Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
Add more filters











Publication year range
1.
Lab Anim ; 47(2): 116-21, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23492514

ABSTRACT

Mouse parvoviruses are among the most prevalent infectious pathogens in contemporary mouse colonies. To improve the efficiency of routine screening for mouse parvovirus infections, a multiplex polymerase chain reaction (PCR) assay targeting the VP gene was developed. The assay detected minute virus of mice (MVM), mouse parvovirus (MPV) and a mouse housekeeping gene (α-actin) and was able to specifically detect MVM and MPV at levels as low as 50 copies. Co-infection with the two viruses with up to 200-fold differences in viral concentrations can easily be detected. The multiplex PCR assay developed here could be a useful tool for monitoring mouse health and the viral contamination of biological materials.


Subject(s)
Mice , Multiplex Polymerase Chain Reaction/methods , Parvoviridae Infections/diagnosis , Parvovirus/isolation & purification , Rodent Diseases/diagnosis , Actins/genetics , Actins/metabolism , Animals , Minute Virus of Mice/genetics , Minute Virus of Mice/isolation & purification , Minute Virus of Mice/metabolism , Parvoviridae Infections/virology , Parvovirus/genetics , Parvovirus/metabolism , Rodent Diseases/virology , Sensitivity and Specificity , Sequence Analysis, DNA , Viral Proteins/genetics , Viral Proteins/metabolism
2.
Kidney Int ; 73(6): 716-23, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18075499

ABSTRACT

Patients with glycogen storage disease type Ia (GSD-Ia) develop renal disease of unknown etiology despite intensive dietary therapies. This renal disease shares many clinical and pathological similarities to diabetic nephropathy. We studied the expression of angiotensinogen, angiotensin type 1 receptor, transforming growth factor-beta1, and connective tissue growth factor in mice with GSD-Ia and found them to be elevated compared to controls. While increased renal expression of angiotensinogen was evident in 2-week-old mice with GSD-Ia, the renal expression of transforming growth factor-beta and connective tissue growth factor did not increase for another week; consistent with upregulation of these factors by angiotensin II. The expression of fibronectin and collagens I, III, and IV was also elevated in the kidneys of mice with GSD-Ia, compared to controls. Renal fibrosis was characterized by a marked increase in the synthesis and deposition of extracellular matrix proteins in the renal cortex and histological abnormalities including tubular basement membrane thickening, tubular atrophy, tubular dilation, and multifocal interstitial fibrosis. Our results suggest that activation of the angiotensin system has an important role in the pathophysiology of renal disease in patients with GSD-Ia.


Subject(s)
Angiotensins/metabolism , Glycogen Storage Disease Type I/complications , Kidney Diseases/etiology , Kidney Diseases/pathology , Kidney/pathology , Angiotensin II/genetics , Angiotensin II/metabolism , Angiotensinogen/genetics , Angiotensinogen/metabolism , Angiotensins/genetics , Animals , Connective Tissue Growth Factor , Extracellular Matrix/metabolism , Fibrosis , Glucose-6-Phosphatase/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Diseases/metabolism , Mice , Mice, Mutant Strains , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
3.
Gene Ther ; 14(3): 219-26, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17006547

ABSTRACT

Glycogen storage disease type Ib (GSD-Ib) is caused by a deficiency in the glucose-6-phosphate transporter (G6PT), an endoplasmic reticulum-associated transmembrane protein that is ubiquitously expressed. GSD-Ib patients suffer from disturbed glucose homeostasis and myeloid dysfunctions. To evaluate the feasibility of gene replacement therapy for GSD-Ib, we have infused adenoviral (Ad) vector containing human G6PT (Ad-hG6PT) into G6PT-deficient (G6PT(-/-)) mice that manifest symptoms characteristics of the human disorder. Ad-hG6PT infusion restores significant levels of G6PT mRNA expression in the liver, bone marrow and spleen, and corrects metabolic as well as myeloid abnormalities in G6PT(-/-) mice. The G6PT(-/-) mice receiving gene therapy exhibit improved growth; normalized serum profiles for glucose, cholesterol, triglyceride, uric acid and lactic acid; and reduced hepatic glycogen deposition. The therapy also corrects neutropenia and lowers the elevated serum levels of granulocyte colony-stimulating factor. The development of bone and spleen in the infused G6PT(-/-) mice is improved and accompanied by increased cellularity and normalized myeloid progenitor cell frequencies in both tissues. This effective use of gene therapy to correct metabolic imbalances and myeloid dysfunctions in GSD-Ib mice holds promise for the future of gene therapy in humans.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glucose Transport Proteins, Facilitative/genetics , Glucose-6-Phosphate/metabolism , Glycogen Storage Disease Type I/therapy , Animals , Animals, Newborn , Bone Marrow/pathology , Gene Expression , Glucose Transport Proteins, Facilitative/metabolism , Glycogen Storage Disease Type I/metabolism , Glycogen Storage Disease Type I/pathology , Humans , Injections , Mice , Mice, Knockout , Microsomes, Liver/metabolism , Neutropenia/therapy , Spleen/pathology , Transduction, Genetic/methods , Transgenes
4.
Gene Ther ; 13(4): 321-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16195703

ABSTRACT

Glycogen storage disease type Ia (GSD-Ia) is caused by a deficiency in glucose-6-phosphatase-alpha (G6Pase-alpha), a nine-transmembrane domain, endoplasmic reticulum-associated protein expressed primarily in the liver and kidney. Previously, we showed that infusion of an adeno-associated virus (AAV) serotype 2 vector carrying murine G6Pase-alpha (AAV2-G6Pase-alpha) into neonatal GSD-Ia mice failed to sustain their life beyond weaning. We now show that neonatal infusion of GSD-Ia mice with an AAV serotype 1-G6Pase-alpha (AAV1-G6Pase-alpha) or AAV serotype 8-G6Pase-alpha (AAV8-G6Pase-alpha) results in hepatic expression of the G6Pase-alpha transgene and markedly improves the survival of the mice. However, only AAV1-G6Pase-alpha can achieve significant renal transgene expression. A more effective strategy, in which a neonatal AAV1-G6Pase-alpha infusion is followed by a second infusion at age one week, provides sustained expression of a complete, functional, G6Pase-alpha system in both the liver and kidney and corrects the metabolic abnormalities in GSD-Ia mice for the 57 week length of the study. This effective use of gene therapy to correct metabolic imbalances and disease progression in GSD-Ia mice holds promise for the future of gene therapy in humans.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/therapy , Isoenzymes/genetics , Animals , Animals, Newborn , Dependovirus/immunology , Gene Expression , Genetic Vectors/genetics , Glycogen Storage Disease Type I/enzymology , Infusions, Intravenous , Kidney/enzymology , Liver/enzymology , Mice , Mice, Mutant Strains , Microsomes , Serotyping , Transgenes
5.
Diabetologia ; 48(9): 1851-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16012821

ABSTRACT

AIMS/HYPOTHESIS: There are three members of the glucose-6-phosphatase (G6Pase) family: (1) the liver/kidney/intestine G6Pase-alpha (encoded by G6PC), which is a key enzyme in glucose homeostasis; (2) the ubiquitous G6Pase-beta (encoded by G6PC3); and (3) the islet-specific G6Pase-related protein (IGRP, encoded by /G6PC2). While G6Pase-alpha and G6Pase-beta are functional glucose-6-phosphate hydrolases, IGRP possesses almost no hydrolase activity. This was unexpected since G6Pase-alpha is more closely related to IGRP than G6Pase-beta. Recently, amino acids 206-214 in IGRP were identified as a beta cell antigen targeted by a prevalent population of pathogenic CD8+ T cells in autoimmune diabetes, suggesting that this peptide confers functional specificity to IGRP. We therefore investigated the molecular events that inactivate IGRP activity and the effects of the beta cell antigen sequence on the stability and enzymatic activity of G6Pase-alpha. METHODS: Studies were performed using site-directed mutagenesis and transient expression assays. Protein stability was evaluated by Western blotting, proteasome inhibitor studies and in vitro transcription-translation. RESULTS: We showed that the residues responsible for G6Pase activity are more extensive than previously recognised. Introducing the IGRP antigenic motif into G6Pase-alpha does not completely destroy activity, although it does destabilise the protein. The low hydrolytic activity in IGRP is due to the combination of multiple independent mutations. CONCLUSIONS/INTERPRETATION: The loss of catalytic activity in IGRP arises from the sum of many sequence differences. G6Pase-alpha mutants containing the beta cell antigen sequence are preferentially degraded in cells, which prevents targeting by pathogenic CD8+ T cells. It is possible that IGRP levels in beta cells could dictate susceptibilities to diabetes.


Subject(s)
Glucose-6-Phosphatase/physiology , Islets of Langerhans/physiology , Phosphoric Monoester Hydrolases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Conserved Sequence , DNA Primers , Dogs , Glucose-6-Phosphatase/chemistry , Glucose-6-Phosphatase/genetics , Humans , Islets of Langerhans/enzymology , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Rats , Sequence Alignment , Sequence Homology, Amino Acid
6.
Gene Ther ; 12(10): 803-14, 2005 May.
Article in English | MEDLINE | ID: mdl-15772691

ABSTRACT

Newborn gene therapy, because it can prevent the damage caused by the onset of a disease, deserves specific attention. To evaluate gene transfer in tissues of newborn mice, we used a human immunodeficiency virus (HIV)-2 based lentiviral vector pseudotyped with vesicular stomatitis virus G glycoprotein expressing the green fluorescent protein reporter gene under the control of the cytomegalovirus promoter. We found that very low doses of HIV-2 could infect and be expressed in newborn mice. Under these conditions, the virus was preferentially expressed in the liver and hepatocytes were the predominant target. The treatment was not toxic, the infected liver cells proliferated and the transduced gene was stably expressed. Adult mice could be infected by HIV-2, but the vector was detected in the liver only utilizing the sensitive method of polymerase chain reaction coupled with Southern blot. Direct comparison between newborn and adult recipients demonstrated a much greater efficiency of liver transduction in the newborn mouse. These results indicate that the combination of early intervention and low multiplicity of infection may be a strategy for preferentially and efficiently targeting newborn liver for gene therapy applications.


Subject(s)
Animals, Newborn , Genetic Therapy/methods , Genetic Vectors/administration & dosage , HIV-2/genetics , Hepatocytes/metabolism , Transduction, Genetic/methods , Adenoviridae/genetics , Animals , Blotting, Southern , Cell Proliferation , Female , Gene Expression , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Hepatocytes/cytology , Liver/metabolism , Lung/metabolism , Mice , Mice, Inbred Strains , Microscopy, Fluorescence , Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism
7.
J Comp Pathol ; 131(2-3): 214-20, 2004.
Article in English | MEDLINE | ID: mdl-15276861

ABSTRACT

This study established a modified alkaline phosphatase-labelled avidin-biotin-complex (ABC-AP) method for diagnosis of mouse hepatitis virus (MHV) and Mycoplasma pulmonis infection from formalin-fixed, paraffin wax-embedded sections, murine antibody-positive serum being used as the primary reagent. With this method, MHV antigen in cAnNCrj.Cg-Foxn1(nu)/Foxn1(nu) mice and M. pulmonis antigen in Wistar rats were immunolabelled in tissue sections. MHV antigen was clearly detected in samples of liver, stomach, caecal and colonic mucosa, and spleen. M. pulmonis antigen was demonstrated on the luminal surface of bronchiolar epithelial cells. This method may prove useful in diagnosis when commercial antisera are unavailable or when immunosuppression prevents serological diagnosis.


Subject(s)
Coronavirus Infections/diagnosis , Immune Sera , Mycoplasma Infections/diagnosis , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Antigens, Viral/immunology , Antigens, Viral/isolation & purification , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Female , Immunohistochemistry/methods , Male , Mice , Murine hepatitis virus/immunology , Mycoplasma Infections/immunology , Mycoplasma Infections/pathology , Mycoplasma pulmonis/immunology , Rats
8.
Life Sci ; 71(11): 1293-301, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12106594

ABSTRACT

Monitoring gene therapy of glycogen storage disease type 1a in a mouse model was achieved using [(18)F]FDG and a dedicated animal scanner. The G6Pase knockout (KO) mice were compared to the same mice after infusion with a recombinant adenovirus containing the murine G6Pase gene (Ad-mG6Pase). Serial images of the same mouse before and after therapy were obtained and compared with wild-type (WT) mice of the same strain to determine the uptake and retention of [(18)F]FDG in the liver. Image data were acquired from heart, blood pool and liver for twenty minutes after injection of [(18)F]FDG. The retention of [(18)F]FDG was lower for the WT mice compared to the KO mice. The mice treated with adenovirus-mediated gene therapy had retention similar to that found in age-matched WT mice. These studies show that FDG can be used to monitor the G6Pase concentration in liver of WT mice as compared to G6Pase KO mice. In these mice, gene therapy returned the liver function to that found in age matched WT controls as measured by the FDG kinetics in the liver compared to that found in age matched wild type controls.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Genetic Therapy , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/diagnostic imaging , Glycogen Storage Disease Type I/therapy , Tomography, Emission-Computed , Animals , Disease Models, Animal , Glucose/metabolism , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Radiopharmaceuticals/metabolism
9.
DNA Cell Biol ; 20(8): 447-53, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11560776

ABSTRACT

Deficiencies in the glucose-6-phosphate transporter (G6PT) cause glycogen storage disease type 1b (GSD-1b), a heritable metabolic disorder. The G6PT protein translocates glucose-6-phosphate from the cytoplasm to the lumen of the endoplasmic reticulum, where glucose-6-phosphatase metabolizes it to glucose and phosphate. Therefore, G6PT and glucose-6-phosphatase work in concert to maintain glucose homeostasis. To delineate the control of G6PT gene expression, we first demonstrated that transcription of the gene requires hepatocyte nuclear factor 1alpha. Consequently, hepatocyte nuclear factor 1alpha-null mice manifest a G6PT deficiency like that of GSD-1b patients. In this study, we delineated the role of glucocorticoids in the transcription of the G6PT gene. We showed that the basal G6PT promoter is contained within nucleotides -369 to -1 upstream of the translation start site, which contains three activation elements. Further, we demonstrated that glucocorticoids activate G6PT transcription and that glucocorticoid action is mediated through a glucocorticoid response element within activation element-2 of the promoter. Taken together, the results suggest that glucocorticoids play a pivotal role in regulating the G6PT gene.


Subject(s)
Antiporters/genetics , Dexamethasone/metabolism , Glucocorticoids/metabolism , Glycogen Storage Disease Type I/genetics , Monosaccharide Transport Proteins/genetics , Transcriptional Activation , Animals , Base Sequence , Dexamethasone/pharmacology , Gene Expression/drug effects , Glucocorticoids/pharmacology , Humans , Molecular Sequence Data , Rats , Tumor Cells, Cultured
11.
Am J Hum Genet ; 69(4): 712-21, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11505338

ABSTRACT

Although >55 CTNS mutations occur in patients with the lysosomal storage disorder cystinosis, no regulatory mutations have been reported, because the promoter has not been defined. Using CAT reporter constructs of sequences 5' to the CTNS coding sequence, we identified the CTNS promoter as the region encompassing nucleotides -316 to +1 with respect to the transcription start site. This region contains an Sp-1 regulatory element (GGCGGCG) at positions -299 to -293, which binds authentic Sp-1, as shown by electrophoretic-mobility-shift assays. Three patients exhibited mutations in the CTNS promoter. One patient with nephropathic cystinosis carried a -295 G-->C substitution disrupting the Sp-1 motif, whereas two patients with ocular cystinosis displayed a -303 G-->T substitution in one case and a -303 T insertion in the other case. Each mutation drastically reduced CAT activity when inserted into a reporter construct. Moreover, each failed either to cause a mobility shift when exposed to nuclear extract or to compete with the normal oligonucleotide's mobility shift. The CTNS promoter region shares 41 nucleotides with the promoter region of an adjacent gene of unknown function, CARKL, whose start site is 501 bp from the CTNS start site. However, the patients' CTNS promoter mutations have no effect on CARKL promoter activity. These findings suggest that the CTNS promoter region should be examined in patients with cystinosis who have fewer than two coding-sequence mutations.


Subject(s)
Conserved Sequence/genetics , Cystinosis/genetics , Glycoproteins , Membrane Proteins/genetics , Mutation/genetics , Phosphotransferases , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Adult , Amino Acid Transport Systems, Neutral , Base Sequence , Child , Child, Preschool , DNA/genetics , DNA/metabolism , DNA Mutational Analysis , Female , Gene Order/genetics , Genotype , HeLa Cells , Humans , Infant , Membrane Transport Proteins , Molecular Sequence Data , Phenotype , Phosphotransferases (Alcohol Group Acceptor) , Protein Binding , Response Elements/genetics
12.
Arch Biochem Biophys ; 393(1): 117-22, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11516168

ABSTRACT

The effect of Ehrlich ascites tumor cells, in vivo, on the hepatic glucose-6-phosphatase (G6Pase) system was examined. The V(max) for glucose 6-phosphate hydrolysis by G6Pase was reduced by 40% and a greater than 15-fold decrease in mRNA encoding the catalytic unit of the G6Pase system was observed 8 days after injection with tumor cells. Blood glucose concentration was decreased from 169 +/- 17 to 105 +/- 9 mg/dl in tumor-bearing mice. There was no change in the G6P transporter (G6PT) mRNA level. However, there was a significant decrease in G6P accumulation into hepatic microsomal vesicles derived from tumor-bearing mice. Decreased G6P accumulation was also associated with a decrease in G6Pase hydrolytic activity in the presence of vanadate, a potent catalytic-unit inhibitor. In addition, G6P accumulation was nearly abolished in microsomes treated with N-bromoacetylethanolamine phosphate, an irreversible inhibitor of the G6PT. These results demonstrate that the catalytic unit and G6PT components of the G6Pase system can be discriminantly regulated, and that microsomal glucose 6-phosphate uptake is dependent on catalytic unit activity as well as G6PT action.


Subject(s)
Carcinoma, Ehrlich Tumor/enzymology , Glucose-6-Phosphatase/chemistry , Glucose-6-Phosphatase/metabolism , Liver/enzymology , Animals , Blood Glucose/metabolism , Carcinoma, Ehrlich Tumor/genetics , Carcinoma, Ehrlich Tumor/metabolism , Catalytic Domain , Ethanolamines/pharmacology , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphate/metabolism , Kinetics , Male , Mice , Mice, Inbred ICR , Microsomes, Liver/enzymology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
13.
DNA Cell Biol ; 20(3): 149-57, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11313018

ABSTRACT

The pancreatic secretory phospholipase A(2) (sPLA(2)IB) is considered to be a digestive enzyme, although it has several important receptor-mediated functions. In this study, using the newly isolated murine sPLA(2)IB cDNA clone as a probe, we demonstrate that in addition to the pancreas, the sPLA(2)IB mRNA was expressed in extrapancreatic organs such as the liver, spleen, duodenum, colon, and lungs. We also demonstrate that sPLA(2)IB mRNA expression was detectable from the 17(th) day of gestation in the developing mouse fetus, coinciding with the time of completion of differentiation of the pancreas. Furthermore, the mRNA expression pattern of sPLA(2)IB was distinct from those of sPLA(2)IIA and cPLA(2) in various tissues examined. The murine sPLA(2)IB gene structure is well conserved, consistent with findings in other mammalian species, and this gene mapped to the region of mouse chromosome 5F1-G1.1. Taken together, our results suggest that sPLA(2)IB plays important roles both in the pancreas and in extrapancreatic tissues and that in the mouse, its expression is developmentally regulated.


Subject(s)
Pancreas/enzymology , Phospholipases A/genetics , Phospholipases A/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 12 , Conserved Sequence , Cyclooxygenase 2 , Cysteine , Embryo, Mammalian/enzymology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Membrane Proteins , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism
14.
J Biol Chem ; 276(11): 7963-7, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11121425

ABSTRACT

The clinical manifestations of type 1 glycogen storage disease (GSD-1) in patients deficient in the glucose-6-phosphatase (G6Pase) system (e.g. growth retardation, hepatomegaly, hyperlipidemia, and renal dysfunction) are shared by Hnf1alpha(-/-) mice deficient of a transcriptional activator, hepatocyte nuclear factor 1alpha (HNF1alpha). However, the molecular mechanism is unknown. The G6Pase system, essential for the maintenance of glucose homeostasis, is comprised of glucose 6-phosphate transporter (G6PT) and G6Pase. G6PT translocates G6P from the cytoplasm to the lumen of the endoplasmic reticulum where it is metabolized by G6Pase to glucose and phosphate. Deficiencies in G6Pase and G6PT cause GSD-1a and GSD-1b, respectively. Hnf1alpha(-/-) mice also develop noninsulin-dependent diabetes mellitus caused by defective insulin secretion. In this study, we sought to determine whether there is a molecular link between HNF1alpha deficiency and function of the G6Pase system. Transactivation studies revealed that HNF1alpha is required for transcription of the G6PT gene. Hepatic G6PT mRNA levels and microsomal G6P transport activity are also markedly reduced in Hnf1alpha(-/-) mice as compared with Hnf1alpha(+/+) and Hnf1alpha(+/-) littermates. On the other hand, hepatic G6Pase mRNA expression and activity are up-regulated in Hnf1alpha(-/-) mice, consistent with observations that G6Pase expression is increased in diabetic animals. Taken together, the results strongly suggest that metabolic abnormalities in HNF1alpha-null mice are caused in part by G6PT deficiency and by perturbations of the G6Pase system.


Subject(s)
DNA-Binding Proteins , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/metabolism , Nuclear Proteins , Phosphotransferases/genetics , Transcription Factors/physiology , Animals , Antiporters , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Hepatocyte Nuclear Factor 1-beta , Humans , Mice , Mice, Knockout , Monosaccharide Transport Proteins , Phenotype , Promoter Regions, Genetic , RNA, Messenger/analysis , Transcriptional Activation
15.
Curr Mol Med ; 1(1): 25-44, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11899241

ABSTRACT

Glycogen storage disease type 1 (GSD-1), also known as von Gierke disease, is a group of autosomal recessive metabolic disorders caused by deficiencies in the activity of the glucose-6-phosphatase (G6Pase) system that consists of at least two membrane proteins, glucose-6-phosphate transporter (G6PT) and G6Pase. G6PT translocates glucose-6-phosphate (G6P) from cytoplasm to the lumen of the endoplasmic reticulum (ER) and G6Pase catalyzes the hydrolysis of G6P to produce glucose and phosphate. Therefore, G6PT and G6Pase work in concert to maintain glucose homeostasis. Deficiencies in G6Pase and G6PT cause GSD-1a and GSD-1b, respectively. Both manifest functional G6Pase deficiency characterized by growth retardation, hypoglycemia, hepatomegaly, kidney enlargement, hyperlipidemia, hyperuricemia, and lactic acidemia. GSD-1b patients also suffer from chronic neutropenia and functional deficiencies of neutrophils and monocytes, resulting in recurrent bacterial infections as well as ulceration of the oral and intestinal mucosa. The G6Pase gene maps to chromosome 17q21 and encodes a 36-kDa glycoprotein that is anchored to the ER by 9 transmembrane helices with its active site facing the lumen. Animal models of GSD-1a have been developed and are being exploited to delineate the disease more precisely and to develop new therapies. The G6PT gene maps to chromosome 11q23 and encodes a 37-kDa protein that is anchored to the ER by 10 transmembrane helices. A functional assay for the recombinant G6PT protein has been established, which showed that G6PT functions as a G6P transporter in the absence of G6Pase. However, microsomal G6P uptake activity was markedly enhanced in the simultaneous presence of G6PT and G6Pase. The cloning of the G6PT gene now permits animal models of GSD-1b to be generated. These recent developments are increasing our understanding of the GSD-l disorders and the G6Pase system, knowledge that will facilitate the development of novel therapeutic approaches for these disorders.


Subject(s)
Glucose-6-Phosphatase/metabolism , Glycogen Storage Disease Type I/enzymology , Glycogen Storage Disease Type I/physiopathology , Amino Acid Sequence , Animals , Binding Sites , Disease Models, Animal , Glucose-6-Phosphatase/chemistry , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/epidemiology , Glycogen Storage Disease Type I/therapy , Humans , Mice , Mice, Knockout , Models, Biological , Molecular Sequence Data , Molecular Structure , Mutation , Neutropenia/physiopathology , Protein Structure, Secondary , Structure-Activity Relationship , Tissue Distribution
16.
J Biol Chem ; 275(44): 34280-6, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10940311

ABSTRACT

Deficiencies in glucose 6-phosphate (G6P) transporter (G6PT), a 10-helical endoplasmic reticulum transmembrane protein of 429 amino acids, cause glycogen storage disease type 1b. To date, only three missense mutations in G6PT have been shown to abolish microsomal G6P transport activity. Here, we report the results of structure-function studies on human G6PT and demonstrate that 15 missense mutations and a codon deletion (delta F93) mutation abolish microsomal G6P uptake activity and that two splicing mutations cause exon skipping. While most missense mutants support the synthesis of G6PT protein similar to that of the wild-type transporter, immunoblot analysis shows that G20D, delta F93, and I278N mutations, located in helix 1, 2, and 6, respectively, destabilize the G6PT. Further, we demonstrate that G6PT mutants lacking an intact helix 10 are misfolded and undergo degradation within cells. Moreover, amino acids 415-417 in the cytoplasmic tail of the carboxyl-domain, extending from helix 10, also play a critical role in the correct folding of the transporter. However, the last 12 amino acids of the cytoplasmic tail play no essential role(s) in functional integrity of the G6PT. Our results, for the first time, elucidate the structural requirements for the stability and transport activity of the G6PT protein.


Subject(s)
Phosphotransferases/metabolism , Amino Acid Sequence , Animals , Antiporters , Base Sequence , COS Cells , Cytoplasm/metabolism , DNA Primers , Enzyme Stability , Humans , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Molecular Sequence Data , Monosaccharide Transport Proteins , Mutation , Phosphotransferases/chemistry , Phosphotransferases/genetics , Polymorphism, Single-Stranded Conformational , Protein Conformation , Protein Folding
17.
J Biol Chem ; 275(24): 18489-94, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10764804

ABSTRACT

[(18)F]-2-Fluoro-2-deoxyglucose (FDG) is a glucose analog currently utilized for positron emission tomography imaging studies in humans. FDG taken up by the liver is rapidly released. This property is attributed to elevated glucose-6-phosphatase (Glc-6-Pase) activity. To characterize this issue we studied the relationship between Glc-6-Pase activity and FDG release kinetics in a cell culture system. We overexpressed the Glc-6-Pase catalytic unit in a Glc-6-Pase-deficient mouse hepatocyte (Ho-15) and in A431 tumor cell lines. Glc-6-Pase enzyme activity and FDG release rates were determined in cells transfected with the Glc-6-Pase gene (Ho-15-D3 and A431-AC3), in mock-transfected cells of both cell lines, and in wild-type mouse hepatocytes (WT10) as control. Although the highest level of Glc-6-Pase activity was measured in A431-AC3, Ho-15-D3 cells showed much faster FDG release rates. The faster FDG release correlated with the level of glucose 6-phosphate transporter (Glc-6-PT) mRNA, which was found to be expressed at higher levels in Ho-15 compared with A431 cells. Overexpression of Glc-6-PT in A431-AC3 produced a dramatic increase in FDG release compared with control cells. This study gives the first direct evidence that activity of the Glc-6-Pase complex can be quantified in vivo by measuring FDG release. Adequate levels of Glc-6-Pase catalytic unit and Glc-6-PT are required for this function. FDG-positron emission tomography may be utilized to evaluate functional status of the Glc-6-Pase complex.


Subject(s)
Fluorodeoxyglucose F18/metabolism , Glucose-6-Phosphatase/metabolism , Animals , Blotting, Northern , Catalytic Domain , Cells, Cultured , Glucose-6-Phosphatase/genetics , Kinetics , Liver/metabolism , Mice , Oligopeptides , Peptides/genetics , Peptides/metabolism , Plasmids , RNA, Messenger/metabolism , Transfection
18.
Am J Hum Genet ; 66(2): 347-55, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10677294

ABSTRACT

Methionine adenosyltransferase (MAT) I/III deficiency, caused by mutations in the MAT1A gene, is characterized by persistent hypermethioninemia without elevated homocysteine or tyrosine. Clinical manifestations are variable and poorly understood, although a number of individuals with homozygous null mutations in MAT1A have neurological problems, including brain demyelination. We analyzed MAT1A in seven hypermethioninemic individuals, to provide insight into the relationship between genotype and phenotype. We identified six novel mutations and demonstrated that mutations resulting in high plasma methionines may signal clinical difficulties. Two patients-a compound heterozygote for truncating and severely inactivating missense mutations and a homozygote for an aberrant splicing MAT1A mutation-have plasma methionine in the 1,226-1,870 microM range (normal 5-35 microM) and manifest abnormalities of the brain gray matter or signs of brain demyelination. Another compound heterozygote for truncating and inactivating missense mutations has 770-1,240 microM plasma methionine and mild cognitive impairment. Four individuals carrying either two inactivating missense mutations or the single-allelic R264H mutation have 105-467 microM plasma methionine and are clinically unaffected. Our data underscore the necessity of further studies to firmly establish the relationship between genotypes in MAT I/III deficiency and clinical phenotypes, to elucidate the molecular bases of variability in manifestations of MAT1A mutations.


Subject(s)
Metabolism, Inborn Errors/genetics , Methionine Adenosyltransferase/deficiency , Methionine Adenosyltransferase/genetics , Mutation/genetics , Adolescent , Adult , Alleles , Alternative Splicing/genetics , Brain/abnormalities , Brain/metabolism , Brain/pathology , Brain/physiopathology , Child , Child, Preschool , Exons/genetics , Female , Genes, Dominant/genetics , Genes, Recessive/genetics , Humans , Infant , Infant, Newborn , Introns/genetics , Male , Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/physiopathology , Methionine/blood , Methionine/metabolism , Methionine Adenosyltransferase/metabolism , Mutation, Missense/genetics , Pedigree , Phenotype , Polymorphism, Single-Stranded Conformational , RNA, Messenger/analysis , RNA, Messenger/genetics
19.
Pharmacol Ther ; 85(1): 1-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10674710

ABSTRACT

Hepatic methionine adenosyltransferase (MAT) deficiency is caused by mutations in the human MAT1A gene that abolish or reduce hepatic MAT activity that catalyzes the synthesis of S-adenosylmethionine from methionine and ATP. This genetic disorder is characterized by isolated persistent hypermethioninemia in the absence of cystathionine beta-synthase deficiency, tyrosinemia, or liver disease. Depending on the nature of the genetic defect, hepatic MAT deficiency can be transmitted either as an autosomal recessive or dominant trait. Genetic analyses have revealed that mutations identified in the MAT1A gene only partially inactivate enzymatic activity, which is consistent with the fact that most hepatic MAT-deficient individuals are clinically well. Two hypermethioninemic individuals with null MAT1A mutations have developed neurological problems, including brain demyelination, although this correlation is by no means absolute. Presently, it is recommended that a DNA-based diagnosis should be performed for isolated hypermethioninemic individuals with unusually high plasma methionine levels to assess if therapy aimed at the prevention of neurological manifestations is warranted.


Subject(s)
Isoenzymes/genetics , Liver/enzymology , Methionine Adenosyltransferase/deficiency , Mutation/genetics , Adenosine Triphosphate/metabolism , Humans , Methionine/metabolism , Methionine Adenosyltransferase/genetics , Methionine Adenosyltransferase/physiology , S-Adenosylmethionine/biosynthesis
20.
J Biol Chem ; 275(2): 828-32, 2000 Jan 14.
Article in English | MEDLINE | ID: mdl-10625614

ABSTRACT

Glycogen storage disease type 1a (GSD-1a), characterized by hypoglycemia, liver and kidney enlargement, growth retardation, hyperlipidemia, and hyperuricemia, is caused by a deficiency in glucose-6-phosphatase (G6Pase), a key enzyme in glucose homeostasis. To evaluate the feasibility of gene replacement therapy for GSD-1a, we have infused adenoviral vector containing the murine G6Pase gene (Ad-mG6Pase) into G6Pase-deficient (G6Pase(-/-)) mice that manifest symptoms characteristic of human GSD-1a. Whereas <15% of G6Pase(-/-) mice under glucose therapy survived weaning, a 100% survival rate was achieved when G6Pase(-/-) mice were infused with Ad-mG6Pase, 90% of which lived to 3 months of age. Hepatic G6Pase activity in Ad-mG6Pase-infused mice was restored to 19% of that in G6Pase(+/+) mice at 7-14 days post-infusion; the activity persisted for at least 70 days. Ad-mG6Pase infusion also greatly improved growth of G6Pase(-/-) mice and normalized plasma glucose, cholesterol, triglyceride, and uric acid profiles. Furthermore, liver and kidney enlargement was less pronounced with near-normal levels of glycogen depositions in both organs. Our data demonstrate that a single administration of a recombinant adenoviral vector can alleviate the pathological manifestations of GSD-1a in mice, suggesting that this disorder in humans can potentially be corrected by gene therapy.


Subject(s)
Genetic Therapy , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/therapy , Liver/metabolism , Adenoviridae , Animals , Disease Models, Animal , Genetic Vectors , Glucose-6-Phosphatase/metabolism , Glucose-6-Phosphate/metabolism , Humans , Mice , Mice, Knockout , Microsomes, Liver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL