Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 531
Filter
Add more filters

Publication year range
1.
Blood ; 144(6): 657-671, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38635773

ABSTRACT

ABSTRACT: Pseudouridine is the most prevalent RNA modification, and its aberrant function is implicated in various human diseases. However, the specific impact of pseudouridylation on hematopoiesis remains poorly understood. Here, we investigated the role of transfer RNA (tRNA) pseudouridylation in erythropoiesis and its association with mitochondrial myopathy, lactic acidosis, and sideroblastic anemia syndrome (MLASA) pathogenesis. By using patient-specific induced pluripotent stem cells (iPSCs) carrying a genetic pseudouridine synthase 1 (PUS1) mutation and a corresponding mutant mouse model, we demonstrated impaired erythropoiesis in MLASA-iPSCs and anemia in the MLASA mouse model. Both MLASA-iPSCs and mouse erythroblasts exhibited compromised mitochondrial function and impaired protein synthesis. Mechanistically, we revealed that PUS1 deficiency resulted in reduced mitochondrial tRNA levels because of pseudouridylation loss, leading to aberrant mitochondrial translation. Screening of mitochondrial supplements aimed at enhancing respiration or heme synthesis showed limited effect in promoting erythroid differentiation. Interestingly, the mammalian target of rapamycin (mTOR) inhibitor rapamycin facilitated erythroid differentiation in MLASA-iPSCs by suppressing mTOR signaling and protein synthesis, and consistent results were observed in the MLASA mouse model. Importantly, rapamycin treatment partially ameliorated anemia phenotypes in a patient with MLASA. Our findings provide novel insights into the crucial role of mitochondrial tRNA pseudouridylation in governing erythropoiesis and present potential therapeutic strategies for patients with anemia facing challenges related to protein translation.


Subject(s)
Erythropoiesis , Induced Pluripotent Stem Cells , Mitochondria , RNA, Transfer , Animals , Mice , Humans , RNA, Transfer/genetics , RNA, Transfer/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Induced Pluripotent Stem Cells/metabolism , Pseudouridine/metabolism , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/metabolism , Anemia, Sideroblastic/pathology , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , Hydro-Lyases/metabolism , Hydro-Lyases/genetics , MELAS Syndrome/genetics , MELAS Syndrome/pathology , MELAS Syndrome/metabolism , Disease Models, Animal
2.
Biochemistry ; 63(13): 1636-1646, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38888931

ABSTRACT

The conserved enzyme aminolevulinic acid synthase (ALAS) initiates heme biosynthesis in certain bacteria and eukaryotes by catalyzing the condensation of glycine and succinyl-CoA to yield aminolevulinic acid. In humans, the ALAS isoform responsible for heme production during red blood cell development is the erythroid-specific ALAS2 isoform. Owing to its essential role in erythropoiesis, changes in human ALAS2 (hALAS2) function can lead to two different blood disorders. X-linked sideroblastic anemia results from loss of ALAS2 function, while X-linked protoporphyria results from gain of ALAS2 function. Interestingly, mutations in the ALAS2 C-terminal extension can be implicated in both diseases. Here, we investigate the molecular basis for enzyme dysfunction mediated by two previously reported C-terminal loss-of-function variants, hALAS2 V562A and M567I. We show that the mutations do not result in gross structural perturbations, but the enzyme stability for V562A is decreased. Additionally, we show that enzyme stability moderately increases with the addition of the pyridoxal 5'-phosphate (PLP) cofactor for both variants. The variants display differential binding to PLP and the individual substrates compared to wild-type hALAS2. Although hALAS2 V562A is a more active enzyme in vitro, it is less efficient concerning succinyl-CoA binding. In contrast, the M567I mutation significantly alters the cooperativity of substrate binding. In combination with previously reported cell-based studies, our work reveals the molecular basis by which hALAS2 C-terminal mutations negatively affect ALA production necessary for proper heme biosynthesis.


Subject(s)
5-Aminolevulinate Synthetase , Anemia, Sideroblastic , Humans , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/deficiency , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Loss of Function Mutation , Enzyme Stability , Heme/metabolism , Heme/chemistry , Porphyrias/genetics , Porphyrias/metabolism , Models, Molecular , Mutation , Protoporphyria, Erythropoietic
3.
Blood ; 139(16): 2460-2470, 2022 04 21.
Article in English | MEDLINE | ID: mdl-34932791

ABSTRACT

The erythroid marrow and circulating red blood cells (RBCs) are the key components of the human erythron. Abnormalities of the erythron that are responsible for anemia can be separated into 3 major categories: erythroid hypoproliferation, ineffective erythropoiesis, and peripheral hemolysis. Ineffective erythropoiesis is characterized by erythropoietin-driven expansion of early-stage erythroid precursors, associated with apoptosis of late-stage precursors. This mechanism is primarily responsible for anemia in inherited disorders like ß-thalassemia, inherited sideroblastic anemias, and congenital dyserythropoietic anemias, as well as in acquired conditions like some subtypes of myelodysplastic syndrome (MDS). The inherited anemias that are due to ineffective erythropoiesis are also defined as iron-loading anemias because of the associated parenchymal iron loading caused by the release of erythroid factors that suppress hepcidin production. Novel treatments specifically targeting ineffective erythropoiesis are being developed. Iron restriction through enhancement of hepcidin activity or inhibition of ferroportin function has been shown to reduce ineffective erythropoiesis in murine models of ß-thalassemia. Luspatercept is a transforming growth factor-ß ligand trap that inhibits SMAD2/3 signaling. Based on preclinical and clinical studies, this compound is now approved for the treatment of anemia in adult patients with ß-thalassemia who require regular RBC transfusions. Luspatercept is also approved for the treatment of transfusion-dependent anemia in patients with MDS with ring sideroblasts, most of whom carry a somatic SF3B1 mutation. While the long-term effectiveness and safety of luspatercept need to be evaluated in ß-thalassemia and MDS, defining the molecular mechanisms of ineffective erythropoiesis in different disorders might allow the discovery of new effective compounds.


Subject(s)
Anemia, Sideroblastic , Myelodysplastic Syndromes , beta-Thalassemia , Adult , Humans , Mice , Animals , Hepcidins/pharmacology , Erythropoiesis/genetics , beta-Thalassemia/complications , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Iron , Anemia, Sideroblastic/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics
4.
Haematologica ; 109(8): 2525-2532, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38450522

ABSTRACT

The revised 4th edition of the World Health Organization (WHO4R) classification lists myelodysplastic syndromes with ring sideroblasts (MDS-RS) as a separate entity with single lineage (MDS-RS-SLD) or multilineage (MDS-RS-MLD) dysplasia. The more recent International Consensus Classification (ICC) distinguishes between MDS with SF3B1 mutation (MDS-SF3B1) and MDS-RS without SF3B1 mutation; the latter is instead included under the category of MDS not otherwise specified. The current study includes 170 Mayo Clinic patients with WHO4R-defined MDS-RS, including MDS-RS-SLD (N=83) and MDS-RSMLD (N=87); a subset of 145 patients were also evaluable for the presence of SF3B1 and other mutations, including 126 with (87%) and 19 (13%) without SF3B1 mutation. Median overall survival for all 170 patients was 6.6 years with 5- and 10-year survival rates of 59% and 25%, respectively. A significant difference in overall survival was apparent between MDS-RS-MLD and MDS-RS-SLD (P<0.01) but not between MDS-RS with and without SF3B1 mutation (P=0.36). Multivariable analysis confirmed the independent prognostic contribution of MLD (hazard ratio=1.8, 95% confidence interval: 1.1-2.8; P=0.01) and also identified age (P<0.01), transfusion need at diagnosis (P<0.01), and abnormal karyotype (P<0.01), as additional risk factors; the impact from SF3B1 or other mutations was not significant. Leukemia-free survival was independently affected by abnormal karyotype (P<0.01), RUNX1 (P=0.02) and IDH1 (P=0.01) mutations, but not by MLD or SF3B1 mutation. Exclusion of patients not meeting ICC-criteria for MDS-SF3B1 did not change the observations on overall survival. MLD-based, as opposed to SF3B1 mutation-based, disease classification for MDS-RS might be prognostically more relevant.


Subject(s)
Anemia, Sideroblastic , Mutation , Myelodysplastic Syndromes , Phosphoproteins , RNA Splicing Factors , Humans , RNA Splicing Factors/genetics , Male , Female , Aged , Middle Aged , Prognosis , Aged, 80 and over , Adult , Phosphoproteins/genetics , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/mortality , Anemia, Sideroblastic/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/pathology , Ribonucleoprotein, U2 Small Nuclear/genetics , Cell Lineage , Young Adult
5.
J Biol Chem ; 298(3): 101643, 2022 03.
Article in English | MEDLINE | ID: mdl-35093382

ABSTRACT

Heme is a critical biomolecule that is synthesized in vivo by several organisms such as plants, animals, and bacteria. Reflecting the importance of this molecule, defects in heme biosynthesis underlie several blood disorders in humans. Aminolevulinic acid synthase (ALAS) initiates heme biosynthesis in α-proteobacteria and nonplant eukaryotes. Debilitating and painful diseases such as X-linked sideroblastic anemia and X-linked protoporphyria can result from one of more than 91 genetic mutations in the human erythroid-specific enzyme ALAS2. This review will focus on recent structure-based insights into human ALAS2 function in health and how it dysfunctions in disease. We will also discuss how certain genetic mutations potentially result in disease-causing structural perturbations. Furthermore, we use thermodynamic and structural information to hypothesize how the mutations affect the human ALAS2 structure and categorize some of the unique human ALAS2 mutations that do not respond to typical treatments, that have paradoxical in vitro activity, or that are highly intolerable to changes. Finally, we will examine where future structure-based insights into the family of ALA synthases are needed to develop additional enzyme therapeutics.


Subject(s)
5-Aminolevulinate Synthetase , Anemia, Sideroblastic , Genetic Diseases, X-Linked , 5-Aminolevulinate Synthetase/chemistry , 5-Aminolevulinate Synthetase/genetics , 5-Aminolevulinate Synthetase/metabolism , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/metabolism , Anemia, Sideroblastic/enzymology , Anemia, Sideroblastic/genetics , Animals , Heme , Humans , Structure-Activity Relationship
6.
Ann Hum Genet ; 87(4): 166-173, 2023 07.
Article in English | MEDLINE | ID: mdl-36916508

ABSTRACT

INTRODUCTION: Congenital sideroblastic anemias (CSAs) are a group of inherited bone-marrow disorders manifesting with erythroid hyperplasia and ineffective erythropoiesis. METHODS: We describe a detailed clinical and genetic characterization of three siblings with CSA. RESULTS: Two of them had limb-girdle myopathy and global developmental delay. The two elder siblings performed allogenic hematopoietic stem-cell transplantation 5 and 3 years prior with stabilization of the hematological features. Exome sequencing in the non-transplanted sibling revealed a novel homozygous nonsense variant in SLC25A38 gene NM_017875.2:c.559C > T; p.(Arg187*) causing autosomal-recessive sideroblastic anemia type-2, and a second homozygous pathogenic previously reported variant in GMPPB gene NM_013334.3:c.458C > T; p.(Thr153Ile) causing autosomal-recessive muscular dystrophy-dystroglycanopathy type B14. With the established diagnosis, hematopoietic stem cell transplantation is now being scheduled for the youngest sibling, and a trial therapy with acetylcholine esterase inhibitors was started for the two neurologically affected patients with partial clinical improvement. CONCLUSION: This family emphasizes the importance of whole-exome sequencing for familial cases with complex phenotypes and vague neurological manifestations.


Subject(s)
Anemia, Sideroblastic , Humans , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/pathology , Siblings , Genotype , Phenotype , Mutation
7.
J Clin Immunol ; 43(4): 780-793, 2023 05.
Article in English | MEDLINE | ID: mdl-36729249

ABSTRACT

PURPOSE: Sideroblastic anemia, immunodeficiency, periodic fevers, and developmental delay (SIFD) is an autosomal recessive syndrome caused by biallelic loss-of-function variant of tRNA nucleotidyl transferase 1 (TRNT1). Efficacious methods to treat SIFD are lacking. We identified two novel mutations in TRNT1 and an efficacious and novel therapy for SIFD. METHODS: We retrospectively summarized the clinical records of two patients with SIFD from different families and reviewed all published cases of SIFD. RESULTS: Both patients had periodic fever, developmental delay, rash, microcytic anemia, and B cell lymphopenia with infections. Whole-exome sequencing of patient 1 identified a previously unreported homozygous mutation of TRNT1 (c.706G > A/p.Glu236Lys). He received intravenous immunoglobulin (IVIG) replacement and antibiotics, but died at 1 year of age. Gene testing in patient 2 revealed compound heterozygous mutations (c.907C > G/p.Gln303Glu and c.88A > G/p.Met30Val) in TRNT1, the former of which is a novel mutation. Periodic fever was controlled in the first month after adalimumab therapy and IVIG replacement, but recurred in the second month. Adalimumab was discontinued and replaced with thalidomide, which controlled the periodic fever and normalized inflammatory markers effectively. A retrospective analysis of reported cases revealed 69 patients with SIFD carrying 46 mutations. The male: female ratio was 1: 1, and the mean age of onset was 3.0 months. The most common clinical manifestations in patients with SIFD were microcytic anemia (82.6%), hypogammaglobulinemia/B cell lymphopenia (75.4%), periodic fever (66.7%), and developmental delay (60.0%). In addition to the typical tetralogy, SIFD features several heterogeneous symptoms involving multiple systems. Corticosteroids, immunosuppressants, and anakinra have low efficacy, whereas etanercept suppressed fever and improved anemia in reports. Bone-marrow transplantation can be used to treat severe SIFD, but carries a high risk. In total, 28.2% (20/71) of reported patients died, mainly because of multi-organ failure. Biallelic mutations located in exon1-intron5 lead to more severe phenotypes and higher mortality. Furthermore, 15.5% (11/71) patients survived to adulthood. The symptoms could be resolved spontaneously in five patients. CONCLUSIONS: Thalidomide can control the inflammation of SIFD and represents a new treatment for SIFD.


Subject(s)
Anemia, Sideroblastic , Immunologic Deficiency Syndromes , Lymphopenia , Male , Humans , Female , Thalidomide , Retrospective Studies , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/drug therapy , Anemia, Sideroblastic/genetics , Adalimumab , Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/genetics , Fever/etiology , Fever/genetics , Mutation/genetics , Nucleotidyltransferases/genetics
8.
J Struct Biol ; 214(1): 107832, 2022 03.
Article in English | MEDLINE | ID: mdl-35041979

ABSTRACT

ATP-binding cassette subfamily B member 7 (ABCB7) is localized in the inner membrane of mitochondria, playing a critical role in iron metabolism. Here, we determined the structure of the nonhydrolyzable ATP analog adenosine-5'-(ß-γ-imido) triphosphate (AMP-PNP) bound human ABCB7 at 3.3 Å by single-particle electron cryo-microscopy (cryo-EM). The AMP-PNP-bound human ABCB7 shows an inverted V-shaped homodimeric architecture with an inward-facing open conformation. One AMP-PNP molecule and Mg2+ were identified in each nucleotide-binding domain (NBD) of the hABCB7 monomer. Moreover, four disease-causing missense mutations of human ABCB7 have been mapped to the structure, creating a hotspot map for X-linked sideroblastic anemia and ataxia disease. Our results provide a structural basis for further understanding the transport mechanism of the mitochondrial ABC transporter.


Subject(s)
ATP-Binding Cassette Transporters , Anemia, Sideroblastic , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Adenylyl Imidodiphosphate/metabolism , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/metabolism , Cryoelectron Microscopy , Humans , Mitochondria/metabolism
9.
Am J Med Genet A ; 188(1): 259-268, 2022 01.
Article in English | MEDLINE | ID: mdl-34510712

ABSTRACT

Sideroblastic anemia with immunodeficiency, fevers, and developmental delay (SIFD; MIM #616084) is an autosomal recessive disorder of mitochondrial and cytosolic tRNA processing caused by pathogenic, biallelic variants in TRNT1. Other features of this disorder include central nervous system, renal, cardiac, ophthalmological features, and sensorineural hearing impairment. SIFD was first described in 2013 and to date, it has been reported in 46 patients. Herein, we review the literature and describe two siblings with SIFD and note the novel phenotype of hypoglycemia in the context of growth hormone (GH) deficiency. GH deficiency without hypoglycemia has previously been reported in three patients with SIFD, but GH deficiency had not been firmly ascribed to SIFD. We propose to expand the phenotype to include GH deficiency, hypoglycemia, and previously unreported dysmorphic features. Furthermore, we highlight the intrafamilial variability of the disease by the discordance of our patients' clinical phenotypes and biochemical profiles measured by untargeted metabolomics analysis. Several metabolomic abnormalities were observed in both patients, and these may represent a potential biochemical signature for SIFD.


Subject(s)
Anemia, Sideroblastic , Anemia, Sideroblastic/genetics , Fever/complications , Fever/genetics , Humans , Mutation , Nucleotidyltransferases/genetics , Phenotype
10.
Am J Med Genet A ; 188(7): 2226-2230, 2022 07.
Article in English | MEDLINE | ID: mdl-35393742

ABSTRACT

Myopathy, lactic acidosis, and sideroblastic anemia 2 (MLASA2) is an autosomal recessive mitochondrial disorder caused by pathogenic variants in YARS2. YARS2 variants confer heterogeneous phenotypes ranging from the full MLASA syndrome to a clinically unaffected state. Symptom onset is most common in the first decade of life but can occur in adulthood and has been reported following intercurrent illness. Early death can result from respiratory muscle weakness and cardiomyopathy. We report a case of MLASA2 with compound heterozygous YARS2 pathogenic variants; a known pathogenic nonsense variant [NM_001040436.3:c.98C>A (p.Ser33Ter)] and a likely pathogenic missense variant not previously associated with disease [NM_001040436.3:c.948G>T (p.Arg316Ser)]. The proband initially presented with a relatively mild phenotype of myopathy and lactic acidosis. During pregnancy, anemia emerged as an additional feature and in the postpartum period she experienced severe decompensation of cardiorespiratory function. This is the first reported case of pregnancy-related complications in a patient with YARS2-related mitochondrial disease. This case highlights the need for caution and careful counseling when considering pregnancy in mitochondrial disease, due to the risk of disease exacerbation and pregnancy complications.


Subject(s)
Acidosis, Lactic , Anemia, Sideroblastic , Mitochondrial Myopathies , Muscular Diseases , Tyrosine-tRNA Ligase , Acidosis, Lactic/diagnosis , Acidosis, Lactic/genetics , Adult , Anemia, Sideroblastic/complications , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/genetics , Female , Humans , Mitochondrial Myopathies/complications , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/genetics , Muscular Diseases/genetics , Pregnancy , Tyrosine-tRNA Ligase/genetics
11.
Muscle Nerve ; 65(4): 374-390, 2022 04.
Article in English | MEDLINE | ID: mdl-34985130

ABSTRACT

The diagnostic evaluation of a patient with suspected hereditary muscle disease can be challenging. Clinicians rely largely on clinical history and examination features, with additional serological, electrodiagnostic, radiologic, histopathologic, and genetic investigations assisting in definitive diagnosis. Hematological testing is inexpensive and widely available, but frequently overlooked in the hereditary myopathy evaluation. Hematological abnormalities are infrequently encountered in this setting; however, their presence provides a valuable clue, helps refine the differential diagnosis, tailors further investigation, and assists interpretation of variants of uncertain significance. A diverse spectrum of hematological abnormalities is associated with hereditary myopathies, including anemias, leukocyte abnormalities, and thrombocytopenia. Recurrent rhabdomyolysis in certain glycolytic enzymopathies co-occurs with hemolytic anemia, often chronic and mild in phosphofructokinase and phosphoglycerate kinase deficiencies, or acute and fever-associated in aldolase-A and triosephosphate isomerase deficiency. Sideroblastic anemia, commonly severe, accompanies congenital-to-childhood onset mitochondrial myopathies including Pearson marrow-pancreas syndrome and mitochondrial myopathy, lactic acidosis, and sideroblastic anemia phenotypes. Congenital megaloblastic macrocytic anemia and mitochondrial dysfunction characterize SFXN4-related myopathy. Neutropenia, chronic or cyclical, with recurrent infections, infantile-to-childhood onset skeletal myopathy and cardiomyopathy are typical of Barth syndrome, while chronic neutropenia without infection occurs rarely in DNM2-centronuclear myopathy. Peripheral eosinophilia may accompany eosinophilic inflammation in recessive calpainopathy. Lipid accumulation in leukocytes on peripheral blood smear (Jordans' anomaly) is pathognomonic for neutral lipid storage diseases. Mild thrombocytopenia occurs in autosomal dominant, childhood-onset STIM1 tubular aggregate myopathy, STIM1 and ORAI1 deficiency syndromes, and GNE myopathy. Herein, we review these hereditary myopathies in which hematological features play a prominent role.


Subject(s)
Anemia, Sideroblastic , Lipid Metabolism, Inborn Errors , Mitochondrial Diseases , Mitochondrial Myopathies , Myopathies, Structural, Congenital , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/pathology , Child , Humans , Mitochondrial Myopathies/genetics , Mutation , Myopathies, Structural, Congenital/genetics
12.
Clin Lab ; 68(4)2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35443589

ABSTRACT

BACKGROUND: Myelodysplastic/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/ MPN-RS-T) was newly introduced as a full entity in the 2016 revision of the WHO classification. In this study, we investigated the morphologic, laboratory, and clinical features of MDS/MPN-RS-T. METHODS: We reviewed the bone marrow and genetic studies of patients whose diagnoses were coded as "refractory anemia with ring sideroblasts (RARS)" or "MDS/MPN, unclassifiable" between January 2008 and April 2018. RESULTS: A total of 8 cases fulfilled the criteria for a diagnosis of MDS/MPN-RS-T. All of them had no specific symptoms. Half of the cases had less than 450 × 109/L platelet counts by an automated hematology analyzer; however, all platelet counts exceeded 450 × 109/L when performed manually. JAK2 mutation tests were performed in 7 cases, and a heterozygous mutation was detected in 1 case. SF3B1 mutations were present in 3 of the 4 cases tested. CONCLUSIONS: When RARS is suspected in patients without thrombocytopenia, manual platelet counts should be performed. For patients with suspected essential thrombocythemia, RS evaluation through careful observation of an iron-stained slide is crucial. Since the independent evaluation of RS was reflected in the revised classification, the ambiguous disease classification becomes clearer and more consistent.


Subject(s)
Anemia, Refractory , Anemia, Sideroblastic , Myelodysplastic-Myeloproliferative Diseases , Neoplasms , Thrombocytosis , Anemia, Refractory/diagnosis , Anemia, Refractory/genetics , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/genetics , Humans , Mutation , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Myelodysplastic-Myeloproliferative Diseases/genetics , Thrombocytosis/diagnosis , Thrombocytosis/genetics
13.
Rinsho Ketsueki ; 63(6): 600-607, 2022.
Article in Japanese | MEDLINE | ID: mdl-35831194

ABSTRACT

Sideroblastic anemias (SAs) are a group of heterogeneous congenital and acquired disorders characterized by anemia and presence of ring sideroblasts in the bone marrow. Congenital SA is a rare condition caused by mutations of genes involved in heme biosynthesis, iron-sulfur cluster biosynthesis, and mitochondrial protein synthesis. SAs can also occur following exposure to certain drugs or alcohol or caused by copper deficiency (secondary SA). SAs have been found to be associated with myelodysplastic syndrome (idiopathic SA), which strongly correlates with specific somatic mutations in SF3B1 (splicing factor 3b subunit 1), involved in the RNA splicing machinery. The recent widespread use of genome-editing technology and next-generation sequencing has led to a better understanding of the molecular pathophysiology of SAs. This review discusses the current understanding of the pathophysiology of SAs.


Subject(s)
Anemia, Sideroblastic , Myelodysplastic Syndromes , Anemia, Sideroblastic/genetics , Erythroid Precursor Cells/metabolism , Humans , Mutation , Myelodysplastic Syndromes/complications , RNA Splicing
14.
Cancer Sci ; 112(5): 1888-1898, 2021 May.
Article in English | MEDLINE | ID: mdl-33560542

ABSTRACT

Circular RNAs (circRNAs) have been identified to exert vital biological functions and can be used as new biomarkers in a number of tumors. However, little is known about the functions of circRNAs in myelodysplastic syndrome (MDS). Here, we aimed to investigate circRNA expression profiles and to investigate the functional and clinical value of circRNAs in MDS. Differential expression of circRNAs between MDS and control subjects was analyzed using circRNA arrays, in which we identified 145 upregulated circRNAs and 224 downregulated circRNAs. Validated by real-time quantitative PCR between 100 MDS patients and 20 controls, three upregulated (hsa_circRNA_100352, hsa_circRNA_104056, and hsa_circRNA_104634) and three downregulated (hsa_circRNA_103846, hsa_circRNA_102817, and hsa_circRNA_102526) circRNAs matched the arrays. The receiver operating characteristic curve analysis of these circRNAs showed that the area under the curve was 0.7266, 0.8676, 0.7349, 0.7091, 0.8806, and 0.7472, respectively. Kaplan-Meier survival analysis showed that only hsa_circRNA_100352, hsa_circRNA_104056, and hsa_circRNA_102817 were significantly associated with overall survival. Furthermore, we generated a competing endogenous RNA network focused on hsa_circRNA_100352, hsa_circRNA_104056, and hsa_circRNA_102817. Analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes showed that the three circRNAs were linked with some important cancer-related functions and pathways.


Subject(s)
Biomarkers, Tumor/metabolism , Myelodysplastic Syndromes/metabolism , RNA, Circular/metabolism , Aged , Anemia, Refractory/genetics , Anemia, Refractory/metabolism , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/metabolism , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/metabolism , Area Under Curve , Biomarkers, Tumor/genetics , Bone Marrow/metabolism , Case-Control Studies , Down-Regulation , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , RNA, Circular/genetics , ROC Curve , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric , Up-Regulation
15.
Blood ; 133(1): 59-69, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30401706

ABSTRACT

The sideroblastic anemias (SAs) are a group of inherited and acquired bone marrow disorders defined by pathological iron accumulation in the mitochondria of erythroid precursors. Like most hematological diseases, the molecular genetic basis of the SAs has ridden the wave of technology advancement. Within the last 30 years, with the advent of positional cloning, the human genome project, solid-state genotyping technologies, and next-generation sequencing have evolved to the point where more than two-thirds of congenital SA cases, and an even greater proportion of cases of acquired clonal disease, can be attributed to mutations in a specific gene or genes. This review focuses on an analysis of the genetics of these diseases and how understanding these defects may contribute to the design and implementation of rational therapies.


Subject(s)
Anemia, Sideroblastic/genetics , Erythroid Precursor Cells/pathology , Genetic Diseases, X-Linked/genetics , Iron/metabolism , Anemia, Sideroblastic/metabolism , Anemia, Sideroblastic/pathology , Genetic Diseases, X-Linked/metabolism , Genetic Diseases, X-Linked/pathology , Humans
17.
Protein Expr Purif ; 183: 105860, 2021 07.
Article in English | MEDLINE | ID: mdl-33689857

ABSTRACT

The ATP-binding cassette sub-family B member 7 (ABCB7) is a membrane transport protein located on the inner membrane of mitochondria, which could be involved in the transport of heme from the mitochondria to the cytosol. ABCB7 also plays a central role in the maturation of cytosolic iron-sulfur (Fe/S) cluster-containing proteins, and mutations can cause a series of mitochondrial defects. X-linked sideroblastic anemia and ataxia (XLSA-A) is a rare cause of early onset ataxia, which may be overlooked due to the usually mild asymptomatic anemia. The genetic defect has been identified as a mutation in the ABCB7 gene at Xq12-q13. Here, we report the expression, purification and the 2D projections derived from negatively stained electron micrographs of recombinant H. sapiens ABCB7 (hABCB7), paving the way from an atomic structure determination of ABCB7.


Subject(s)
ATP-Binding Cassette Transporters , Mutation , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/isolation & purification , Anemia, Sideroblastic/enzymology , Anemia, Sideroblastic/genetics , Genetic Diseases, X-Linked/enzymology , Genetic Diseases, X-Linked/genetics , Humans , Protein Conformation , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Spinocerebellar Ataxias/enzymology , Spinocerebellar Ataxias/genetics
18.
Australas J Dermatol ; 62(2): e276-e279, 2021 May.
Article in English | MEDLINE | ID: mdl-33332575

ABSTRACT

Sideroblastic anaemia, B-cell immunodeficiency, periodic fever and developmental delay (SIFD) is caused by mutations of TRNT1, an enzyme essential for mitochondrial protein synthesis, and has been reported in 23 cases. A 6-month-old girl was evaluated with recurrent fever, failure to thrive, skin lesions and anaemia. She received blood transfusions and empirical antibiotics. Skin lesions, previously interpreted as insect bites, consisted of numerous firm asymptomatic erythematous papules and nodules, distributed over trunk and limbs. Skin histopathology revealed an intense dermal neutrophilic infiltrate extending to the subcutaneous, with numerous atypical myeloid cells, requiring the diagnosis of leukaemia cutis, to be ruled out. Over the follow-up, she developed herpetic stomatitis, tonsillitis, lobar pneumonia and Metapneumovirus tracheitis, and also deeper skin lesions, resembling panniculitis. Hypogammaglobulinaemia was diagnosed. An autoinflammatory disease was confirmed by whole exome sequencing: heterozygous mutations for TRNT1 NM_182916 c.495_498del, p.F167Tfs * 9 and TRNT1 NM_182916 c.1246A>G, p.K416E. The patient has been treated with subcutaneous immunoglobulin and etanercept. She presented with developmental delay and short stature for age. The fever, anaemia, skin neutrophilic infiltration and the inflammatory parameters improved. We describe a novel mutation in SIFD and the first to present skin manifestations, namely neutrophilic dermal and hypodermal infiltration.


Subject(s)
Anemia, Sideroblastic/diagnosis , Developmental Disabilities/complications , Immunologic Deficiency Syndromes/diagnosis , Neutrophils/metabolism , Skin Diseases/etiology , Anemia, Sideroblastic/genetics , Dermis/metabolism , Developmental Disabilities/genetics , Female , Fever/etiology , Humans , Immunologic Deficiency Syndromes/congenital , Immunologic Deficiency Syndromes/genetics , Infant , Mutation , Nucleotidyltransferases/genetics , Exome Sequencing
19.
Hum Mutat ; 41(8): 1425-1434, 2020 08.
Article in English | MEDLINE | ID: mdl-32442335

ABSTRACT

LARS2 variants are associated with Perrault syndrome, characterized by premature ovarian failure and hearing loss, and with an infantile lethal multisystem disorder: Hydrops, lactic acidosis, sideroblastic anemia (HLASA) in one individual. Recently we reported LARS2 deafness with (ovario) leukodystrophy. Here we describe five patients with a range of phenotypes, in whom we identified biallelic LARS2 variants: three patients with a HLASA-like phenotype, an individual with Perrault syndrome whose affected siblings also had leukodystrophy, and an individual with a reversible mitochondrial myopathy, lactic acidosis, and developmental delay. Three HLASA cases from two unrelated families were identified. All were males with genital anomalies. Two survived multisystem disease in the neonatal period; both have developmental delay and hearing loss. A 55-year old male with deafness has not displayed neurological symptoms while his female siblings with Perrault syndrome developed leukodystrophy and died in their 30s. Analysis of muscle from a child with a reversible myopathy showed reduced LARS2 and mitochondrial complex I levels, and an unusual form of degeneration. Analysis of recombinant LARS2 variant proteins showed they had reduced aminoacylation efficiency, with HLASA-associated variants having the most severe effect. A broad phenotypic spectrum should be considered in association with LARS2 variants.


Subject(s)
Amino Acyl-tRNA Synthetases/genetics , Gonadal Dysgenesis, 46,XX/genetics , Hearing Loss, Sensorineural/genetics , Mitochondrial Myopathies/genetics , Acidosis, Lactic/genetics , Adult , Anemia, Sideroblastic/genetics , Edema/genetics , Female , Humans , Infant , Male , Middle Aged , Phenotype , Protein Structure, Tertiary
20.
Am J Med Genet A ; 182(2): 365-373, 2020 02.
Article in English | MEDLINE | ID: mdl-31825167

ABSTRACT

Pearson syndrome (PS) is a multisystem mitochondrial respiratory chain disorder typically characterized by sideroblastic anemia and exocrine pancreatic insufficiency. PS is caused by a single large-scale mitochondrial DNA (mtDNA) deletion. PS classically presents in the first year of life and may be fatal in infancy. Children who survive PS may progress to develop Kearns-Sayre syndrome later in life. The full phenotypic spectrum and prognosis of the condition continue to evolve. Here we report five new patients with PS with unique clinical presentations, including four patients with onset later than previously reported in the literature, and one patient with prenatal onset of symptoms. The timing and unique features of these presentations support an expanded phenotypic spectrum of single large-scale mtDNA deletion syndromes (SLSMDS) and reinforce the importance of including SLSMDS in the differential for children with complex multisystem presentations.


Subject(s)
Congenital Bone Marrow Failure Syndromes/genetics , DNA, Mitochondrial/genetics , Kearns-Sayre Syndrome/genetics , Lipid Metabolism, Inborn Errors/genetics , Mitochondrial Diseases/genetics , Muscular Diseases/genetics , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/physiopathology , Child , Child, Preschool , Congenital Bone Marrow Failure Syndromes/physiopathology , Exocrine Pancreatic Insufficiency/genetics , Exocrine Pancreatic Insufficiency/physiopathology , Female , Gene Deletion , Humans , Infant , Kearns-Sayre Syndrome/physiopathology , Lipid Metabolism, Inborn Errors/physiopathology , Male , Mitochondria/genetics , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Muscular Diseases/physiopathology , Phenotype , Sequence Deletion/genetics
SELECTION OF CITATIONS
SEARCH DETAIL