Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
2.
Mol Med ; 21: 487-95, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26062020

ABSTRACT

Acute intermittent porphyria (AIP) is an autosomal-dominant hepatic disorder caused by the half-normal activity of hydroxymethylbilane (HMB) synthase. Symptomatic individuals experience life-threatening acute neurovisceral attacks that are precipitated by factors that induce the hepatic expression of 5-aminolevulinic acid synthase 1 (ALAS1), resulting in the marked accumulation of the putative neurotoxic porphyrin precursors 5-aminolevulinic acid (ALA) and porphobilinogen (PBG). Here, we provide the first detailed description of the biochemical and pathologic alterations in the explanted liver of an AIP patient who underwent orthotopic liver transplantation (OLT) due to untreatable and debilitating chronic attacks. After OLT, the recipient's plasma and urinary ALA and PBG rapidly normalized, and her attacks immediately stopped. In the explanted liver, (a) ALAS1 mRNA and activity were elevated approximately ~3- and 5-fold, and ALA and PBG concentrations were increased ~3- and 1,760-fold, respectively; (b) uroporphyrin III concentration was elevated; (c) microsomal heme content was sufficient, and representative cytochrome P450 activities were essentially normal; (d) HMB synthase activity was approximately half-normal (~42%); (e) iron concentration was slightly elevated; and (f) heme oxygenase I mRNA was increased approximately three-fold. Notable pathologic findings included nodular regenerative hyperplasia, previously not reported in AIP livers, and minimal iron deposition, despite the large number of hemin infusions received before OLT. These findings suggest that the neurovisceral symptoms of AIP are not associated with generalized hepatic heme deficiency and support the neurotoxicity of ALA and/or PBG. Additionally, they indicate that substrate inhibition of hepatic HMB synthase activity by PBG is not a pathogenic mechanism in acute attacks.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Hydroxymethylbilane Synthase/biosynthesis , Liver/metabolism , Porphyria, Acute Intermittent/genetics , 5-Aminolevulinate Synthetase/biosynthesis , Adult , Aminolevulinic Acid/blood , Aminolevulinic Acid/urine , Female , Heme/metabolism , Humans , Hydroxymethylbilane Synthase/antagonists & inhibitors , Liver/pathology , Liver Transplantation , Porphobilinogen/blood , Porphobilinogen/urine , Porphyria, Acute Intermittent/enzymology , Porphyria, Acute Intermittent/pathology , RNA, Messenger/biosynthesis , Uroporphyrins/metabolism
3.
J Clin Invest ; 125(4): 1665-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25705881

ABSTRACT

Macrocytic anemia with abnormal erythropoiesis is a common feature of megaloblastic anemias, congenital dyserythropoietic anemias, and myelodysplastic syndromes. Here, we characterized a family with multiple female individuals who have macrocytic anemia. The proband was noted to have dyserythropoiesis and iron overload. After an extensive diagnostic evaluation that did not provide insight into the cause of the disease, whole-exome sequencing of multiple family members revealed the presence of a mutation in the X chromosomal gene ALAS2, which encodes 5'-aminolevulinate synthase 2, in the affected females. We determined that this mutation (Y365C) impairs binding of the essential cofactor pyridoxal 5'-phosphate to ALAS2, resulting in destabilization of the enzyme and consequent loss of function. X inactivation was not highly skewed in wbc from the affected individuals. In contrast, and consistent with the severity of the ALAS2 mutation, there was a complete skewing toward expression of the WT allele in mRNA from reticulocytes that could be recapitulated in primary erythroid cultures. Together, the results of the X inactivation and mRNA studies illustrate how this X-linked dominant mutation in ALAS2 can perturb normal erythropoiesis through cell-nonautonomous effects. Moreover, our findings highlight the value of whole-exome sequencing in diagnostically challenging cases for the identification of disease etiology and extension of the known phenotypic spectrum of disease.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Anemia, Dyserythropoietic, Congenital/genetics , Anemia, Macrocytic/genetics , Erythropoiesis/genetics , Genetic Diseases, X-Linked/genetics , Mutation, Missense , Point Mutation , 5-Aminolevulinate Synthetase/metabolism , Adult , Cells, Cultured , Exome/genetics , Female , Genes, Dominant , Genes, X-Linked , Genetic Diseases, X-Linked/blood , Hemorrhage/etiology , Humans , Iron Overload/etiology , Male , Models, Molecular , Molecular Sequence Data , Pregnancy , Pregnancy Complications, Hematologic/genetics , Protein Binding , Protein Conformation , Puerperal Disorders/etiology , Pyridoxal Phosphate/metabolism , RNA, Messenger/genetics , Reticulocytes/metabolism , X Chromosome Inactivation
4.
Proc Natl Acad Sci U S A ; 111(21): 7777-82, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24821812

ABSTRACT

The acute hepatic porphyrias are inherited disorders of heme biosynthesis characterized by life-threatening acute neurovisceral attacks. Factors that induce the expression of hepatic 5-aminolevulinic acid synthase 1 (ALAS1) result in the accumulation of the neurotoxic porphyrin precursors 5-aminolevulinic acid (ALA) and porphobilinogen (PBG), which recent studies indicate are primarily responsible for the acute attacks. Current treatment of these attacks involves i.v. administration of hemin, but a faster-acting, more effective, and safer therapy is needed. Here, we describe preclinical studies of liver-directed small interfering RNAs (siRNAs) targeting Alas1 (Alas1-siRNAs) in a mouse model of acute intermittent porphyria, the most common acute hepatic porphyria. A single i.v. dose of Alas1-siRNA prevented the phenobarbital-induced biochemical acute attacks for approximately 2 wk. Injection of Alas1-siRNA during an induced acute attack significantly decreased plasma ALA and PBG levels within 8 h, more rapidly and effectively than a single hemin infusion. Alas1-siRNA was well tolerated and a therapeutic dose did not cause hepatic heme deficiency. These studies provide proof-of-concept for the clinical development of RNA interference therapy for the prevention and treatment of the acute attacks of the acute hepatic porphyrias.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Liver/metabolism , Porphyria, Acute Intermittent/prevention & control , RNA Interference/immunology , RNA, Small Interfering/pharmacology , 5-Aminolevulinate Synthetase/genetics , Analysis of Variance , Animals , Blotting, Western , Drug Evaluation, Preclinical , Electrophoresis, Polyacrylamide Gel , Female , Mice , Mice, Inbred C57BL , Particle Size , RNA Interference/drug effects , Real-Time Polymerase Chain Reaction
5.
Exp Hematol ; 40(6): 477-86.e1, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22269113

ABSTRACT

Erythroid-specific 5-aminolevulinate synthase (ALAS2) is essential for hemoglobin production, and a loss-of-function mutation of ALAS2 gene causes X-linked sideroblastic anemia. Human ALAS2 protein consists of 587 amino acids and its carboxyl(C)-terminal region of 33 amino acids is conserved in higher eukaryotes, but is not present in prokaryotic ALAS. We explored the role of this C-terminal region in the pathogenesis of X-linked sideroblastic anemia. In vitro enzymatic activity was measured using bacterially expressed recombinant proteins. In vivo catalytic activity was evaluated by comparing the accumulation of porphyrins in eukaryotic cells stably expressing each mutant ALAS2 tagged with FLAG, and the half-life of each FLAG-tagged ALAS2 protein was determined by Western blot analysis. Two novel mutations (Val562Ala and Met567Ile) were identified in patients with X-linked sideroblastic anemia. Val562Ala showed the higher catalytic activity in vitro, but a shorter half-life in vivo compared to those of wild-type ALAS2 (WT). In contrast, the in vitro activity of Met567Ile mutant was about 25% of WT, while its half-life was longer than that of WT. However, in vivo catalytic activity of each mutant was lower than that of WT. In addition, the deletion of 33 amino acids at C-terminal end resulted in higher catalytic activity both in vitro and in vivo with the longer half-life compared to WT. In conclusion, the C-terminal region of ALAS2 protein may function as an intrinsic modifier that suppresses catalytic activity and increases the degradation of its protein, each function of which is enhanced by the Met567Ile mutation and the Val562Ala mutation, respectively.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/genetics , Adolescent , Adult , Base Sequence , Biocatalysis , Blotting, Western , DNA Primers , Enzyme Stability , Humans , Male , Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...