been described. Despite this, GSD-Ia patients exhibit phenotypic heterogeneity and a stringent genotype-phenotype relationship does not exist.


Subject(s)
Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/etiology , Humans , Mutation , Phenotype
8.
J Pediatr Endocrinol Metab ; 31(4): 473-478, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29374762

ABSTRACT

BACKGROUND: Glycogen storage diseases (GSDs) are a collection of disorders related to glycogen synthesis or degradation that classically present in infancy with hypoglycemia, failure to thrive and hepatomegaly; however, their phenotype can vary significantly. CASE PRESENTATION: We present the cases of two children, 5 years old and 3.5 years old, who were referred to endocrinology for short stature. They were ultimately found to have hepatomegaly, fasting hypoglycemia, mild elevation of transaminases and ketosis. Laboratory and genetic studies were consistent with double heterozygosity for GSDs Ia and III, with one novel mutation discovered in each patient. Nightly, both children were treated with cornstarch, which resulted in resolution of laboratory abnormalities and improvement in their growth velocity. These cases are unusual in that GSD was diagnosed relatively late in life in patients with no previous history of severe hypoglycemia. CONCLUSIONS: They highlight the importance of considering glycogen storage disease in a child presenting with short stature, as it is a treatable disease that can be diagnosed non-invasively with genetic testing.


Subject(s)
Dwarfism/complications , Glycogen Storage Disease Type III/diagnosis , Glycogen Storage Disease Type I/diagnosis , Hepatomegaly/complications , Child, Preschool , Genetic Testing , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type III/etiology , Humans , Male , Mutation , Phenotype , Prognosis
9.
FEBS Lett ; 581(20): 3833-8, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17659284

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) patients deficient in glucose-6-phosphatase-alpha manifest a disturbed glucose homeostasis. We hypothesized that disturbed glucose homeostasis might affect myeloid functions. Here, we show that GSD-Ia mice exhibit normal neutrophil activities but have elevated myeloid progenitor cells in the bone marrow and spleen. Interestingly, GSD-Ia mice exhibit a persistent increase in peripheral blood neutrophil counts along with elevated serum levels of granulocyte colony stimulating factor and cytokine-induced neutrophil chemoattractant. Taken together, our results suggest that a loss of glucose homeostasis can compromise the immune system, resulting in neutrophilia. This may explain some of the unexpected clinical manifestations seen in GSD-Ia.


Subject(s)
Cytokines/blood , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/metabolism , Neutrophils/metabolism , Animals , Calcium/analysis , Calcium/metabolism , Chemokine CXCL2 , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Glycogen Storage Disease Type I/pathology , Mice , Mice, Knockout , Monokines/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Respiratory Burst/physiology
10.
Semin Hematol ; 39(2): 103-6, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11957192

ABSTRACT

Glycogen storage disease (GSD) is a rare autosomal-recessive disorder characterized by hypoglycemia, hepatosplenomegaly, seizures, and failure to thrive in infants. Neutropenia and/or neutrophil dysfunction develops in GSD1b, but not in other types. GSD1b results from a deficiency of the glucose-6-phosphate translocase enzyme and the genetic defect maps to chromosome 11q23. Patients with GSD1b are susceptible to recurrent bacterial infections, commonly involving the perirectal area, ears, skin, and urinary tract, although life-threatening infections, such as septicemia, pneumonia, and meningitis occur less frequently. Although the exact mechanism of neutropenia in patients with GSD1b is not known, treatment with recombinant human granulocyte colony-stimulating factor (G-CSF) has reduced the incidence of infections and has improved the quality of life of these patients. Defects in neutrophil chemotaxis and intracellular bacterial killing have been described and appear to be corrected by the use of G-CSF. To date, no cases of myelodysplasia or acute myeloid leukemia have been observed in patients with GSD1b treated with G-CSF. A significant complication of cytokine therapy is the development of hypersplenism, requiring either a reduction in the dosage of G-CSF or splenectomy.


Subject(s)
Glycogen Storage Disease , Bacterial Infections/etiology , Glycogen Storage Disease/complications , Glycogen Storage Disease/drug therapy , Glycogen Storage Disease/etiology , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/drug therapy , Glycogen Storage Disease Type I/etiology , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Neutropenia/etiology
11.
Vopr Med Khim ; 25(4): 371-5, 1979.
Article in Russian | MEDLINE | ID: mdl-382625

ABSTRACT

More than ten patients with glycogen-storage disease, which were classified as patients with glycogenosis of the I type--deficiency in glucose-6-phosphatase) on the basis of clinical data and biochemical analyses in vivo, were detected within the last few years. But activity of glucose-6-phosphatase was found to be normal in biopsy of samples of the liver tissue obtained from these patients. This disease was termed as glycogenosis of the Ib type. A hypothesis is advanced, according to which the discrepancy in data on biochemical study of the patients in vivo and in vitro is due to absence of a specific permease in liver tissue, which transfers glucose-6-phosphate from cytosol onto the innesurface of membranes of cytoplasmic network, where glucose-6-phosphatase is located.


Subject(s)
Glucosephosphates/blood , Glycogen Storage Disease Type I/etiology , Adolescent , Biological Transport , Blood Glucose/metabolism , Child , Child, Preschool , Enzyme Activation , Female , Glucosephosphate Dehydrogenase/blood , Glycogen Storage Disease Type I/blood , Humans , Infant , Lactates/blood , Liver/metabolism , Liver Glycogen/metabolism , Male
12.
Nat Rev Endocrinol ; 6(12): 676-88, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20975743

ABSTRACT

Glycogen storage disease type I (GSD-I) consists of two subtypes: GSD-Ia, a deficiency in glucose-6-phosphatase-α (G6Pase-α) and GSD-Ib, which is characterized by an absence of a glucose-6-phosphate (G6P) transporter (G6PT). A third disorder, G6Pase-ß deficiency, shares similarities with this group of diseases. G6Pase-α and G6Pase-ß are G6P hydrolases in the membrane of the endoplasmic reticulum, which depend on G6PT to transport G6P from the cytoplasm into the lumen. A functional complex of G6PT and G6Pase-α maintains interprandial glucose homeostasis, whereas G6PT and G6Pase-ß act in conjunction to maintain neutrophil function and homeostasis. Patients with GSD-Ia and those with GSD-Ib exhibit a common metabolic phenotype of disturbed glucose homeostasis that is not evident in patients with G6Pase-ß deficiency. Patients with a deficiency in G6PT and those lacking G6Pase-ß display a common myeloid phenotype that is not shared by patients with GSD-Ia. Previous studies have shown that neutrophils express the complex of G6PT and G6Pase-ß to produce endogenous glucose. Inactivation of either G6PT or G6Pase-ß increases neutrophil apoptosis, which underlies, at least in part, neutrophil loss (neutropenia) and dysfunction in GSD-Ib and G6Pase-ß deficiency. Dietary and/or granulocyte colony-stimulating factor therapies are available; however, many aspects of the diseases are still poorly understood. This Review will address the etiology of GSD-Ia, GSD-Ib and G6Pase-ß deficiency and highlight advances in diagnosis and new treatment approaches, including gene therapy.


Subject(s)
Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/therapy , Animals , Genetic Therapy/methods , Genotype , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/complications , Glycogen Storage Disease Type I/metabolism , Humans , Kidney Transplantation/physiology , Liver Transplantation/physiology , Models, Biological , Phenotype
15.
Sci Signal ; 2(58): pe8, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19224896

ABSTRACT

The enzyme glucose-6-phosphatase is critical for maintaining fasting blood sugar levels by increasing hepatic glucose production. Its absence in patients with von Gierke's disease leads to severe hypoglycemia and abnormal accumulation of glycogen (glycogenosis) in the liver. New players that control the expression of glucose-6-phosphatase have been identified that may provide insight into this metabolic disorder, as well as type 2 diabetes.


Subject(s)
Glycogen Storage Disease Type I/etiology , Diabetes Mellitus, Type 2/etiology , Gene Expression Regulation, Enzymologic , Glucose-6-Phosphatase/metabolism , Humans
16.
Am J Perinatol ; 19(4): 215-9, 2002 May.
Article in English | MEDLINE | ID: mdl-12012283

ABSTRACT

The objective of this study is to study the absolute number of nucleated red blood cells (RBC) at birth, an index of active fetal erythropoiesis, in infants with G6PD deficiency and in controls. We tested the hypothesis that hematocrit and hemoglobin would be lower, and absolute nucleated RBC counts higher, in the G6PD deficient and that these changes would be more prominent in infants exposed passively to fava bean through maternal diet. Thirty-two term infants with G6PD deficiency were compared with 30 term controls. Complete blood counts with manual differential counts were obtained within 12 hours of life. Absolute nucleated RBC and corrected leukocyte counts were computed from the Coulter results and the differential count. G6PD deficient patients did not differ from controls in terms of gestational age, birth weight, or Apgar scores or in any of the hematologic parameters studied, whether or not the mother reported fava beans consumption in the days prior to delivery. Although intrauterine hemolysis is possible in G6PD deficient fetuses exposed passively to fava beans, our study supports that such events must be very rare.


Subject(s)
Erythroblasts , Glycogen Storage Disease Type I/blood , Adult , Erythroblastosis, Fetal/etiology , Erythrocyte Count , Favism/complications , Female , Glycogen Storage Disease Type I/etiology , Hematocrit , Humans , Infant, Newborn , Pilot Projects , Pregnancy , Prenatal Exposure Delayed Effects , Prospective Studies
17.
Enzyme ; 25(2): 132-7, 1980.
Article in English | MEDLINE | ID: mdl-6929752

ABSTRACT

Based on the consideration that normal metabolic processes depend upon the activity of key enzymes (and membrane carriers) as modulated by regulatory factors (hormones, diet, endogenous compounds, age, physical activity, environmental agents), metabolic disorders might be classified into two groups: (I) defects in enzyme synthesis, leading to enzyme deficiency (classical inborn errors of metabolism) or to qualitative (structural) enzyme alterations (entailing unresponsiveness to regulation), in the presence of normal regulatory factors; (II) defects in enzyme regulation, which include metabolic syndromes such as diabetes mellitus, obesity and hyperlipoproteinemias (other than type I), and are due to changes in enzyme activities caused by alterations in regulatory factor(s) (secondary to various causes), in the presence of normally responsive enzymes.


Subject(s)
Diabetes Mellitus/enzymology , Enzymes/physiology , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/metabolism , Metabolic Diseases/enzymology , Allosteric Regulation , Enzymes/biosynthesis , Enzymes/metabolism , Humans , Metabolic Diseases/etiology , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/etiology , Mutation
18.
Cancer Detect Prev ; 8(1-2): 1-9, 1985.
Article in English | MEDLINE | ID: mdl-2415253

ABSTRACT

Animal experiments on markers of tumors induced by physical and chemical carcinogens are discussed. The results lead to the conclusion that there are no specific markers induced by certain carcinogenic agents.


Subject(s)
Carcinogens/pharmacology , Neoplasms, Experimental/analysis , Animals , Carcinoembryonic Antigen/analysis , Cytoskeleton , Glycogen Storage Disease Type I/etiology , Hormones, Ectopic/analysis , Humans , Isoenzymes/analysis , Liver Glycogen/metabolism , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/metabolism , Polyamines/analysis , Rats , alpha-Fetoproteins/analysis
19.
Hum Mol Genet ; 12(19): 2547-58, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12925567

ABSTRACT

Glycogen storage disease type Ib (GSD-Ib) is caused by a deficiency in the glucose-6-phosphate transporter (G6PT). In addition to disrupted glucose homeostasis, GSD-Ib patients have unexplained and unexpected defects in neutrophil respiratory burst, chemotaxis and calcium flux, in response to the bacterial peptide f-Met-Leu-Phe, as well as intermittent neutropenia. We generated a G6PT knockout (G6PT-/-) mouse that mimics all known defects of the human disorder and used the model to further our understanding of the pathogenesis of GSD-Ib. We demonstrate that the neutropenia is caused directly by the loss of G6PT activity; that chemotaxis and calcium flux, induced by the chemokines KC and macrophage inflammatory protein-2, are defective in G6PT-/- neutrophils; and that local production of these chemokines and the resultant neutrophil trafficking in vivo are depressed in G6PT-/- ascites during an inflammatory response. The bone and spleen of G6PT-/- mice are developmentally delayed and accompanied by marked hypocellularity of the bone marrow, elevation of myeloid progenitor cell frequencies in both organs and a corresponding dramatic increase in granulocyte colony stimulating factor levels in both GSD-Ib mice and humans. So, in addition to transient neutropenia, a sustained defect in neutrophil trafficking due to both the resistance of neutrophils to chemotactic factors, and reduced local production of neutrophil-specific chemokines at sites of inflammation, may underlie the myeloid deficiency in GSD-Ib. These findings demonstrate that G6PT is not just a G6P transport protein but also an important immunomodulatory protein whose activities need to be addressed in treating the myeloid complications in GSD-Ib patients.


Subject(s)
Antiporters/deficiency , Glucose/metabolism , Homeostasis , Monosaccharide Transport Proteins/deficiency , Neutrophils/metabolism , Animals , Calcium/metabolism , Chemokines/metabolism , Chemotaxis, Leukocyte , Disease Models, Animal , Genes, Recessive , Genetic Variation , Glycogen Storage Disease Type I/etiology , Glycogen Storage Disease Type I/pathology , Kidney/pathology , Kinetics , Liver/pathology , Mice , Mice, Knockout , Neutropenia/etiology , Neutropenia/physiopathology , Respiratory Burst , Restriction Mapping , Time Factors
20.
South Med J ; 74(6): 761-4, 1981 Jun.
Article in English | MEDLINE | ID: mdl-6264632

ABSTRACT

We have described a 20-month-old child with type IB glycogen storage disease, based on clinical and biochemical manifestations. Functional testing data were similar to those found in glucose-6-phosphatase deficiency, but in vitro studies showed normal hepatic glucose-6-phosphatase activity. Disruption of membranes with deoxycholic acid was followed by an increase in enzyme activity compared to a control liver tissue, suggesting "latency" of enzyme. We suggest that this patient had glycogen storage type IB and that this disorder may represent a specific glucose-6-phosphate transport defect.


Subject(s)
Glycogen Storage Disease Type I/etiology , Adult , Biological Transport, Active/drug effects , Blood Glucose/metabolism , Deoxycholic Acid/pharmacology , Female , Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/metabolism , Humans , Lactates/blood , Liver/drug effects , Liver/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL