Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Cell ; 172(5): 952-965.e18, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474921

ABSTRACT

Viruses that are typically benign sometimes invade the brainstem in otherwise healthy children. We report bi-allelic DBR1 mutations in unrelated patients from different ethnicities, each of whom had brainstem infection due to herpes simplex virus 1 (HSV1), influenza virus, or norovirus. DBR1 encodes the only known RNA lariat debranching enzyme. We show that DBR1 expression is ubiquitous, but strongest in the spinal cord and brainstem. We also show that all DBR1 mutant alleles are severely hypomorphic, in terms of expression and function. The fibroblasts of DBR1-mutated patients contain higher RNA lariat levels than control cells, this difference becoming even more marked during HSV1 infection. Finally, we show that the patients' fibroblasts are highly susceptible to HSV1. RNA lariat accumulation and viral susceptibility are rescued by wild-type DBR1. Autosomal recessive, partial DBR1 deficiency underlies viral infection of the brainstem in humans through the disruption of tissue-specific and cell-intrinsic immunity to viruses.


Subject(s)
Brain Diseases, Metabolic, Inborn/genetics , Brain Stem/metabolism , Brain Stem/virology , RNA/chemistry , RNA/metabolism , Alleles , Amino Acid Sequence , Animals , Brain Diseases, Metabolic, Inborn/pathology , Brain Stem/pathology , Encephalitis, Viral/genetics , Escherichia coli/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Herpesvirus 1, Human , Humans , Interferons/metabolism , Introns/genetics , Male , Mice , Mutant Proteins/metabolism , Mutation/genetics , Open Reading Frames/genetics , Pedigree , RNA Nucleotidyltransferases/chemistry , RNA Nucleotidyltransferases/deficiency , RNA Nucleotidyltransferases/genetics , Toll-Like Receptor 3/metabolism , Virus Replication
2.
J Biol Chem ; 291(39): 20487-502, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27496950

ABSTRACT

Human NADPH-cytochrome P450 oxidoreductase (POR) gene mutations are associated with severe skeletal deformities and disordered steroidogenesis. The human POR mutation A287P presents with disordered sexual development and skeletal malformations. Difficult recombinant expression and purification of this POR mutant suggested that the protein was less stable than WT. The activities of cytochrome P450 17A1, 19A1, and 21A2, critical in steroidogenesis, were similar using our purified, full-length, unmodified A287P or WT POR, as were those of several xenobiotic-metabolizing cytochromes P450, indicating that the A287P protein is functionally competent in vitro, despite its functionally deficient phenotypic behavior in vivo Differential scanning calorimetry and limited trypsinolysis studies revealed a relatively unstable A287P compared with WT protein, leading to the hypothesis that the syndrome observed in vivo results from altered POR protein stability. The crystal structures of the soluble domains of WT and A287P reveal only subtle differences between them, but these differences are consistent with the differential scanning calorimetry results as well as the differential susceptibility of A287P and WT observed with trypsinolysis. The relative in vivo stabilities of WT and A287P proteins were also examined in an osteoblast cell line by treatment with cycloheximide, a protein synthesis inhibitor, showing that the level of A287P protein post-inhibition is lower than WT and suggesting that A287P may be degraded at a higher rate. Current studies demonstrate that, unlike previously described mutations, A287P causes POR deficiency disorder due to conformational instability leading to proteolytic susceptibility in vivo, rather than through an inherent flavin-binding defect.


Subject(s)
Antley-Bixler Syndrome Phenotype , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Mutation, Missense , Amino Acid Substitution , Antley-Bixler Syndrome Phenotype/enzymology , Antley-Bixler Syndrome Phenotype/genetics , Crystallography, X-Ray , Cytochrome P-450 Enzyme System/metabolism , Enzyme Stability/genetics , Humans
3.
Arch Biochem Biophys ; 513(1): 42-50, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21741353

ABSTRACT

Genetic variations in POR, encoding NADPH-cytochrome P450 oxidoreductase (CYPOR), can diminish the function of numerous cytochromes P450, and also have the potential to block degradation of heme by heme oxygenase-1 (HO-1). Purified full-length human CYPOR, HO-1, and biliverdin reductase were reconstituted in lipid vesicles and assayed for NADPH-dependent conversion of heme to bilirubin. Naturally-occurring human CYPOR variants queried were: WT, A115V, Y181D, P228L, M263V, A287P, R457H, Y459H, and V492E. All CYPOR variants exhibited decreased bilirubin production relative to WT, with a lower apparent affinity of the CYPOR-HO-1 complex than WT. Addition of FMN or FAD partially restored the activities of Y181D, Y459H, and V492E. When mixed with WT CYPOR, only the Y181D CYPOR variant inhibited heme degradation by sequestering HO-1, whereas Y459H and V492E were unable to inhibit HO-1 activity suggesting that CYPOR variants might have differential binding affinities with redox partners. Titrating the CYPOR-HO-1 complex revealed that the optimal CYPOR:HO-1 ratio for activity was 1:2, lending evidence in support of productive HO-1 oligomerization, with higher ratios of CYPOR:HO-1 showing decreased activity. In conclusion, human POR mutations, shown to impact P450 activities, also result in varying degrees of diminished HO-1 activity, which may further complicate CYPOR deficiency.


Subject(s)
Heme Oxygenase-1/chemistry , Multienzyme Complexes/chemistry , Mutation, Missense , NADPH-Ferrihemoprotein Reductase/chemistry , NADPH-Ferrihemoprotein Reductase/genetics , Protein Multimerization , Flavin Mononucleotide/chemistry , Flavin Mononucleotide/genetics , Flavin Mononucleotide/metabolism , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/genetics , Flavin-Adenine Dinucleotide/metabolism , Heme/chemistry , Heme/genetics , Heme/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Oxidoreductases Acting on CH-CH Group Donors/chemistry , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism
4.
Drug Metab Dispos ; 38(2): 332-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19884324

ABSTRACT

Patients with congenital adrenal hyperplasia, exhibiting combined CYP17 and CYP21 deficiency, were shown by Arlt et al. (2004) to harbor a 541T-->G mutation in exon 5 of POR (encoding NADPH-cytochrome P450 reductase, CYPOR), which resulted in a Y181D substitution that obliterated electron transfer capacity. Using bacterial expression models, we examined catalytic and physical properties of the human CYPOR Y181D variant. As purified, Y181D lacked flavin mononucleotide (FMN) and NADPH-cytochrome c reductase (NCR) activity but retained normal flavin adenine dinucleotide binding and NADPH utilization. Titration of the purified protein with FMN restored 64 of wild-type (WT) NCR activity in Y181D with an activation constant of approximately 2 microM. As determined by FMN fluorescence quenching, Y181D had K(d)(FMN) = 7.3 microM. Biplasmid coexpression of CYPOR and CYP1A2, at the physiological ratio of approximately 1:10 in the engineered MK_1A2_POR Escherichia coli strain, showed the compromised capacity of Y181D to support CYP1A2-catalyzed metabolism of the procarcinogens 2-aminoanthracene, 2-amino-3-methylimidazo(4,5-f)quinoline, and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Isolated MK1A2_POR membranes confirmed FMN stimulation of Y181D NCR activity with a 1.6 microM activation constant. CYP1A2 ethoxyresorufin-O-dealkylase activity of the MK1A2_POR(Y181D) membranes, undetectable in the absence of added FMN, increased to 37% of MK1A2_POR(WT) membranes with a 1.2 microM FMN activation constant. Therefore, we conclude that compromised FMN binding is the specific molecular defect causing POR deficiency in patients with Y181D mutation and that this defect, in large part, can be overcome in vitro by FMN addition.


Subject(s)
Flavin Mononucleotide/metabolism , NADPH-Ferrihemoprotein Reductase/deficiency , Amino Acid Substitution , Carcinogens/metabolism , Circular Dichroism , Cytochrome P-450 CYP1A2/chemistry , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Escherichia coli , Flavin Mononucleotide/chemistry , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Humans , Kinetics , Membranes/metabolism , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , NADH, NADPH Oxidoreductases/metabolism , NADPH-Ferrihemoprotein Reductase/chemistry , NADPH-Ferrihemoprotein Reductase/genetics , NADPH-Ferrihemoprotein Reductase/metabolism , Plasmids , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Spectrometry, Fluorescence , Spectrophotometry
5.
Free Radic Biol Med ; 110: 261-269, 2017 09.
Article in English | MEDLINE | ID: mdl-28666850

ABSTRACT

Previously published studies strongly suggested that insulin- and exercise-induced skeletal muscle glucose uptake require nitric oxide (NO) production. However, the signal transduction mechanisms by which insulin and contraction regulated NO production and subsequent glucose transport are not known. In the present study, we utilized the myotube cell lines treated with insulin or hydrogen peroxide, the latter to mimic contraction-induced oxidative stress, to characterize these mechanisms. We found that insulin stimulation of neuronal nitric oxide synthase (nNOS) phosphorylation, NO production, and GLUT4 translocation were all significantly reduced by inhibition of either nNOS or Akt2. Hydrogen peroxide (H2O2) induced phosphorylation of nNOS at the same residue as did insulin, and also stimulated NO production and GLUT4 translocation. nNOS inhibition prevented H2O2-induced GLUT4 translocation. AMP activated protein kinase (AMPK) inhibition prevented H2O2 activation and phosphorylation of nNOS, leading to reduced NO production and significantly attenuated GLUT4 translocation. We conclude that nNOS phosphorylation and subsequently increased NO production are required for both insulin- and H2O2-stimulated glucose transport. Although the two stimuli result in phosphorylation of the same residue on nNOS, they do so through distinct protein kinases. Thus, insulin and H2O2-activated signaling pathways converge on nNOS, which is a common mediator of glucose uptake in both pathways. However, the fact that different kinases are utilized provides a basis for the use of exercise to activate glucose transport in the face of insulin resistance.


Subject(s)
Glucose/metabolism , Hydrogen Peroxide/pharmacology , Insulin/pharmacology , Muscle Fibers, Skeletal/drug effects , Nitric Oxide Synthase Type I/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Insulin Resistance , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress , Phosphorylation , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL