Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
2.
Nat Commun ; 11(1): 2813, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32499479

ABSTRACT

5'-aminolevulinate synthase (ALAS) catalyzes the first step in heme biosynthesis, generating 5'-aminolevulinate from glycine and succinyl-CoA. Inherited frameshift indel mutations of human erythroid-specific isozyme ALAS2, within a C-terminal (Ct) extension of its catalytic core that is only present in higher eukaryotes, lead to gain-of-function X-linked protoporphyria (XLP). Here, we report the human ALAS2 crystal structure, revealing that its Ct-extension folds onto the catalytic core, sits atop the active site, and precludes binding of substrate succinyl-CoA. The Ct-extension is therefore an autoinhibitory element that must re-orient during catalysis, as supported by molecular dynamics simulations. Our data explain how Ct deletions in XLP alleviate autoinhibition and increase enzyme activity. Crystallography-based fragment screening reveals a binding hotspot around the Ct-extension, where fragments interfere with the Ct conformational dynamics and inhibit ALAS2 activity. These fragments represent a starting point to develop ALAS2 inhibitors as substrate reduction therapy for porphyria disorders that accumulate toxic heme intermediates.


Subject(s)
5-Aminolevulinate Synthetase/chemistry , Gene Expression Regulation, Enzymologic , 5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/genetics , Acyl Coenzyme A/chemistry , Catalysis , Catalytic Domain , Crystallography, X-Ray , Genetic Diseases, X-Linked/genetics , Heme/chemistry , Humans , Kinetics , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Protein Domains , Protoporphyria, Erythropoietic/genetics , Substrate Specificity
4.
JAMA Dermatol ; 153(8): 789-796, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28614581

ABSTRACT

Importance: Autosomal recessive erythropoietic protoporphyria (EPP) and X-linked protoporphyria (XLP) are rare photodermatoses presenting with variable degrees of painful phototoxicity that markedly affects quality of life. The clinical variability, determinants of severity, and genotype/phenotype correlations of these diseases are not well characterized. Objective: To describe the baseline clinical characteristics, genotypes, and determinants of disease severity in a large patient cohort with EPP or XLP. Design, Setting, and Participants: A prospective observational study was conducted among patients with confirmed diagnoses of EPP or XLP from November 1, 2010, to December 6, 2015, at 6 academic medical centers of the Porphyrias Consortium of the National Institutes of Health Rare Diseases Clinical Research Network. Detailed medical histories, including history of phototoxicity and treatment, were collected on standardized case report forms. Patients underwent baseline laboratory testing, total erythrocyte protoporphyrin (ePPIX) testing, and molecular genetic testing. Data were entered into a centralized database. Main Outcomes and Measures: Results of biochemical and genetic tests were explored for association with clinical phenotype in patients with EPP or XLP. Results: Of the 226 patients in the study (113 female and 113 male patients; mean [SD] age, 36.7 [17.0] years), 186 (82.3%) had EPP with a FECH (OMIM 612386) mutation and the common low-expression FECH allele IVS3-48T>C, and only 1 patient had 2 FECH mutations. Twenty-two patients had XLP (9.7%; 10 male and 12 female patients), and 9 patients (4.0%) had elevated ePPIX levels and symptoms consistent with protoporphyria but no detectable mutation in the FECH or ALAS2 (OMIM 301300) gene. Samples of DNA could not be obtained from 8 patients. Patients' mean (SD) age at symptom onset was 4.4 (4.4) years. Anemia (107 [47.3%]), history of liver dysfunction (62 [27.4%]), and gallstones (53 [23.5%]) were commonly reported. Higher ePPIX levels were associated with earlier age of symptom onset (median ePPIX levels for those who developed symptoms before vs after 1 year of age, 1744 vs 1567 µg/dL; P = .02), less sun tolerance (median ePPIX levels for those reporting symptoms before vs after 10 minutes of sun exposure, 2233 vs 1524 µg/dL; P ≤ .001), and increased risk of liver dysfunction (median ePPIX levels for those with liver dysfunction vs normal liver function, 2016 vs 1510 µg/dL; P = .003). Patients with EPP and FECH missense mutations had significantly lower ePPIX levels than those with other mutations (1462 vs 1702 µg/dL; P = .01). Male patients with XLP had significantly higher ePPIX levels, on average, than did patients with EPP (3574 vs 1669 µg/dL; P < .001). Marked clinical variability was seen in female patients with XLP owing to random X-chromosomal inactivation. Conclusions and Relevance: These data suggest that higher ePPIX levels are a major determinant of disease severity and risk of liver dysfunction in patients with EPP or XLP. These findings provide a framework for clinical monitoring and management of these disorders.


Subject(s)
5-Aminolevulinate Synthetase/deficiency , Ferrochelatase/genetics , Genetic Diseases, X-Linked/physiopathology , Protoporphyria, Erythropoietic/physiopathology , Quality of Life , 5-Aminolevulinate Synthetase/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Genetic Diseases, X-Linked/genetics , Genotype , Humans , Liver Diseases/genetics , Liver Diseases/physiopathology , Male , Middle Aged , Mutation , Phenotype , Prospective Studies , Protoporphyria, Erythropoietic/genetics , Severity of Illness Index , Young Adult
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(2): 428-439, 2017 02.
Article in English | MEDLINE | ID: mdl-27838491

ABSTRACT

Mutations in the C-terminus of human erythroid 5-aminolevulinate synthase (hALAS2), a pyridoxal 5'-phosphate (PLP)-dependent enzyme, are associated with two different blood disorders, X-linked sideroblastic anemia (XLSA) and X-linked protoporphyria (XLPP). XLSA-causing mutations yield hALAS2 variants with decreased activity, while XLPP-causing mutations result in a gain-of-function of hALAS2. There are no specific treatments for XLPP. Isonicotinic acid hydrazide (isoniazid, INH), an antituberculosis agent, can cause sideroblastic anemia as a side-effect, by limiting PLP availability to hALAS2, via inhibition of pyridoxal kinase or reaction with pyridoxal to form pyridoxal isonicotinoyl hydrazone. We hypothesized that INH also binds and directly inhibits hALAS2. Using fluorescence-activated cell sorting and confocal fluorescence microscopy, we demonstrate that INH reduces protoporphyrin IX levels in HeLa cells expressing either wild-type hALAS2 or XLPP variants. In addition, PLP and pyridoxamine 5'-phosphate (PMP) reversed the cellular inhibition of hALAS2 activity by INH. Steady-state kinetic analyses with purified hALAS2 indicated that INH directly inhibits the enzyme, noncompetitively or uncompetitively, with an apparent Ki of 1.2µM. Circular dichroism spectroscopy revealed that INH triggered tertiary structural changes in hALAS2 that altered the microenvironment of the PLP cofactor and hampered the association of PLP with apo-hALAS2. Treatment of four XLPP patients with INH (5mg·kg-1·day-1) over a six-month period was well tolerated but without statistically significant modification of PPIX levels. These results, taken together, permit us to further an INH inhibition kinetic mechanism for ALAS, which suggests the possible use of INH-derived drugs in treating patients with XLPP and potentially other protoporphyrin-accumulating porphyrias.


Subject(s)
5-Aminolevulinate Synthetase/deficiency , Enzyme Inhibitors/pharmacology , Genetic Diseases, X-Linked/drug therapy , Isoniazid/pharmacology , Protoporphyria, Erythropoietic/drug therapy , 5-Aminolevulinate Synthetase/antagonists & inhibitors , 5-Aminolevulinate Synthetase/blood , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/metabolism , Anemia, Sideroblastic/enzymology , Enzyme Inhibitors/therapeutic use , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/enzymology , HeLa Cells , Humans , Isoniazid/therapeutic use , Protein Binding/drug effects , Protein Structure, Tertiary/drug effects , Protoporphyria, Erythropoietic/blood , Protoporphyria, Erythropoietic/enzymology , Protoporphyrins/blood , Pyridoxal Phosphate/metabolism , Pyridoxine/pharmacology , Vitamin B Complex/pharmacology
6.
J Dermatol ; 43(4): 414-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26387792

ABSTRACT

A 12-year-old boy with photosensitivity since 3 years of age presented with small concavities on both cheeks, the nasal root and the dorsal surface of both hands. According to the clinical features, erythropoietic protoporphyria (EPP) was suspected. Urine and blood samples were tested for porphyrin derivatives, which revealed a markedly elevated level of erythrocyte protoporphyrin (EP) and a diagnosis of EPP was made. The patient's mother had no photosensitivity, however, lesions appearing slightly as small scars were found on the dorsum of her right hand; his elder sister and father showed no rash. The EP levels were elevated in samples from his mother and mildly elevated in those from his elder sister and father. To obtain a definitive diagnosis, genetic analyses were performed using samples from all family members, which revealed no mutations in the ferrochelatase-encoding gene (FECH), which is responsible for EPP. Instead, a pathological mutation of the 5-aminolevulinic acid synthase-encoding gene (ALAS2) was identified in samples from the patient, his mother and his elder sister, confirming a definitive diagnosis of X-linked dominant protoporphyria (XLDPP). This is the first Japanese family reported to have XLDPP, demonstrating evidence of the condition in Japan. In addition, because XLDPP is very similar to EPP in its clinical aspects and laboratory findings, a genetic analysis is required for the differential diagnosis.


Subject(s)
5-Aminolevulinate Synthetase/deficiency , Ferrochelatase/genetics , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/genetics , Protoporphyria, Erythropoietic/diagnosis , Protoporphyria, Erythropoietic/genetics , Protoporphyrins/analysis , 5-Aminolevulinate Synthetase/analysis , 5-Aminolevulinate Synthetase/genetics , Alleles , Cheek , Child , Diagnosis, Differential , Hand , Humans , Japan , Male , Mutation , Pedigree
7.
Biochemistry ; 54(36): 5617-31, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26300302

ABSTRACT

Regulation of 5-aminolevulinate synthase (ALAS) is at the origin of balanced heme production in mammals. Mutations in the C-terminal region of human erythroid-specific ALAS (hALAS2) are associated with X-linked protoporphyria (XLPP), a disease characterized by extreme photosensitivity, with elevated blood concentrations of free protoporphyrin IX and zinc protoporphyrin. To investigate the molecular basis for this disease, recombinant hALAS2 and variants of the enzyme harboring the gain-of-function XLPP mutations were constructed, purified, and analyzed kinetically, spectroscopically, and thermodynamically. Enhanced activities of the XLPP variants resulted from increases in the rate at which the product 5-aminolevulinate (ALA) was released from the enzyme. Circular dichroism spectroscopy revealed that the XLPP mutations altered the microenvironment of the pyridoxal 5'-phosphate cofactor, which underwent further and specific alterations upon succinyl-CoA binding. Transient kinetic analyses of the variant-catalyzed reactions and protein fluorescence quenching upon binding of ALA to the XLPP variants demonstrated that the protein conformational transition step associated with product release was predominantly affected. Of relevance is the fact that XLPP could also be modeled in cell culture. We propose that (1) the XLPP mutations destabilize the succinyl-CoA-induced hALAS2 closed conformation and thus accelerate ALA release, (2) the extended C-terminus of wild-type mammalian ALAS2 provides a regulatory role that allows for allosteric modulation of activity, thereby controlling the rate of erythroid heme biosynthesis, and (3) this control is disrupted in XLPP, resulting in porphyrin accumulation.


Subject(s)
5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/metabolism , Aminolevulinic Acid/metabolism , Genetic Diseases, X-Linked/enzymology , Protoporphyria, Erythropoietic/enzymology , Protoporphyrins/metabolism , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/genetics , Aminolevulinic Acid/chemistry , Enzyme Stability , Escherichia coli/cytology , Genetic Diseases, X-Linked/genetics , HeLa Cells , Hot Temperature , Humans , K562 Cells , Kinetics , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Protoporphyria, Erythropoietic/genetics , Protoporphyrins/chemistry , Thermodynamics
8.
J Biol Chem ; 289(50): 34827-37, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25352601

ABSTRACT

Heme is an essential cofactor for aerobic organisms. Its redox chemistry is central to a variety of biological functions mediated by hemoproteins. In blood stages, malaria parasites consume most of the hemoglobin inside the infected erythrocytes, forming nontoxic hemozoin crystals from large quantities of heme released during digestion. At the same time, the parasites possess a heme de novo biosynthetic pathway. This pathway in the human malaria parasite Plasmodium falciparum has been considered essential and is proposed as a potential drug target. However, we successfully disrupted the first and last genes of the pathway, individually and in combination. These knock-out parasite lines, lacking 5-aminolevulinic acid synthase and/or ferrochelatase (FC), grew normally in blood-stage culture and exhibited no changes in sensitivity to heme-related antimalarial drugs. We developed a sensitive LC-MS/MS assay to monitor stable isotope incorporation into heme from its precursor 5-[(13)C4]aminolevulinic acid, and this assay confirmed that de novo heme synthesis was ablated in FC knock-out parasites. Disrupting the FC gene also caused no defects in gametocyte generation or maturation but resulted in a greater than 70% reduction in male gamete formation and completely prevented oocyst formation in female Anopheles stephensi mosquitoes. Our data demonstrate that the heme biosynthesis pathway is not essential for asexual blood-stage growth of P. falciparum parasites but is required for mosquito transmission. Drug inhibition of pathway activity is therefore unlikely to provide successful antimalarial therapy. These data also suggest the existence of a parasite mechanism for scavenging host heme to meet metabolic needs.


Subject(s)
Anopheles/parasitology , Erythrocytes/parasitology , Heme/biosynthesis , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , 5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/genetics , Animals , Female , Ferrochelatase/genetics , Gene Knockout Techniques , Heme/metabolism , Humans , Male , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Tandem Mass Spectrometry
9.
Clin Exp Dermatol ; 39(1): 35-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24131146

ABSTRACT

X-linked dominant protoporphyria (XLDPP) was first reported in the genetics literature in 2008. It has a phenotype very similar to erythropoietic protoporphyria (EPP), but is distinguished from EPP by higher concentrations of erythrocyte protoporphyrin (of which a high proportion is zinc-chelated), its apparently higher incidence of liver disease, and an X-linked dominant pattern of inheritance. Dermatologists should understand how XLDPP differs from EPP, in order to advise newly diagnosed patients correctly about the genetic implications and the long-term management strategy. We present a case series of XLDPP to introduce this condition to the dermatology literature.


Subject(s)
5-Aminolevulinate Synthetase/deficiency , Genetic Diseases, X-Linked/diagnosis , Protoporphyria, Erythropoietic/diagnosis , 5-Aminolevulinate Synthetase/genetics , Adolescent , Female , Humans , Mutation , Pedigree , Protoporphyrins/blood
10.
Dermatology ; 227(3): 238-42, 2013.
Article in English | MEDLINE | ID: mdl-24135682

ABSTRACT

X-linked dominant protoporphyria (XLDPP) is a genetic disorder that affects the synthesis of the heme group due to an increase in delta-aminolaevulinate synthase 2 (ALAS2) enzyme activity. Moreover, annular elastolytic giant-cell granuloma (AEGCG) is a rare reactive granulomatous dermatosis, usually associated with actinic damage. An 86-year-old man presented with edematous-erythematous lesions in photoexposed areas of the face and on the dorsum of both hands. Protoporphyrin levels in serum and feces were significantly elevated and a heterozygous frameshift mutation in the exon 11 of the ALAS2 gene: c.1706-1709del (p.Glu569GlyfsX24) was identified. Concomitantly, we observed an annular plaque with raised borders on the back of his right hand, clinically and histologically compatible with a diagnosis of AEGCG. Skin lesions disappeared only upon use of a physical sunscreen. We report two rare photodermatoses in an elderly patient and discuss the significance of dermal elastic fiber damage induced by the XLDPP as a main triggering factor of AEGCG.


Subject(s)
5-Aminolevulinate Synthetase/deficiency , Facial Dermatoses/complications , Genetic Diseases, X-Linked/complications , Granuloma, Giant Cell/complications , Hand Dermatoses/complications , Photosensitivity Disorders/complications , Protoporphyria, Erythropoietic/complications , Aged, 80 and over , Feces/chemistry , Granuloma, Giant Cell/pathology , Humans , Male , Photosensitivity Disorders/drug therapy , Protoporphyrins/analysis , Protoporphyrins/blood , Sunscreening Agents/therapeutic use
11.
J Invest Dermatol ; 133(6): 1467-71, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23673504

ABSTRACT

Photosensitivity is the clinical hallmark of both erythropoietic protoporphyria (EPP) and X-linked dominant protoporphyria (XLDPP). Both disorders result from a hereditary dysfunction in heme biosynthesis. Disease onset is usually in early childhood. However, rare patients with late-onset EPP in association with a myeloproliferative disorder or myelodysplastic syndrome have been reported. In this issue, Livideanu et al. describe the first patient with late-onset XLDPP.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Genetic Diseases, X-Linked/genetics , Photosensitivity Disorders/genetics , Protoporphyria, Erythropoietic/genetics , 5-Aminolevulinate Synthetase/deficiency , Female , Humans , Male
12.
Mol Med ; 19: 18-25, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23348515

ABSTRACT

X-linked protoporphyria (XLP) (MIM 300752) is a recently recognized erythropoietic porphyria due to gain-of-function mutations in the erythroid-specific aminolevulinate synthase gene (ALAS2). Previously, two exon 11 small deletions, c.1699_1670ΔAT (ΔAT) and c.1706_1709ΔAGTG (ΔAGTG), that prematurely truncated or elongated the ALAS2 polypeptide, were reported to increase enzymatic activity 20- to 40-fold, causing the erythroid accumulation of protoporphyrins, cutaneous photosensitivity and liver disease. The mutant ΔAT and ΔAGTG ALAS2 enzymes, two novel mutations, c.1734ΔG (ΔG) and c.1642C>T (p.Q548X), and an engineered deletion c.1670-1671TC>GA p.F557X were expressed, and their purified enzymes were characterized. Wild-type and ΔAGTG enzymes exhibited similar amounts of 54- and 52-kDa polypeptides on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), whereas the ΔAT and p.F557X had only 52-kDa polypeptides. Compared to the purified wild-type enzyme, ΔAT, ΔAGTG and Q548X enzymes had increased specific activities that were only 1.8-, 3.1- and 1.6-fold, respectively. Interestingly, binding studies demonstrated that the increased activity Q548X enzyme did not bind to succinyl-CoA synthetase. The elongated ΔG enzyme had wild-type specific activity, kinetics and thermostability; twice the wild-type purification yield (56 versus 25%); and was primarily a 54-kDa form, suggesting greater stability in vivo. On the basis of studies of mutant enzymes, the maximal gain-of function region spanned 57 amino acids between 533 and 580. Thus, these ALAS2 gain-of-function mutations increased the specific activity (ΔAT, ΔAGTG and p.Q548X) or stability (ΔG) of the enzyme, thereby leading to the increased erythroid protoporphyrin accumulation causing XLP.


Subject(s)
5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Erythrocytes/enzymology , Genetic Diseases, X-Linked/genetics , Mutation , Protoporphyria, Erythropoietic/genetics , 5-Aminolevulinate Synthetase/deficiency , Enzyme Stability , Erythrocytes/metabolism , Female , Humans , Kinetics , Male , Temperature
13.
Hum Mol Genet ; 22(7): 1280-8, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23263862

ABSTRACT

Frameshift mutations in the last coding exon of the 5-aminolevulinate synthase (ALAS) 2 gene were described to activate the enzyme causing increased levels of zinc- and metal-free protoporphyrin in patients with X-linked dominant protoporphyria (XLDPP). Only two such so-called gain-of-function mutations have been reported since the description of XLDPP in 2008. In this study of four newly identified XLDPP families, we identified two novel ALAS2 gene mutations, a nonsense p.Q548X and a frameshift c.1651-1677del26bp, along with a known mutation (delAGTG) found in two unrelated families. Of relevance, a de novo somatic and germinal mosaicism was present in a delAGTG family. Such a phenomenon may explain the high proportion of this mutation in XLDPP worldwide. Enhancements of over 3- and 14-fold in the catalytic rate and specificity constant of purified recombinant XLDPP variants in relation to those of wild-type ALAS2 confirmed the gain of function ascribed to these enzymes. The fact that both p.Q548X and c.1651-1677del26bp are located in close proximity and upstream from the two previously described mutations led us to propose the presence of a large gain-of-function domain within the C-terminus of ALAS2. To test this hypothesis, we generated four additional nonsense mutants (p.A539X, p.G544X, p.G576X and p.V583X) surrounding the human XLDPP mutations and defined an ALAS2 gain-of-function domain with a minimal size of 33 amino acids. The identification of this gain-of-function domain provides important information on the enzymatic activity of ALAS2, which was proposed to be constitutively inhibited, either directly or indirectly, through its own C-terminus.


Subject(s)
5-Aminolevulinate Synthetase/genetics , Genetic Diseases, X-Linked/genetics , Protoporphyria, Erythropoietic/genetics , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/metabolism , Amino Acid Sequence , Base Sequence , Child, Preschool , Codon, Nonsense , DNA Mutational Analysis , Exons , Female , Frameshift Mutation , Genetic Association Studies , Genetic Diseases, X-Linked/enzymology , Humans , Infant , Kinetics , Molecular Sequence Data , Mosaicism , Mutagenesis, Site-Directed , Pedigree , Protein Structure, Tertiary , Protoporphyria, Erythropoietic/enzymology , Sequence Analysis, DNA , Young Adult
15.
Ann Hematol ; 92(1): 1-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22983749

ABSTRACT

Sideroblastic anemia is characterized by anemia with the emergence of ring sideroblasts in the bone marrow. There are two forms of sideroblastic anemia, i.e., congenital sideroblastic anemia (CSA) and acquired sideroblastic anemia. In order to clarify the pathophysiology of sideroblastic anemia, a nationwide survey consisting of clinical and molecular genetic analysis was performed in Japan. As of January 31, 2012, data of 137 cases of sideroblastic anemia, including 72 cases of myelodysplastic syndrome (MDS)-refractory cytopenia with multilineage dysplasia (RCMD), 47 cases of MDS-refractory anemia with ring sideroblasts (RARS), and 18 cases of CSA, have been collected. Hemoglobin and MCV level in CSA are significantly lower than those of MDS, whereas serum iron level in CSA is significantly higher than those of MDS. Of 14 CSA for which DNA was available for genetic analysis, 10 cases were diagnosed as X-linked sideroblastic anemia due to ALAS2 gene mutation. The mutation of SF3B1 gene, which was frequently mutated in MDS-RS, was not detected in CSA patients. Together with the difference of clinical data, it is suggested that genetic background, which is responsible for the development of CSA, is different from that of MDS-RS.


Subject(s)
Anemia, Sideroblastic/congenital , 5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Adolescent , Adult , Age of Onset , Aged , Anemia, Sideroblastic/blood , Anemia, Sideroblastic/classification , Anemia, Sideroblastic/epidemiology , Anemia, Sideroblastic/genetics , Child , Child, Preschool , Chromosome Aberrations , Female , Gene Frequency , Genes, X-Linked , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/genetics , Glutaredoxins/deficiency , Glutaredoxins/genetics , Health Surveys , Humans , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Infant , Infant, Newborn , Japan/epidemiology , Male , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Middle Aged , Mitochondrial Membrane Transport Proteins/deficiency , Mitochondrial Membrane Transport Proteins/genetics , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/genetics , Phosphoproteins/deficiency , Phosphoproteins/genetics , RNA Splicing Factors , Recombinant Fusion Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/deficiency , Ribonucleoprotein, U2 Small Nuclear/genetics , Treatment Outcome , Vitamin B 6/therapeutic use , Young Adult
16.
Biochem Biophys Res Commun ; 340(1): 105-10, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16356476

ABSTRACT

To identify erythroid-specific heme-regulated genes, we performed differential expression analysis between wild-type and heme-deficient erythroblasts, which had been prepared from wild-type and erythroid-specific delta-aminolevulinate synthase-null mouse ES cells, respectively. Among 8737 clones on cDNA array, 40 cDNA clones, including 34 unknown ESTs, were first selected by their high expression profiles in wild-type erythroblasts, and evaluated further for their erythroid-lineage specificity, expression in hematopoietic tissues in vivo, and heme-dependent expression, which yielded 11, 4, and 4 genes, respectively. Because of the selection strategy employed, the final 4 were considered as the newly identified erythroid-specific heme-regulated genes. These 4 genes were uncoupling protein 2, nucleolar spindle-associated protein, cellular nucleic acid-binding protein, and a novel acetyltransferase-like protein. These findings thus suggest that heme may regulate a wide variety of hitherto unrecognized genes, and further analysis of these genes may clarify their role in erythroid cell differentiation.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Erythroblasts/metabolism , Gene Expression Regulation, Developmental/physiology , Heme/metabolism , 5-Aminolevulinate Synthetase/deficiency , Animals , Cells, Cultured , Gene Expression Profiling , Mice
18.
Blood ; 101(3): 1188-93, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12393610

ABSTRACT

Alas2 encodes the erythroid-specific delta-aminolevulinate synthase (ALAS2 or ALAS-E), the first enzyme in heme biosynthesis in erythroid cells. Mice with the Alas2-null phenotype showed massive cytoplasmic, but not mitochondrial, iron accumulation in their primitive erythroblasts. Because these animals died by day 11.5 in utero, studies of iron metabolism in definitive erythroblasts were not possible using the in vivo model. In this study, embryonic stem (ES) cells lacking the Alas2 gene were induced to undergo differentiation to the definitive erythroblast stage in culture, and the phenotype of Alas2-null definitive erythroblasts was examined. Alas2-null definitive erythroblasts cell pellets were entirely colorless due to a marked deficiency of heme, although their cell morphology was similar to that of the wild-type erythroblasts. The level of expression of erythroid-specific genes in Alas2-null definitive erythroblasts was also similar to that of the wild-type erythroblasts. These findings indicate that Alas2-null definitive erythroblasts developed to a stage similar to that of the wild-type erythroblasts, which were also shown to be very similar to the bone marrow erythroblasts in vivo. In contrast, Alas2-null definitive erythroblasts contained 15 times more nonheme iron than did the wild-type erythroblasts, and electron microscopy found this iron to be distributed in the cytoplasm but not in mitochondria. Consistent with the aberrant increase in iron, Alas2-null definitive erythroblasts were more peroxidized than wild-type erythroblasts. These findings suggest that ALAS2 deficiency itself does not interfere with the development of definitive erythroid cells, but it results in a profound iron accumulation and a peroxidized state in erythroblasts.


Subject(s)
5-Aminolevulinate Synthetase/deficiency , Erythroblasts/metabolism , Iron/metabolism , Animals , Cell Line , Cytoplasm/metabolism , Erythroblasts/cytology , Erythroblasts/enzymology , Gene Expression Profiling , Heme/metabolism , Mice , Mice, Knockout , Mitochondria/metabolism , Oxidation-Reduction
19.
Blood ; 101(5): 1996-2000, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12406866

ABSTRACT

The sideroblastic anemias are characterized by ring sideroblasts, that is, red cell precursors with mitochondrial iron accumulation. We therefore studied the expression of mitochondrial ferritin (MtF) in these conditions. Erythroid cells from 13 patients with refractory anemia with ring sideroblasts (RARS) and 3 patients with X-linked sideroblastic anemia (XLSA) were analyzed for the distribution of cytoplasmic H ferritin (HF) and MtF using immunocytochemical methods. We also studied 11 healthy controls, 5 patients with refractory anemia without ring sideroblasts (RA), and 7 patients with RA with excess of blasts (RAEB). About one fourth of normal immature red cells, mostly proerythroblasts and basophilic erythroblasts, showed diffuse cytoplasmic positivity for HF, but very few were positive for MtF (0%-10%). Similar patterns were found in anemic patients without ring sideroblasts. In contrast, many erythroblasts from patients with sideroblastic anemia (82%-90% in XLSA and 36%-84% in RARS) were positive for MtF, which regularly appeared as granules ringing the nucleus. Double immunocytochemical staining confirmed the different cellular distribution of HF and MtF. There was a highly significant relationship between the percentage of MtF(+) erythroblasts and that of ring sideroblasts (Spearman R = 0.90; P <.0001). Reverse transcription-polymerase chain reaction studies demonstrated the presence of MtF mRNA in circulating reticulocytes of 2 patients with XLSA but not in controls. These findings suggest that most of the iron deposited in perinuclear mitochondria of ring sideroblasts is present in the form of MtF and that this latter might be a specific marker of sideroblastic anemia.


Subject(s)
Anemia, Sideroblastic/blood , Ceruloplasmin/metabolism , Erythroblasts/metabolism , Ferritins/blood , Genetic Diseases, X-Linked/blood , Iron/blood , Mitochondria/metabolism , Mitochondrial Proteins/blood , Reticulocytes/metabolism , 5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/genetics , Anemia, Refractory/blood , Anemia, Refractory, with Excess of Blasts/blood , Anemia, Sideroblastic/genetics , Ceruloplasmin/genetics , Chromosomes, Human, Pair 5/genetics , Cytoplasm/chemistry , Erythroblasts/ultrastructure , Humans , Mitochondrial Proteins/genetics , Myelodysplastic Syndromes/blood , RNA, Messenger/biosynthesis , Reticulocytes/ultrastructure
20.
Cell Mol Biol (Noisy-le-grand) ; 48(1): 5-10, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11929048

ABSTRACT

Hereditary sideroblastic anemia (HSA) is a heterogeneous group of inherited anemic disorders which is characterized by the presence of ringed sideroblasts in the bone marrow, microcytic hypochromic anemia and typically its X-linked inheritance in patients. It has been shown that a deficiency of the erythroid-specific delta-aminolevulinate synthase (ALAS-E) activity is responsible for pyridoxine-responsive HSA in many patients, however, the pathogenesis of other types of HSA remains still unknown. In this article, recent evidence suggesting multiple causes for HSA is summarized and discussed.


Subject(s)
Anemia, Sideroblastic/genetics , 5-Aminolevulinate Synthetase/deficiency , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Anemia, Sideroblastic/enzymology , Anemia, Sideroblastic/etiology , Family Health , Genetic Heterogeneity , Humans , Point Mutation , Protein Binding , Succinate-CoA Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...