1
|
Raynor A, Peoc'h K, Boi C, Manceau H, Pissard S, Diallo K, Kannengiesser C, Rohrlich P. Marked microcytosis and increased transferrin saturation: Think about variants in SLC11A2 (DMT1). Blood Cells Mol Dis 2025; 110:102898. [PMID: 39531753 DOI: 10.1016/j.bcmd.2024.102898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Congenital microcytic anemias are rare diseases associated with decreased hemoglobin synthesis and red blood cells of low corpuscular volume. DMT1/NRAMP2 is a highly conserved divalent cation transporter encoded by the SLC11A2 gene, expressed at the membrane of various cells. It ensures ferrous iron absorption from the apical membrane of enterocytes, iron recovery from urine by renal tubules, and acidified endosome uptake after transferrin internalization. Pathogenic DMT1 variants have been described in 10 individuals to date, associated with recessive hypochromic anemia and iron overload. Herein, we report a new variant of SLC11A2 (c.469A>G, p.Ile157Val) compound with known p.Arg416Cys associated with a frankly microcytic anemia and increased transferrin saturation. The clinical picture observed in the patient was exceptionally mild, extending the field of the DMT1 phenotypes to borderline anemias.
Collapse
Affiliation(s)
- Alexandre Raynor
- Service de Biochimie, Hôpital Bichat, APHP, Nord, 75018 Paris, France; Université Paris Cité, INSERM UMRs-1149, HIROS Heme Iron and Oxidative Stress, Centre de recherche sur l'inflammation, 75018 Paris, France
| | - Katell Peoc'h
- Service de Biochimie, Hôpital Bichat, APHP, Nord, 75018 Paris, France; Université Paris Cité, INSERM UMRs-1149, HIROS Heme Iron and Oxidative Stress, Centre de recherche sur l'inflammation, 75018 Paris, France; LBMR Maladies héréditaires du métabolisme (phénotype et génotype), Métabolisme des métaux (Fer), Hôpital Bichat, APHP, Nord, 75018 Paris, France; Université Paris Cité, UFR de Médecine Xavier Bichat, France
| | - Camille Boi
- LBMR Maladies héréditaires du métabolisme (phénotype et génotype), Métabolisme des métaux (Fer), Hôpital Bichat, APHP, Nord, 75018 Paris, France; Service de Génétique, Hôpital Bichat, APHP, Nord, 75018 Paris, France
| | - Hana Manceau
- Service de Biochimie, Hôpital Bichat, APHP, Nord, 75018 Paris, France; Université Paris Cité, INSERM UMRs-1149, HIROS Heme Iron and Oxidative Stress, Centre de recherche sur l'inflammation, 75018 Paris, France; LBMR Maladies héréditaires du métabolisme (phénotype et génotype), Métabolisme des métaux (Fer), Hôpital Bichat, APHP, Nord, 75018 Paris, France; Université Paris Cité, UFR de Médecine Xavier Bichat, France
| | - Serge Pissard
- departement de génétique, GHU Henri Mondor, APHP, Sud, Créteil, France
| | - Karim Diallo
- LBMR Maladies héréditaires du métabolisme (phénotype et génotype), Métabolisme des métaux (Fer), Hôpital Bichat, APHP, Nord, 75018 Paris, France; Service de Génétique, Hôpital Bichat, APHP, Nord, 75018 Paris, France
| | - Caroline Kannengiesser
- LBMR Maladies héréditaires du métabolisme (phénotype et génotype), Métabolisme des métaux (Fer), Hôpital Bichat, APHP, Nord, 75018 Paris, France; Université Paris Cité, UFR de Médecine Xavier Bichat, France; Service de Génétique, Hôpital Bichat, APHP, Nord, 75018 Paris, France.
| | - Pierre Rohrlich
- Service d'hémato-oncologie pédiatrique, Hôpital l'Archet, Nice, France
| |
Collapse
|
2
|
Yvan-Charvet L, Barouillet T, Borowczyk C. Haematometabolism rewiring in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01108-9. [PMID: 39743562 DOI: 10.1038/s41569-024-01108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 01/04/2025]
Abstract
Atherosclerotic cardiovascular diseases are the most frequent cause of death worldwide. The clinical complications of atherosclerosis are closely linked to the haematopoietic and immune systems, which maintain homeostatic functions and vital processes in the body. The nodes linking metabolism and inflammation are receiving increasing attention because they are inextricably linked to inflammatory manifestations of non-communicable diseases, including atherosclerosis. Although metabolism and inflammation are essential to survival and involve all tissues, we still know little about how these processes influence each other. In an effort to understand these mechanisms, in this Review we explore whether and how potent cardiovascular risk factors and metabolic modifiers of atherosclerosis influence the molecular and cellular machinery of 'haematometabolism' (metabolic-dependent haematopoietic stem cell skewing) and 'efferotabolism' (metabolic-dependent efferocyte reprogramming). These changes might ultimately propagate a quantitative and qualitative drift of the macrophage supply chain and affect the clinical manifestations of atherosclerosis. Refining our understanding of the different metabolic requirements of these processes could open the possibility of developing therapeutics targeting haematometabolism that, in conjunction with improved dietary habits, help rebalance and promote efficient haematopoiesis and efferocytosis and decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
3
|
Castruccio Castracani C, Breda L, Papp TE, Guerra A, Radaelli E, Assenmacher CA, Finesso G, Mui BL, Tam YK, Fontana S, Riganti C, Fiorito V, Petrillo S, Tolosano E, Parhiz H, Rivella S. An erythroid-specific lentiviral vector improves anemia and iron metabolism in a new model of XLSA. Blood 2025; 145:98-113. [PMID: 39656107 PMCID: PMC11738033 DOI: 10.1182/blood.2024025846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/14/2024] [Indexed: 01/03/2025] Open
Abstract
ABSTRACT X-linked sideroblastic anemia (XLSA) is a congenital anemia caused by mutations in ALAS2, a gene responsible for heme synthesis. Treatments are limited to pyridoxine supplements and blood transfusions, offering no definitive cure except for allogeneic hematopoietic stem cell transplantation, only accessible to a subset of patients. The absence of a suitable animal model has hindered the development of gene therapy research for this disease. We engineered a conditional Alas2-knockout (KO) mouse model using tamoxifen administration or treatment with lipid nanoparticles carrying Cre-mRNA and conjugated to an anti-CD117 antibody. Alas2-KOBM animals displayed a severe anemic phenotype characterized by ineffective erythropoiesis (IE), leading to low numbers of red blood cells, hemoglobin, and hematocrit. In particular, erythropoiesis in these animals showed expansion of polychromatic erythroid cells, characterized by reduced oxidative phosphorylation, mitochondria's function, and activity of key tricarboxylic acid cycle enzymes. In contrast, glycolysis was increased in the unsuccessful attempt to extend cell survival despite mitochondrial dysfunction. The IE was associated with marked splenomegaly and low hepcidin levels, leading to iron accumulation in the liver, spleen, and bone marrow and the formation of ring sideroblasts. To investigate the potential of a gene therapy approach for XLSA, we developed a lentiviral vector (X-ALAS2-LV) to direct ALAS2 expression in erythroid cells. Infusion of bone marrow (BM) cells with 0.6 to 1.4 copies of the X-ALAS2-LV in Alas2-KOBM mice improved complete blood cell levels, tissue iron accumulation, and survival rates. These findings suggest our vector could be curative in patients with XLSA.
Collapse
Affiliation(s)
| | - Laura Breda
- Department of Pediatrics, Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Tyler E. Papp
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amaliris Guerra
- Department of Pediatrics, Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Enrico Radaelli
- Comparative Pathology Core, Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Charles-Antoine Assenmacher
- Comparative Pathology Core, Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Giovanni Finesso
- Comparative Pathology Core, Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Barbara L. Mui
- Acuitas Therapeutics, Vancouver, British Columbia, Canada
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, British Columbia, Canada
| | - Simona Fontana
- Department of Oncology, University of Torino, Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | - Veronica Fiorito
- Department of Biotechnology and Health Sciences and Molecular Biotechnology Center "Guido Tarone," University of Torino, Torino, Italy
| | - Sara Petrillo
- Department of Biotechnology and Health Sciences and Molecular Biotechnology Center "Guido Tarone," University of Torino, Torino, Italy
| | - Emanuela Tolosano
- Department of Biotechnology and Health Sciences and Molecular Biotechnology Center "Guido Tarone," University of Torino, Torino, Italy
| | - Hamideh Parhiz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Stefano Rivella
- Department of Pediatrics, Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics–The Children's Hospital of Philadelphia, Philadelphia, PA
- Penn Center for Musculoskeletal Disorders, The Children's Hospital of Philadelphia, Philadelphia, PA
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
4
|
Stenton SL, Laricchia K, Lake NJ, Chaluvadi S, Ganesh V, DiTroia S, Osei-Owusu I, Pais L, O’Heir E, Austin-Tse C, O’Leary M, Abu Shanap M, Barrows C, Berger S, Bönnemann CG, Bujakowska KM, Campagna DR, Compton AG, Donkervoort S, Fleming MD, Gallacher L, Gleeson JG, Haliloglu G, Pierce EA, Place EM, Sankaran VG, Shimamura A, Stark Z, Tan TY, Thorburn DR, White SM, Vilain E, Lek M, Rehm HL, O’Donnell-Luria A. Mitochondrial DNA variant detection in over 6,500 rare disease families by the systematic analysis of exome and genome sequencing data resolves undiagnosed cases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.22.24319370. [PMID: 39763565 PMCID: PMC11703311 DOI: 10.1101/2024.12.22.24319370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Background Variants in the mitochondrial genome (mtDNA) cause a diverse collection of mitochondrial diseases and have extensive phenotypic overlap with Mendelian diseases encoded on the nuclear genome. The mtDNA is often not specifically evaluated in patients with suspected Mendelian disease, resulting in overlooked diagnostic variants. Methods Using dedicated pipelines to address the technical challenges posed by the mtDNA - circular genome, variant heteroplasmy, and nuclear misalignment - single nucleotide variants, small indels, and large mtDNA deletions were called from exome and genome sequencing data, in addition to RNA-sequencing when available. A cohort of 6,660 rare disease families were analyzed (5,625 genetically undiagnosed, 84%) from the Genomics Research to Elucidate the Genetics of Rare diseases (GREGoR) Consortium as well as other rare disease cohorts. Results Diagnostic mtDNA variants were identified in 10 previously genetically undiagnosed families (one large deletion, eight reported pathogenic variants, one novel pathogenic variant). In one additional undiagnosed proband, the detection of >900 heteroplasmic variants provided functional evidence of pathogenicity to a novel de novo variant in the nuclear gene POLG (DNA polymerase gamma), responsible for mtDNA replication and repair. Conclusion mtDNA variant calling from data generated by exome and genome sequencing for nuclear variant analysis resulted in a genetic diagnosis or detection of a candidate variant for 0.4% of undiagnosed families affected by a broad range of rare diseases.
Collapse
Affiliation(s)
- Sarah L. Stenton
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristen Laricchia
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Nicole J. Lake
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Sushma Chaluvadi
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Vijay Ganesh
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Stephanie DiTroia
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Ikeoluwa Osei-Owusu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Lynn Pais
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Emily O’Heir
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Christina Austin-Tse
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Melanie O’Leary
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Mayada Abu Shanap
- Hematology/Oncology, Bone Marrow Transplantation and Cellular Therapy, Pediatric Department, King Hussein Cancer Centre (KHCC), Amman, Jordan
| | - Chelsea Barrows
- University of California, Department of Neurosciences, San Diego, CA; Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
| | - Seth Berger
- Children’s National Research Institute, Washington, DC, USA
| | - Carsten G. Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kinga M. Bujakowska
- Ocular Genomics Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dean R. Campagna
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alison G. Compton
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Sandra Donkervoort
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mark D. Fleming
- Department of Pathology, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lyndon Gallacher
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Joseph G. Gleeson
- University of California, Department of Neurosciences, San Diego, CA; Rady Children’s Institute for Genomic Medicine, San Diego, CA, USA
| | - Goknur Haliloglu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eric A. Pierce
- Ocular Genomics Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Emily M. Place
- Ocular Genomics Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Vijay G. Sankaran
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
- Howard Hughes Medical Institute, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Akiko Shimamura
- Department of Hematology and Oncology, Boston Children’s Hospital, Boston, MA, USA
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Tiong Yang Tan
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - David R. Thorburn
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Susan M. White
- Victorian Clinical Genetics Services, Murdoch Children’s Research Institute, Flemington Road, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | | | - Eric Vilain
- Institute for Clinical and Translational Science, University of California, Irvine, CA, USA
| | - Monkol Lek
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Heidi L. Rehm
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anne O’Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
5
|
Gattermann N. Iron overload in acquired sideroblastic anemias and MDS: pathophysiology and role of chelation and luspatercept. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:443-449. [PMID: 39644054 DOI: 10.1182/hematology.2024000569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
Besides transfusion therapy, ineffective erythropoiesis contributes to systemic iron overload in myelodysplastic syndromes with ring sideroblasts (MDS-RS) via erythroferrone-induced suppression of hepcidin synthesis in the liver, leading to increased intestinal iron absorption. The underlying pathophysiology of MDS-RS, characterized by disturbed heme synthesis and mitochondrial iron accumulation, is less well understood. Several lines of evidence indicate that the mitochondrial transporter ABCB7 is critically involved. ABCB7 is misspliced and underexpressed in MDS-RS, due to somatic mutations in the splicing factor SF3B1. The pathogenetic significance of ABCB7 seems related to its role in stabilizing ferrochelatase, the enzyme incorporating iron into protoporphyrin IX to make heme. Although iron-related oxidative stress is toxic, many patients with MDS do not live long enough to develop clinical complications of iron overload. Furthermore, it is difficult to determine the extent to which iron overload contributes to morbidity and mortality in older patients with MDS, because iron-related complications overlap with age-related medical problems. Nevertheless, high-quality registry studies showed that transfusion dependency is associated with the presence of toxic iron species and inferior survival and confirmed a significant survival benefit of iron chelation therapy. The most widely used iron chelator in patients with MDS is deferasirox, owing to its effectiveness and convenient oral administration. Luspatercept, which can reduce SMAD2/SMAD3-dependent signaling implicated in suppression of erythropoiesis, may obviate the need for red blood cell transfusion in MDS-RS for more than a year, thereby diminishing further iron loading. However, luspatercept cannot be expected to substantially reduce the existing iron overload.
Collapse
|
6
|
Bick NR, Dreishpoon MB, Perry A, Rogachevskaya A, Bottomley SS, Fleming MD, Ducamp S, Tsvetkov P. Engineered bacterial lipoate protein ligase A (lplA) restores lipoylation in cell models of lipoylation deficiency. J Biol Chem 2024; 300:107995. [PMID: 39547509 DOI: 10.1016/j.jbc.2024.107995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/25/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Protein lipoylation, a vital lysine post-translational modification, plays a crucial role in the function of key mitochondrial tricarboxylic acid cycle enzymatic complexes. In eukaryotes, lipoyl post-translational modification synthesis occurs exclusively through de novo pathways, relying on lipoyl synthesis/transfer enzymes, dependent upon mitochondrial fatty acid and Fe-S cluster biosynthesis. Dysregulation in any of these pathways leads to diminished cellular lipoylation. Efficient restoration of lipoylation in lipoylation deficiency cell states using either chemical or genetic approaches has been challenging because of pathway complexity and multiple upstream regulators. To address this challenge, we explored the possibility that a bacterial lipoate protein ligase A (lplA) enzyme, which can salvage free lipoic acid bypassing the dependency on de novo synthesis, could be engineered to be functional in human cells. Overexpression of the engineered lplA in lipoylation null cells restored lipoylation levels, cellular respiration, and growth in low glucose conditions. Engineered lplA restored lipoylation in all tested lipoylation null cell models, mimicking defects in mitochondrial fatty acid synthesis (MECR KO), Fe-S cluster biosynthesis (BOLA3 KO), and specific lipoylation-regulating enzymes (FDX1 [ferredoxin 1], LIAS [lipoyl synthase], and LIPT1 [lipoyl (octanoyl) transferase 1] KOs). Furthermore, we describe a patient with a homozygous c.212C>T variant LIPT1 with a previously uncharacterized syndromic congenital sideroblastic anemia. K562 erythroleukemia cells engineered to harbor this missense LIPT1 allele recapitulate the lipoylation-deficient phenotype and exhibit impaired proliferation in low glucose that is completely restored by engineered lplA. This synthetic approach offers a potential therapeutic strategy for treating lipoylation disorders.
Collapse
Affiliation(s)
- Nolan R Bick
- Broad Institute of MIT and Harvard, Cambridge, Massachusets, USA
| | - Margaret B Dreishpoon
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ava Perry
- Broad Institute of MIT and Harvard, Cambridge, Massachusets, USA
| | - Anna Rogachevskaya
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sylvia S Bottomley
- Department of Medicine, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sarah Ducamp
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Tsvetkov
- Broad Institute of MIT and Harvard, Cambridge, Massachusets, USA; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
7
|
Dongerdiye R, Kedar PS, Saptarshi A, Sampagar A, Shanmukhaiah C, Mudaliar S, Kanvinde P, Desai M, Madkaikar M. Non-syndromic congenital sideroblastic anaemia; phenotype, and genotype of 15 Indian patients. Ann Hematol 2024; 103:3987-3998. [PMID: 39196378 DOI: 10.1007/s00277-024-05969-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
Sideroblastic anaemias are a diverse group of congenital and acquired bone marrow failure disorders marked by the presence of ring sideroblasts, ineffective erythropoiesis, and systemic iron overload. Congenital Sideroblastic anaemia (CSA) is mainly caused by gene mutations associated with heme synthesis, iron-sulfur [Fe-S] cluster, and mitochondrial protein synthesis pathways. The most prevalent form of CSA is caused by mutations in the erythroid-specific -amino levulinate synthase (ALAS2) gene, which encodes the first enzyme in the heme synthesis pathway in red blood cells. The second most prevalent form of CSA is caused by a mutation in the Solute carrier family 25 member 38 (SLC25A38) gene, which codes for an erythroid-specific protein of the inner mitochondrial membrane. Additionally, 15-20 genes are altogether associated with CSA. In this study, we aim to identify the CSA patients, understand their genetics and establish genotype-phenotype correlation. We have identified fifteen cases of CSA using our targeted NGS (t-NGS) panel. The major clinical findings in our cohort were microcytic anaemia, ring sideroblasts, and dyserythropoiesis in the bone marrow. Currently, two patients are responsive to pyridoxine, while the rest are on blood transfusion support. We have identified ten variants in three different genes of CSA (ALAS2, SLC25A38 & HSPA9). Five patients harbour four hemizygous variants- p.Ala282Ser, p.Arg170Cys, p.Arg204Gln and exon 2 duplication in the ALAS2 gene. In seven patients, we have identified three homozygous mutations - p.Pro190Arg, p.Arg187Gln and p.Arg134Cys in the SLC25A38 gene. These mutations have been predominantly identified in the European population. Three patients revealed three heterozygous variants p. Thr463Ile, D326Tyr, and Arg284Trp in the HSPA9 gene. PyMoL was used to evaluate the functional effects of these variations and understand their effect on the structure of the protein. We believe that by combining a bone marrow examination with genetic sequencing, CSA patients can acquire a definitive diagnosis.
Collapse
Affiliation(s)
- Rashmi Dongerdiye
- Department of Haematogenetics, ICMR-National Institute of Immunohematology, Indian Council of Medical Research, 13th Floor, NMS Building, King Edward Memorial (KEM.) Hospital Campus, Parel, Mumbai, 400012, India
| | - Prabhakar S Kedar
- Department of Haematogenetics, ICMR-National Institute of Immunohematology, Indian Council of Medical Research, 13th Floor, NMS Building, King Edward Memorial (KEM.) Hospital Campus, Parel, Mumbai, 400012, India.
| | - Arati Saptarshi
- Department of Haematogenetics, ICMR-National Institute of Immunohematology, Indian Council of Medical Research, 13th Floor, NMS Building, King Edward Memorial (KEM.) Hospital Campus, Parel, Mumbai, 400012, India
| | - Abhilasha Sampagar
- Pediatric Hematologist Oncologist, KLE'S Dr Prabhakar Kore Hospital, Belagavi, Karnataka, India
| | | | - Sangeeta Mudaliar
- Department of Haematology-Oncology, B J Wadia Hospital, Parel, Mumbai, 400012, India
| | - Purva Kanvinde
- Department of Haematology-Oncology, B J Wadia Hospital, Parel, Mumbai, 400012, India
| | - Mukesh Desai
- Department of Haematology-Oncology, B J Wadia Hospital, Parel, Mumbai, 400012, India
| | - Manisha Madkaikar
- Department of Haematogenetics, ICMR-National Institute of Immunohematology, Indian Council of Medical Research, 13th Floor, NMS Building, King Edward Memorial (KEM.) Hospital Campus, Parel, Mumbai, 400012, India
| |
Collapse
|
8
|
Ducamp S, Sendamarai AK, Campagna DR, Chin DWL, Fujiwara Y, Schmidt PJ, Fleming MD. Murine models of erythroid 5ALA synthesis disorders and their conditional synthetic lethal dependency on pyridoxine. Blood 2024; 144:1418-1432. [PMID: 38900972 DOI: 10.1182/blood.2023023078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
ABSTRACT X-linked sideroblastic anemia (XLSA) and X-linked protoporphyria (XLPP) are uncommon diseases caused by loss-of-function and gain-of-function mutations, respectively, in the erythroid form of 5-aminolevulinic acid synthetase (ALAS), ALAS2, which encodes the first enzyme in heme biosynthesis. A related congenital sideroblastic anemia (CSA) is due to mutations in SLC25A38 (solute carrier family 25 member A38), which supplies mitochondrial glycine for ALAS2 (SLC25A38-CSA). The lack of viable animal models has limited the studies on pathophysiology and development of therapies for these conditions. Here, using CRISPR-CAS9 gene editing technology, we have generated knockin mouse models that recapitulate the main features of XLSA and XLPP; and using conventional conditional gene targeting in embryonic stem cells, we also developed a faithful model of the SLC25A38-CSA. In addition to examining the phenotypes and natural history of each disease, we determine the effect of restriction or supplementation of dietary pyridoxine (vitamin B6), the essential cofactor of ALAS2, on the anemia and porphyria. In addition to the well-documented response of XLSA mutations to pyridoxine supplementation, we also demonstrate the relative insensitivity of the XLPP/EPP protoporphyrias, severe sensitivity of the XLSA models, and an extreme hypersensitivity of the SLC25A38-CSA model to pyridoxine deficiency, a phenotype that is not shared with another mouse hereditary anemia model, Hbbth3/+ β-thalassemia intermedia. Thus, in addition to generating animal models useful for examining the pathophysiology and treatment of these diseases, we have uncovered an unsuspected conditional synthetic lethality between the heme synthesis-related CSAs and pyridoxine deficiency. These findings have the potential to inform novel therapeutic paradigms for the treatment of these diseases.
Collapse
Affiliation(s)
- Sarah Ducamp
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Anoop K Sendamarai
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Dean R Campagna
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | | | - Yuko Fujiwara
- Division of Hematology/Oncology at Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Paul J Schmidt
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Fujiwara T, Harigae H. Pyridoxine: beyond a cofactor of ALAS. Blood 2024; 144:1358-1359. [PMID: 39325482 DOI: 10.1182/blood.2024025575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
|
10
|
Marin MJ, van Wijk XMR, Boothe PD, Harris NS, Winter WE. An Introduction to the Complete Blood Count for Clinical Chemists: Red Blood Cells. J Appl Lab Med 2024; 9:1025-1039. [PMID: 38646908 DOI: 10.1093/jalm/jfae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 03/06/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND The most frequently ordered laboratory test worldwide is the complete blood count (CBC). CONTENT In this primer, the red blood cell test components of the CBC are introduced, followed by a discussion of the laboratory evaluation of anemia and polycythemia. SUMMARY As clinical chemists are increasingly tasked to direct laboratories outside of the traditional clinical chemistry sections such as hematology, expertise must be developed. This review article is a dedication to that effort.
Collapse
Affiliation(s)
- Maximo J Marin
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | | | - Paul D Boothe
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - Neil S Harris
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| | - William E Winter
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
11
|
Cai J, Liu T, Huang Y, Chen H, Yu M, Zhang D, Huang Z. A novel and apparent de novo ALAS2 missense variant associated with congenital sideroblastic anemia. Front Pediatr 2024; 12:1411676. [PMID: 39281190 PMCID: PMC11394181 DOI: 10.3389/fped.2024.1411676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/19/2024] [Indexed: 09/18/2024] Open
Abstract
Background Congenital sideroblastic anemia (CSA) constitutes a group of inherited erythropoietic disorders. Some affect mainly or exclusively erythroid cells; other syndromic forms occur within multisystem disorders with extensive nonhematopoietic manifestations. In this study, we have performed clinical and molecular investigations on a 10-year-old boy suspected of having CSA. Methods Routine blood examination, peripheral blood and bone marrow smears, and serum iron tests were performed. Gene mutation analysis was conducted using whole-exome sequencing (WES) and the results were confirmed using Sanger sequencing. Furthermore, the functional impact of the identified variant was assessed/predicted with bioinformatics methods. Results The patient presented with severe microcytic anemia (hemoglobin, 50 g/L), iron overload and ring sideroblasts in the bone marrow. Moreover, WES revealed the presence of a hemizygous missense variant in ALAS2 (c.1102C > T), changing an encoded arginine to tryptophan (p. Arg368Trp). This variant was verified via Sanger sequencing, and neither of the parents carried this variant, which was suspected to be a de novo variant. Using in silico analysis with four different software programs, the variant was predicted to be harmful. PyMol and LigPlot software showed that the p. Arg368Trp variant may result in changes in hydrogen bonds. The patient was treated with vitamin B6 combined with deferasirox. After 6 months, the hemoglobin increased to 99 g/L and the serum ferritin decreased significantly. Conclusion We report a novel pathogenic variant in the ALAS2 gene (c.1102C > T:p. Arg368Trp), which caused CSA in a 10-year-old boy. Mutational analysis is important in patients with CSA when family history data are unavailable. Anemia due to the ALAS2 Arg368Trp variant responds to pyridoxine supplements.
Collapse
Affiliation(s)
- Jianling Cai
- Department of Pediatrics, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Tianming Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yuxuan Huang
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Hongxing Chen
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Meidie Yu
- Department of Clinical Medicine, Shantou University Medical College, Shantou, Guangdong, China
| | - Dongqing Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zhanqin Huang
- Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
12
|
Wang B, Shi D, Yang S, Lian Y, Li H, Cao M, He Y, Zhang L, Qiu C, Liu T, Wen W, Ma Y, Shi L, Cheng T, Shi L, Yuan W, Chu Y, Shi J. Mitochondrial tRNA pseudouridylation governs erythropoiesis. Blood 2024; 144:657-671. [PMID: 38635773 DOI: 10.1182/blood.2023022004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/20/2024] Open
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.
Collapse
Affiliation(s)
- Bichen Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Deyang Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuang Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yu Lian
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Regenerative Medicine Clinic and Red Blood Cell Diseases Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Haoyuan Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mutian Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yifei He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lele Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Regenerative Medicine Clinic and Red Blood Cell Diseases Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Chen Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tong Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Wen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuanwu Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Shi
- Key Laboratory of Breast Cancer Prevention and Therapy (Ministry of Education), Haihe Laboratory of Cell Ecosystem, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yajing Chu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jun Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
- Regenerative Medicine Clinic and Red Blood Cell Diseases Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
13
|
Gomes RAMP, Santos AL, Catarino L. Using portable X-ray fluorescence elemental analysis to explore porous skeletal lesions: Interplay of sex, age at death, and cause of death. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2024; 184:e24954. [PMID: 38747122 DOI: 10.1002/ajpa.24954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/04/2024] [Accepted: 04/29/2024] [Indexed: 07/10/2024]
Abstract
OBJECTIVES Search for possible associations between bone elemental concentration and the presence of porous skeletal lesions (PSLs), considering the sex, age, and cause of death (COD) of the individuals. MATERIALS AND METHODS The sample comprised 107 non-adult individuals (56 females, 51 males) aged 0-20 (x̄ = 13.2, SD = 5.8) from the Coimbra and Lisbon Identified Skeletal Collections. Cribra cranii, orbitalia, humeralis, and femoralis were recorded as present/absent, and elemental concentrations were assessed by portable x-ray fluorescence (pXRF). A multivariate statistical approach was applied. RESULTS Well-preserved skeletons with minimal diagenesis showed no sex-related elemental variations or PSL associations. In contrast, age-at-death correlated with elevated Ca, P, Sr, and Pb levels. Cribra cranii increased with age while other cribra declined post-adolescence. Higher concentrations of Fe and lower of S were linked to cribra cranii. Respiratory infections as COD increased the odds of expressing cribra femoralis (OR = 5.25, CI = 1.25-15.14), cribra cranii (OR = 2.91, CI = 0.97-8.69), and cribra orbitalia (OR = 2.76, CI = 1.06-7.24). DISCUSSION Feasible pXRF results and low cribra intraobserver error assure replicability. Elevated Ca, P, and Sr in older individuals may relate to skeletal growth, while increased Pb suggests bioaccumulation. Cribra's increase with age reflects different rates of marrow conversion and bone remodeling. Higher Fe and lower S in individuals with cribra cranii possibly reflects poor nutrition, early alcohol use, and sideroblastic anemia, aligning with 19th-20th-century Portugal's living conditions. Respiratory infections increased cribra expression, revealing intricate interplays among inflammation, anemia(s), marrow expansion, and diet. This research highlights a complex scenario and blazes a new path for cribra interpretation.
Collapse
Affiliation(s)
- Ricardo A M P Gomes
- Department of Life Sciences, Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
- Carrera de Antropología, University of Concepción, Barrio Universitário S/N, Concepción, Chile
| | - Ana Luisa Santos
- Department of Life Sciences, Research Centre for Anthropology and Health (CIAS), University of Coimbra, Coimbra, Portugal
| | - Lidia Catarino
- Department of Earth Sciences, Geosciences Center (CGeo), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
14
|
Selvanathan A, Teo J, Parayil Sankaran B. Hematologic Manifestations in Primary Mitochondrial Diseases. J Pediatr Hematol Oncol 2024; 46:e338-e347. [PMID: 38857202 DOI: 10.1097/mph.0000000000002890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/23/2024] [Indexed: 06/12/2024]
Abstract
Primary mitochondrial disorders (PMDs) are known for their pleiotropic manifestations in humans, affecting almost any organ or system at any time. Hematologic manifestations, such as cytopenias and sideroblastic anemia, occur in 10% to 30% of patients with confirmed PMDs. These can be the initial presenting features or complications that develop over time. Surveillance for these manifestations allows for prompt identification and treatment. This article provides an overview of the pathophysiology underpinning the hematologic effects of mitochondrial dysfunction, discussing the 3 key roles of the mitochondria in hematopoiesis: providing energy for cell differentiation and function, synthesizing heme, and generating iron-sulfur clusters. Subsequently, the diagnosis and management of mitochondrial disorders are discussed, focusing on hematologic manifestations and the specific conditions commonly associated with them. Through this, we aimed to provide a concise point of reference for those considering a mitochondrial cause for a patient's hematologic abnormality, or for those considering a hematologic manifestation in a patient with known or suspected mitochondrial disease.
Collapse
Affiliation(s)
- Arthavan Selvanathan
- Genetic Metabolic Disorders Service, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Juliana Teo
- Haematology Department, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Bindu Parayil Sankaran
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| |
Collapse
|
15
|
Ogunbileje JO, Harris N, Wynn T, Kashif R, Stover B, Osa-Andrews B. ATP-Binding Cassette Transporter of Clinical Significance: Sideroblastic Anemia. J Pers Med 2024; 14:636. [PMID: 38929857 PMCID: PMC11204910 DOI: 10.3390/jpm14060636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/16/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The ATP-binding cassette (ABC) transporters are a vast group of 48 membrane proteins, some of which are of notable physiological and clinical importance. Some ABC transporters are involved in functions such as the transport of chloride ions, bilirubin, reproductive hormones, cholesterol, and iron. Consequently, genetic or physiological disruption in these functions is manifested in various disease processes like cystic fibrosis, Tangier disease, and sideroblastic anemia. Among other etiologies, primary sideroblastic anemia results from a genetic mutation in the ATP-binding cassette-7 (ABCB7), a member of the ABC transporter family. There are not many articles specifically tackling the disease processes caused by ABC transporters in detail. Some testing methodologies previously reported in the available literature for investigating sideroblastic anemia need updating. Here, we expound on the relevance of ABCB7 as a clinically important ABC transporter and a rare participant in the disease process of Sideroblastic anemia. The other genetic and secondary etiologies of sideroblastic anemia, which do not involve mutations in the ABCB7 protein, are also described. We review the pathophysiology, clinical course, symptoms, diagnosis, and treatment of sideroblastic anemia with a focus on modern technologies for laboratory testing.
Collapse
Affiliation(s)
| | - Neil Harris
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Tung Wynn
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Reema Kashif
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Brian Stover
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | | |
Collapse
|
16
|
Vives Corrons JL. Understanding Rare Anemias: Emerging Frontiers for Diagnosis and Treatment. J Clin Med 2024; 13:3180. [PMID: 38892889 PMCID: PMC11172750 DOI: 10.3390/jcm13113180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Background-This review provides a comprehensive overview of rare anemias, emphasizing their hereditary and acquired causes, diagnostic advancements, and evolving treatment strategies. It outlines the significance of rare anemias within public health, historical challenges in recognition and treatment, and the role of European initiatives like ENERCA and EuroBloodNet in advancing care. Content-This document discusses diagnostic technologies like next-generation sequencing and the impact of artificial intelligence, alongside the promising avenues of gene therapy, targeted drug treatments, and stem cell transplantation. It underscores the importance of a patient-tailored approach, advances in diagnostic tools, and the necessity for continued research, patient advocacy, and international collaboration to improve outcomes for individuals with rare anemias.
Collapse
Affiliation(s)
- Joan-Lluis Vives Corrons
- Rare Anaemias and Erythropoietic Disorders Research, Institute for Leukaemia Research Josep Carreras, 08916 Barcelona, Spain;
- Ektacytometry Unit, Clinical Centre for Ambulatory Medicine, 08036 Barcelona, Spain
| |
Collapse
|
17
|
Liu G, Hou Y, Jin X, Zhang Y, Sun C, Huang C, Ren Y, Gao J, Wang X, Jiang X. PI3K/HSCB axis facilitates FOG1 nuclear translocation to promote erythropoiesis and megakaryopoiesis. eLife 2024; 13:RP95815. [PMID: 38757931 PMCID: PMC11101173 DOI: 10.7554/elife.95815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
Erythropoiesis and megakaryopoiesis are stringently regulated by signaling pathways. However, the precise molecular mechanisms through which signaling pathways regulate key transcription factors controlling erythropoiesis and megakaryopoiesis remain partially understood. Herein, we identified heat shock cognate B (HSCB), which is well known for its iron-sulfur cluster delivery function, as an indispensable protein for friend of GATA 1 (FOG1) nuclear translocation during erythropoiesis of K562 human erythroleukemia cells and cord-blood-derived human CD34+CD90+hematopoietic stem cells (HSCs), as well as during megakaryopoiesis of the CD34+CD90+HSCs. Mechanistically, HSCB could be phosphorylated by phosphoinositol-3-kinase (PI3K) to bind with and mediate the proteasomal degradation of transforming acidic coiled-coil containing protein 3 (TACC3), which otherwise detained FOG1 in the cytoplasm, thereby facilitating FOG1 nuclear translocation. Given that PI3K is activated during both erythropoiesis and megakaryopoiesis, and that FOG1 is a key transcription factor for these processes, our findings elucidate an important, previously unrecognized iron-sulfur cluster delivery independent function of HSCB in erythropoiesis and megakaryopoiesis.
Collapse
Affiliation(s)
- Gang Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Yunxuan Hou
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Yixue Zhang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Chaoyue Sun
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Chengquan Huang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Yujie Ren
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Jianmin Gao
- School of Chemistry, Northeast Normal UniversityChangchunChina
| | - Xiuli Wang
- School of Life Science, Northeast Normal UniversityChangchunChina
| | - Xiumei Jiang
- School of Chemistry, Northeast Normal UniversityChangchunChina
| |
Collapse
|
18
|
Giovannetti M, Rodríguez-Palero MJ, Fabrizio P, Nicolle O, Bedet C, Michaux G, Witting M, Artal-Sanz M, Palladino F. SIN-3 transcriptional coregulator maintains mitochondrial homeostasis and polyamine flux. iScience 2024; 27:109789. [PMID: 38746662 PMCID: PMC11091686 DOI: 10.1016/j.isci.2024.109789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/30/2024] [Accepted: 04/16/2024] [Indexed: 06/05/2024] Open
Abstract
Mitochondrial function relies on the coordinated transcription of mitochondrial and nuclear genomes to assemble respiratory chain complexes. Across species, the SIN3 coregulator influences mitochondrial functions, but how its loss impacts mitochondrial homeostasis and metabolism in the context of a whole organism is unknown. Exploring this link is important because SIN3 haploinsufficiency causes intellectual disability/autism syndromes and SIN3 plays a role in tumor biology. Here we show that loss of C. elegans SIN-3 results in transcriptional deregulation of mitochondrial- and nuclear-encoded mitochondrial genes, potentially leading to mito-nuclear imbalance. Consistent with impaired mitochondrial function, sin-3 mutants show extensive mitochondrial fragmentation by transmission electron microscopy (TEM) and in vivo imaging, and altered oxygen consumption. Metabolomic analysis of sin-3 mutant animals revealed a mitochondria stress signature and deregulation of methionine flux, resulting in decreased S-adenosyl methionine (SAM) and increased polyamine levels. Our results identify SIN3 as a key regulator of mitochondrial dynamics and metabolic flux, with important implications for human pathologies.
Collapse
Affiliation(s)
- Marina Giovannetti
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - María-Jesús Rodríguez-Palero
- Andalusian Centre for Developmental Biology (CABD), Consejo Superior de Investigaciones Científicas/Junta de Andalucía/Universidad Pablo de Olavide and Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Paola Fabrizio
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Ophélie Nicolle
- University Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, 35000 Rennes, France
| | - Cécile Bedet
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| | - Grégoire Michaux
- University Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, 35000 Rennes, France
| | - Michael Witting
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
- Chair of Analytical Food Chemistry, TUM School of Life Sciences, Technical University of Munich, Maximus-von-Imhof Forum 2, 85354 Freising, Weihenstephan, Germany
| | - Marta Artal-Sanz
- Andalusian Centre for Developmental Biology (CABD), Consejo Superior de Investigaciones Científicas/Junta de Andalucía/Universidad Pablo de Olavide and Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Francesca Palladino
- Laboratory of Biology and Modeling of the Cell, UMR5239 CNRS/Ecole Normale Supérieure de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, Université de Lyon, Lyon, France
| |
Collapse
|
19
|
Gomes RAMP, Catarino L, Santos AL. The Role of Fe, S, P, Ca, and Sr in Porous Skeletal Lesions: A Study on Non-adult Individuals Using pXRF. Biol Trace Elem Res 2024:10.1007/s12011-024-04187-4. [PMID: 38691307 DOI: 10.1007/s12011-024-04187-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 04/12/2024] [Indexed: 05/03/2024]
Abstract
Portable X-ray fluorescence is a new tool in the study of human bone. This research aims to investigate if variations in bone elemental concentrations are related with porous skeletal lesions (PSLs). One hundred well-preserved non-adult skeletons aged 0-11 years were selected from the archaeological site Convent of São Domingos, Lisbon (18th-19th century). Measuring a standard reference material and calculating the technical error of measurement assured elemental data reliability. Moreover, measuring soil samples excluded possible contamination of bones with elements from the soil, except for Pb. Additionally, the Ca/P ratio indicates maintenance of bone integrity. Cribra cranii, orbitalia, humeralis, and femoralis were recorded as present/absent, and the estimated intra-/inter-observer errors were low. The multivariate analysis found higher odds of having cribra orbitalia (OR = 1.76; CI = 0.97-3.20) and cribra femoralis (OR = 1.42; CI = 0.73-2.74) in individuals with lower Fe and higher S. Furthermore, higher levels of P, Ca, and Sr increased the odds of individuals developing cribra femoralis (OR = 2.30; CI = 1.23-4.29). Age also correlated with increased odds of exhibiting cribra orbitalia (OR = 1.86; CI = 0.94-3.68), cribra femoralis (OR = 6.97; CI = 2.78-17.45), and cribra humeralis (OR = 8.32; CI = 2.71-25.60). These findings suggest a shared etiology for these three cribras, contrasting with the higher Fe levels in individuals with cribra cranii. Lower Fe and higher S levels in individuals with cribra suggest a complex etiology, possibly involving conditions like megaloblastic or chronic disease anemia(s). Age-related elemental changes support the hypothesis that age influences cribra frequencies. This study highlights PSL complexity and opens new avenues for research.
Collapse
Affiliation(s)
- Ricardo A M P Gomes
- Research Centre for Anthropology and Health (CIAS), Department of Life Sciences, University of Coimbra, Rua Do Arco da Traição, 3000-056, Coimbra, Portugal.
- Carrera de Antropologia, University of Concepción, Barrio Universitário S/N, Concepción, Chile.
| | - Lidia Catarino
- Geosciences Center, Department of Earth Sciences, University of Coimbra, Rua Sílvio Lima - Pólo II, 3030-790, Coimbra, Portugal
| | - Ana Luisa Santos
- Research Centre for Anthropology and Health (CIAS), Department of Life Sciences, University of Coimbra, Rua Do Arco da Traição, 3000-056, Coimbra, Portugal
| |
Collapse
|
20
|
Dhonju K, Gautam A, Dahal A, Sharma NK, Adhikari D, Devkota L, Adhikari P, Tuladhar S, Pathak BD, Banmala S. Dolutegravir-induced acquired sideroblastic anemia in a HIV positive patient: A challenging hematologic complication. Clin Case Rep 2023; 11:e8301. [PMID: 38084360 PMCID: PMC10710374 DOI: 10.1002/ccr3.8301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/30/2023] [Accepted: 11/27/2023] [Indexed: 10/16/2024] Open
Abstract
Dolutegravir, the most recent antiretroviral drug with high efficacy, good tolerability, infrequent drug-drug interactions, and a favorable safety profile has not been reported in current literature as a cause of acquired sideroblastic anemia. Here, we present a 35-year-old male patient who was diagnosed with acquired sideroblastic anemia to Dolutegravir therapy.
Collapse
Affiliation(s)
- Kiran Dhonju
- Sukraraj Tropical and Infectious Disease HospitalKathmanduNepal
| | - Ashmita Gautam
- Tribhuvan University Institute of MedicineKathmanduNepal
| | - Abhinav Dahal
- Sukraraj Tropical and Infectious Disease HospitalKathmanduNepal
| | | | | | - Lina Devkota
- Sukraraj Tropical and Infectious Disease HospitalKathmanduNepal
| | | | | | | | - Sabin Banmala
- Nepalese Army Institute of Health SciencesKathmanduNepal
| |
Collapse
|
21
|
Ding Y, Yang K, Liu X, Xiao J, Li W, Zhong H. A Novel ALAS2 Mutation Causes Congenital Sideroblastic Anemia. Mediterr J Hematol Infect Dis 2023; 15:e2023062. [PMID: 38028395 PMCID: PMC10631716 DOI: 10.4084/mjhid.2023.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Affiliation(s)
- Yuxi Ding
- Department of Hematology, Zigong First People’s Hospital, Zigong, China
| | - Kun Yang
- Department of Hematology, Zigong First People’s Hospital, Zigong, China
| | - Xiaodong Liu
- Department of Hematology, Zigong First People’s Hospital, Zigong, China
| | - Jian Xiao
- Department of Hematology, Zigong First People’s Hospital, Zigong, China
| | - Wanting Li
- Department of Hematology, Zigong First People’s Hospital, Zigong, China
| | - Huixiu Zhong
- Department of Laboratory Medicine, Zigong First People’s Hospital, Zigong, China
| |
Collapse
|
22
|
Hoving V, Nijssen LE, Donker AE, Roelofs R, Schols SEM, Swinkels DW. Erythropoiesis-hepcidin-iron axis in patients with X-linked sideroblastic anaemia: An explorative biomarker study. Br J Haematol 2023; 202:1216-1219. [PMID: 37469032 DOI: 10.1111/bjh.18986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Vera Hoving
- Department of Haematology, Radboud university medical center, Nijmegen, The Netherlands
| | - Lieke E Nijssen
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Department of Paediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Albertine E Donker
- Department of Paediatrics, Máxima Medical Center, Veldhoven, The Netherlands
| | - Rian Roelofs
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Saskia E M Schols
- Department of Haematology, Radboud university medical center, Nijmegen, The Netherlands
| | - Dorine W Swinkels
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud university medical center, Nijmegen, The Netherlands
- Sanquin Blood Bank, Sanquin Diagnostics BV, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Qi Y, Hu M, Wang Z, Shang W. Mitochondrial iron regulation as an emerging target in ischemia/reperfusion injury during kidney transplantation. Biochem Pharmacol 2023; 215:115725. [PMID: 37524207 DOI: 10.1016/j.bcp.2023.115725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/02/2023]
Abstract
The injury caused by ischemia and subsequent reperfusion (I/R) is inevitable during kidney transplantation and its current management remains unsatisfactory. Iron is considered to play a remarkable pathologic role in the initiation or progression of tissue damage induced by I/R, whereas the effects of iron-related therapy remain controversial owing to the complicated nature of iron's involvement in multiple biological processes. A significant portion of the cellular iron is located in the mitochondria, which exerts a central role in the development and progression of I/R injury. Recent studies of iron regulation associated with mitochondrial function represents a unique opportunity to improve our knowledge on the pathophysiology of I/R injury. However, the molecular mechanisms linking mitochondria to the iron homeostasis remain unclear. In this review, we provide a comprehensive analysis of the alterations to iron metabolism in I/R injury during kidney transplantation, analyze the current understanding of mitochondrial regulation of iron homeostasis and discussed its potential application in I/R injury. The elucidation of regulatory mechanisms regulating mitochondrial iron homeostasis will offer valuable insights into potential therapeutic targets for alleviating I/R injury with the ultimate aim of improving kidney graft outcomes, with potential implications that could also extend to acute kidney injury or other I/R injuries.
Collapse
Affiliation(s)
- Yuanbo Qi
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| | - Mingyao Hu
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China
| | - Zhigang Wang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| | - Wenjun Shang
- Department of Kidney Transplantation, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
24
|
Melo Arias AF, Escribano Serrat S, Martínez Nieto J, Medina Salazar F, Ropero Gradilla P, Benavente Cuesta C, González Fernández FA. Two new mutations in the GLRX5 gene cause sideroblastic anemia. Blood Cells Mol Dis 2023; 102:102763. [PMID: 37301020 DOI: 10.1016/j.bcmd.2023.102763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Affiliation(s)
| | - Silvia Escribano Serrat
- Department of Hematology and Hemotherapy, Hospital Clínico San Carlos, IdiSSC, Madrid, Spain.
| | - Jorge Martínez Nieto
- Department of Hematology and Hemotherapy, Hospital Clínico San Carlos, IdiSSC, Madrid, Spain
| | - Fiorella Medina Salazar
- Department of Hematology and Hemotherapy, Hospital Clínico San Carlos, IdiSSC, Madrid, Spain
| | - Paloma Ropero Gradilla
- Department of Hematology and Hemotherapy, Hospital Clínico San Carlos, IdiSSC, Madrid, Spain
| | - Celina Benavente Cuesta
- Department of Hematology and Hemotherapy, Hospital Clínico San Carlos, IdiSSC, Madrid, Spain
| | | |
Collapse
|
25
|
Zhou J, Zhang H, Qin Y, Liu T. Severe Microcytic Anemia Caused by Complex Hereditary Spherocytosis and X-Linked Sideroblastic Anemia with Mutations in SPTB and ALAS2 Genes. J Clin Med 2023; 12:jcm12051990. [PMID: 36902777 PMCID: PMC10004689 DOI: 10.3390/jcm12051990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/17/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
We report a case of severe anemia caused by complex hereditary spherocytosis (HS) and X-linked sideroblastic anemia (XLSA) with two mutations in the spectrin beta (SPTB) and 5-aminolevulinic acid synthase (ALAS2) genes. The proband was a 16-year-old male with severe jaundice and microcytic hypochromic anemia since his childhood. He had more severe anemia requiring erythrocyte transfusion, and had no response to vitamin B6 treatment. Next-generation sequencing (NGS) revealed double heterozygous mutations, one in exon 19 (c.3936G > A:p.W1312X) of the SPTB gene and another in exon 2 (c.37A > G:p.K13E) of the ALAS2 gene, and confirmed again by Sanger sequencing. The mutation of ALAS2 (c.37A > G) is inherited from his asymptomatic heterozygous mother, causing amino acid p.K13E, and the mutation has not yet been reported. The mutation of SPTB (c.3936G > A) is a nonsense mutation, leading to a premature termination codon in exon 19, and the mutation in the SPTB gene is not found in any of his relatives, which indicates a de novo monoallelic mutation. Conclusions: The double heterozygous mutations in the SPTB and ALAS2 genes lead to the joint occurrence of HS and XLSA in this patient, and are implicated in the more severe clinical phenotypes.
Collapse
Affiliation(s)
| | | | | | - Ting Liu
- Correspondence: ; Tel.: +86-18980601240
| |
Collapse
|
26
|
Watanabe A, Fujiwara T, Ohta A, Shimizu Y, Tanaka R. HSPA9 frameshift and loss-of-function mutations in a patient manifesting syndromic sideroblastic anemia and congenital anomalies. Pediatr Blood Cancer 2023; 70:e29971. [PMID: 36094340 DOI: 10.1002/pbc.29971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 02/06/2023]
Affiliation(s)
- Atsuko Watanabe
- Department of Pediatric Oncology/Hematology, International Medical Center, Saitama Medical University, Hidaka, Japan
| | - Tohru Fujiwara
- Department of Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsuhiko Ohta
- Department of Pediatric Oncology/Hematology, International Medical Center, Saitama Medical University, Hidaka, Japan
| | - Yuki Shimizu
- Department of Pediatric Oncology/Hematology, International Medical Center, Saitama Medical University, Hidaka, Japan
| | - Ryuhei Tanaka
- Department of Pediatric Oncology/Hematology, International Medical Center, Saitama Medical University, Hidaka, Japan
| |
Collapse
|
27
|
Silvestri L, Pettinato M, Furiosi V, Bavuso Volpe L, Nai A, Pagani A. Managing the Dual Nature of Iron to Preserve Health. Int J Mol Sci 2023; 24:ijms24043995. [PMID: 36835406 PMCID: PMC9961779 DOI: 10.3390/ijms24043995] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/09/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Because of its peculiar redox properties, iron is an essential element in living organisms, being involved in crucial biochemical processes such as oxygen transport, energy production, DNA metabolism, and many others. However, its propensity to accept or donate electrons makes it potentially highly toxic when present in excess and inadequately buffered, as it can generate reactive oxygen species. For this reason, several mechanisms evolved to prevent both iron overload and iron deficiency. At the cellular level, iron regulatory proteins, sensors of intracellular iron levels, and post-transcriptional modifications regulate the expression and translation of genes encoding proteins that modulate the uptake, storage, utilization, and export of iron. At the systemic level, the liver controls body iron levels by producing hepcidin, a peptide hormone that reduces the amount of iron entering the bloodstream by blocking the function of ferroportin, the sole iron exporter in mammals. The regulation of hepcidin occurs through the integration of multiple signals, primarily iron, inflammation and infection, and erythropoiesis. These signals modulate hepcidin levels by accessory proteins such as the hemochromatosis proteins hemojuvelin, HFE, and transferrin receptor 2, the serine protease TMPRSS6, the proinflammatory cytokine IL6, and the erythroid regulator Erythroferrone. The deregulation of the hepcidin/ferroportin axis is the central pathogenic mechanism of diseases characterized by iron overload, such as hemochromatosis and iron-loading anemias, or by iron deficiency, such as IRIDA and anemia of inflammation. Understanding the basic mechanisms involved in the regulation of hepcidin will help in identifying new therapeutic targets to treat these disorders.
Collapse
Affiliation(s)
- Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-0226436889; Fax: +39-0226434723
| | - Mariateresa Pettinato
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Valeria Furiosi
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Letizia Bavuso Volpe
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonella Nai
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
28
|
Ma Z, Li D, Yang X, Liang J, Zhu Y. Case report: An infant boy with X-linked sideroblastic anaemia successfully treated by umbilical cord blood haematopoietic stem cell transplantation. Front Genet 2022; 13:1009988. [DOI: 10.3389/fgene.2022.1009988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
X-linked sideroblastic anaemia (XLSA) is an inherited disorder caused by mutations in genes encoding proteins involved in the biosynthesis of haem. The pathogenic gene, as well as the pathogenesis and diagnosis of XLSA, have been fully elucidated in previous studies. However, only a few new advances have been made in managing XLSA in recent years, and blood transfusion remains the primary treatment. We report a case of umbilical cord blood haematopoietic stem cell transplantation in a male infant diagnosed with XLSA who was born with asphyxia due to severe anaemia. Early hepatic vein occlusion occurred after transplantation. However, this complication was rapidly controlled after active treatment, and the child’s quality of life improved significantly. Haematopoietic stem cell transplantation is a promising alternative treatment for XLSA.
Collapse
|
29
|
Nasr W, Filippi MD. Acquired and hereditary bone marrow failure: A mitochondrial perspective. Front Oncol 2022; 12:1048746. [PMID: 36408191 PMCID: PMC9666693 DOI: 10.3389/fonc.2022.1048746] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022] Open
Abstract
The disorders known as bone marrow failure syndromes (BMFS) are life-threatening disorders characterized by absence of one or more hematopoietic lineages in the peripheral blood. Myelodysplastic syndromes (MDS) are now considered BMF disorders with associated cellular dysplasia. BMFs and MDS are caused by decreased fitness of hematopoietic stem cells (HSC) and poor hematopoiesis. BMF and MDS can occur de novo or secondary to hematopoietic stress, including following bone marrow transplantation or myeloablative therapy. De novo BMF and MDS are usually associated with specific genetic mutations. Genes that are commonly mutated in BMF/MDS are in DNA repair pathways, epigenetic regulators, heme synthesis. Despite known and common gene mutations, BMF and MDS are very heterogenous in nature and non-genetic factors contribute to disease phenotype. Inflammation is commonly found in BMF and MDS, and contribute to ineffective hematopoiesis. Another common feature of BMF and MDS, albeit less known, is abnormal mitochondrial functions. Mitochondria are the power house of the cells. Beyond energy producing machinery, mitochondrial communicate with the rest of the cells via triggering stress signaling pathways and by releasing numerous metabolite intermediates. As a result, mitochondria play significant roles in chromatin regulation and innate immune signaling pathways. The main goal of this review is to investigate BMF processes, with a focus mitochondria-mediated signaling in acquired and inherited BMF.
Collapse
Affiliation(s)
- Waseem Nasr
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, OH, United States,University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati, OH, United States,University of Cincinnati College of Medicine, Cincinnati, OH, United States,*Correspondence: Marie-Dominique Filippi,
| |
Collapse
|
30
|
Alam MM, Alathaibi A, Attar RA, Kashif M, Al-Ghamdi HS, Alharthi SA, Bokhary A, Althomali M. Pyridoxine Refractory Sideroblastic Anemia: Diagnosis and Misdiagnosis. CLINICAL PEDIATRIC HEMATOLOGY-ONCOLOGY 2022. [DOI: 10.15264/cpho.2022.29.2.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
| | - Abdulrhman Alathaibi
- Department of Hematology/Oncology, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | - Ruwayd Adel Attar
- Department of Hematology/Oncology, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | - Muhammad Kashif
- Department of Laboratory, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | | | | | | | - Muteb Althomali
- Department of Hematology/Oncology, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| |
Collapse
|
31
|
Abstract
A 57-year-old gentleman presented to the hospital with progressive fatigue and dyspnea on exertion three months after recovering from COVID-19. He was noted to have severe anemia with reticulocytopenia. After excluding vitamin deficiencies and heavy metal toxicities, a bone marrow aspirate and biopsy were performed, which showed erythroid predominant trilineage maturing hematopoiesis with 79% ring sideroblasts and no dysplasia. SF3B1 mutation was negative. He was diagnosed with sideroblastic anemia and became transfusion-dependent. He was treated with an erythropoiesis-stimulating agent and luspatercept with transient improvement in anemia. After 12 months of treatment, anemia spontaneously improved. Repeat bone marrow biopsy showed hypercellular marrow with 39% ringed sideroblasts. We suspect that this possibly was a delayed manifestation of COVID-19 infection.
Collapse
|
32
|
Campbell T, Slone J, Huang T. Mitochondrial Genome Variants as a Cause of Mitochondrial Cardiomyopathy. Cells 2022; 11:cells11182835. [PMID: 36139411 PMCID: PMC9496904 DOI: 10.3390/cells11182835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondria are small double-membraned organelles responsible for the generation of energy used in the body in the form of ATP. Mitochondria are unique in that they contain their own circular mitochondrial genome termed mtDNA. mtDNA codes for 37 genes, and together with the nuclear genome (nDNA), dictate mitochondrial structure and function. Not surprisingly, pathogenic variants in the mtDNA or nDNA can result in mitochondrial disease. Mitochondrial disease primarily impacts tissues with high energy demands, including the heart. Mitochondrial cardiomyopathy is characterized by the abnormal structure or function of the myocardium secondary to genetic defects in either the nDNA or mtDNA. Mitochondrial cardiomyopathy can be isolated or part of a syndromic mitochondrial disease. Common manifestations of mitochondrial cardiomyopathy are a phenocopy of hypertrophic cardiomyopathy, dilated cardiomyopathy, and cardiac conduction defects. The underlying pathophysiology of mitochondrial cardiomyopathy is complex and likely involves multiple abnormal processes in the cell, stemming from deficient oxidative phosphorylation and ATP depletion. Possible pathophysiology includes the activation of alternative metabolic pathways, the accumulation of reactive oxygen species, dysfunctional mitochondrial dynamics, abnormal calcium homeostasis, and mitochondrial iron overload. Here, we highlight the clinical assessment of mtDNA-related mitochondrial cardiomyopathy and offer a novel hypothesis of a possible integrated, multivariable pathophysiology of disease.
Collapse
|
33
|
Iron Metabolism in the Disorders of Heme Biosynthesis. Metabolites 2022; 12:metabo12090819. [PMID: 36144223 PMCID: PMC9505951 DOI: 10.3390/metabo12090819] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 01/19/2023] Open
Abstract
Given its remarkable property to easily switch between different oxidative states, iron is essential in countless cellular functions which involve redox reactions. At the same time, uncontrolled interactions between iron and its surrounding milieu may be damaging to cells and tissues. Heme—the iron-chelated form of protoporphyrin IX—is a macrocyclic tetrapyrrole and a coordination complex for diatomic gases, accurately engineered by evolution to exploit the catalytic, oxygen-binding, and oxidoreductive properties of iron while minimizing its damaging effects on tissues. The majority of the body production of heme is ultimately incorporated into hemoglobin within mature erythrocytes; thus, regulation of heme biosynthesis by iron is central in erythropoiesis. Additionally, heme is a cofactor in several metabolic pathways, which can be modulated by iron-dependent signals as well. Impairment in some steps of the pathway of heme biosynthesis is the main pathogenetic mechanism of two groups of diseases collectively known as porphyrias and congenital sideroblastic anemias. In porphyrias, according to the specific enzyme involved, heme precursors accumulate up to the enzyme stop in disease-specific patterns and organs. Therefore, different porphyrias manifest themselves under strikingly different clinical pictures. In congenital sideroblastic anemias, instead, an altered utilization of mitochondrial iron by erythroid precursors leads to mitochondrial iron overload and an accumulation of ring sideroblasts in the bone marrow. In line with the complexity of the processes involved, the role of iron in these conditions is then multifarious. This review aims to summarise the most important lines of evidence concerning the interplay between iron and heme metabolism, as well as the clinical and experimental aspects of the role of iron in inherited conditions of altered heme biosynthesis.
Collapse
|
34
|
Rodriguez-Sevilla JJ, Calvo X, Arenillas L. Causes and Pathophysiology of Acquired Sideroblastic Anemia. Genes (Basel) 2022; 13:1562. [PMID: 36140729 PMCID: PMC9498732 DOI: 10.3390/genes13091562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 01/19/2023] Open
Abstract
The sideroblastic anemias are a heterogeneous group of inherited and acquired disorders characterized by anemia and the presence of ring sideroblasts in the bone marrow. Ring sideroblasts are abnormal erythroblasts with iron-loaded mitochondria that are visualized by Prussian blue staining as a perinuclear ring of green-blue granules. The mechanisms that lead to the ring sideroblast formation are heterogeneous, but in all of them, there is an abnormal deposition of iron in the mitochondria of erythroblasts. Congenital sideroblastic anemias include nonsyndromic and syndromic disorders. Acquired sideroblastic anemias include conditions that range from clonal disorders (myeloid neoplasms as myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms with ring sideroblasts) to toxic or metabolic reversible sideroblastic anemia. In the last 30 years, due to the advances in genomic techniques, a deep knowledge of the pathophysiological mechanisms has been accomplished and the bases for possible targeted treatments have been established. The distinction between the different forms of sideroblastic anemia is based on the study of the characteristics of the anemia, age of diagnosis, clinical manifestations, and the performance of laboratory analysis involving genetic testing in many cases. This review focuses on the differential diagnosis of acquired disorders associated with ring sideroblasts.
Collapse
Affiliation(s)
| | - Xavier Calvo
- Laboratori de Citologia Hematològica, Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
- Group of Translational Research on Hematological Neoplasms (GRETNHE), IMIM-Hospital del Mar, 08003 Barcelona, Spain
| | - Leonor Arenillas
- Laboratori de Citologia Hematològica, Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
- Group of Translational Research on Hematological Neoplasms (GRETNHE), IMIM-Hospital del Mar, 08003 Barcelona, Spain
| |
Collapse
|
35
|
Abstract
Heme (protoheme IX) is an essential cofactor for a large variety of proteins whose functions vary from one electron reactions to binding gases. While not ubiquitous, heme is found in the great majority of known life forms. Unlike most cofactors that are acquired from dietary sources, the vast majority of organisms that utilize heme possess a complete pathway to synthesize the compound. Indeed, dietary heme is most frequently utilized as an iron source and not as a source of heme. In Nature there are now known to exist three pathways to synthesize heme. These are the siroheme dependent (SHD) pathway which is the most ancient, but least common of the three; the coproporphyrin dependent (CPD) pathway which with one known exception is found only in gram positive bacteria; and the protoporphyrin dependent (PPD) pathway which is found in gram negative bacteria and all eukaryotes. All three pathways share a core set of enzymes to convert the first committed intermediate, 5-aminolevulinate (ALA) into uroporphyrinogen III. In the current review all three pathways are reviewed as well as the two known pathways to synthesize ALA. In addition, interesting features of some heme biosynthesis enzymes are discussed as are the regulation and disorders of heme biosynthesis.
Collapse
Affiliation(s)
- Harry A Dailey
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602-1111, USA
- Department of Microbiology, University of Georgia, Athens, GA 30602-1111, USA
| | - Amy E Medlock
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602-1111, USA
- Augusta University/University of Georgia Medical Partnership, University of Georgia, Athens, GA, USA
| |
Collapse
|
36
|
Exploring the mechanistic link between SF3B1 mutation and ring sideroblast formation in myelodysplastic syndrome. Sci Rep 2022; 12:14562. [PMID: 36028755 PMCID: PMC9418223 DOI: 10.1038/s41598-022-18921-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/22/2022] [Indexed: 12/02/2022] Open
Abstract
Acquired sideroblastic anemia, characterized by bone marrow ring sideroblasts (RS), is predominantly associated with myelodysplastic syndrome (MDS). Although somatic mutations in splicing factor 3b subunit 1 (SF3B1), which is involved in the RNA splicing machinery, are frequently found in MDS-RS, the detailed mechanism contributing to RS formation is unknown. To explore the mechanism, we established human umbilical cord blood-derived erythroid progenitor-2 (HUDEP-2) cells stably expressing SF3B1K700E. SF3B1K700E expressing cells showed higher proportion of RS than the control cells along with erythroid differentiation, indicating the direct contribution of mutant SF3B1 expression in erythroblasts to RS formation. In SF3B1K700E expressing cells, ABCB7 and ALAS2, known causative genes for congenital sideroblastic anemia, were downregulated. Additionally, mis-splicing of ABCB7 was observed in SF3B1K700E expressing cells. ABCB7-knockdown HUDEP-2 cells revealed an increased frequency of RS formation along with erythroid differentiation, demonstrating the direct molecular link between ABCB7 defects and RS formation. ALAS2 protein levels were obviously decreased in ABCB7-knockdown cells, indicating decreased ALAS2 translation owing to impaired Fe–S cluster export by ABCB7 defects. Finally, RNA-seq analysis of MDS clinical samples demonstrated decreased expression of ABCB7 by the SF3B1 mutation. Our findings contribute to the elucidation of the complex mechanisms of RS formation in MDS-RS.
Collapse
|
37
|
Abstract
The World Health Organization estimates that approximately a quarter of the world's population suffers from anemia, including almost half of preschool-age children. Globally, iron deficiency anemia is the most common cause of anemia. Other important causes of anemia in children are hemoglobinopathies, infection, and other chronic diseases. Anemia is associated with increased morbidity, including neurologic complications, increased risk of low birth weight, infection, and heart failure, as well as increased mortality. When approaching a child with anemia, detailed historical information, particularly diet, environmental exposures, and family history, often yield important clues to the diagnosis. Dysmorphic features on physical examination may indicate syndromic causes of anemia. Diagnostic testing involves a stepwise approach utilizing various laboratory techniques. The increasing availability of genetic testing is providing new mechanistic insights into inherited anemias and allowing diagnosis in many previously undiagnosed cases. Population-based approaches are being taken to address nutritional anemias. Novel pharmacologic agents and advances in gene therapy-based therapeutics have the potential to ameliorate anemia-associated disease and provide treatment strategies even in the most difficult and complex cases.
Collapse
Affiliation(s)
- Patrick G Gallagher
- Departments of Pediatrics, Pathology, and Genetics, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
38
|
Girard S, Genevieve F, Rault E, Fenneteau O, Lesesve JF. When Ring Sideroblasts on Bone Marrow Smears Are Inconsistent with the Diagnosis of Myelodysplastic Neoplasms. Diagnostics (Basel) 2022; 12:1752. [PMID: 35885655 PMCID: PMC9320983 DOI: 10.3390/diagnostics12071752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/08/2022] [Accepted: 07/16/2022] [Indexed: 01/19/2023] Open
Abstract
Ring sideroblasts are commonly seen in myelodysplastic neoplasms and are a key condition for identifying distinct entities of myelodysplastic neoplasms according to the WHO classification. However, the presence of ring sideroblasts is not exclusive to myelodysplastic neoplasms. Ring sideroblasts are as well either encountered in non-clonal secondary acquired disorders, such as exposure to toxic substances, drug/medicine, copper deficiency, zinc overload, lead poison, or hereditary sideroblastic anemias related to X-linked, autosomal, or mitochondrial mutations. This review article will discuss diseases associated with ring sideroblasts outside the context of myelodysplastic neoplasms. Knowledge of the differential diagnoses characterized by the presence of ring sideroblasts in bone marrow is essential to prevent any misdiagnosis, which leads to delayed diagnosis and subsequent management of patients that differ in the different forms of sideroblastic anemia.
Collapse
Affiliation(s)
- Sandrine Girard
- Laboratory of Hematology, Center of Biology and Pathology East, Hospices Civils de Lyon, 69500 Bron, France
- French-Speaking Cellular Hematology Group, 69500 Bron, France; (F.G.); (E.R.); (O.F.); (J.-F.L.)
| | - Franck Genevieve
- French-Speaking Cellular Hematology Group, 69500 Bron, France; (F.G.); (E.R.); (O.F.); (J.-F.L.)
- Laboratory of Hematology, Angers University Hospital, 49100 Angers, France
| | - Emmanuelle Rault
- French-Speaking Cellular Hematology Group, 69500 Bron, France; (F.G.); (E.R.); (O.F.); (J.-F.L.)
- Department of Biological Hematology, Tours University Hospital, 37081 Tours, France
| | - Odile Fenneteau
- French-Speaking Cellular Hematology Group, 69500 Bron, France; (F.G.); (E.R.); (O.F.); (J.-F.L.)
- Laboratory of Hematology, Robert Debré Hospital, APHP, 75019 Paris, France
| | - Jean-François Lesesve
- French-Speaking Cellular Hematology Group, 69500 Bron, France; (F.G.); (E.R.); (O.F.); (J.-F.L.)
- Laboratory of Hematology, Nancy University Hospital, 54000 Nancy, France
| |
Collapse
|
39
|
Maio N, Rouault TA. Mammalian iron sulfur cluster biogenesis and human diseases. IUBMB Life 2022; 74:705-714. [PMID: 35098635 PMCID: PMC9247042 DOI: 10.1002/iub.2597] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 07/30/2023]
Affiliation(s)
- Nunziata Maio
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | - Tracey A Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| |
Collapse
|
40
|
Yien YY, Perfetto M. Regulation of Heme Synthesis by Mitochondrial Homeostasis Proteins. Front Cell Dev Biol 2022; 10:895521. [PMID: 35832791 PMCID: PMC9272004 DOI: 10.3389/fcell.2022.895521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/12/2022] [Indexed: 11/19/2022] Open
Abstract
Heme plays a central role in diverse, life-essential processes that range from ubiquitous, housekeeping pathways such as respiration, to highly cell-specific ones such as oxygen transport by hemoglobin. The regulation of heme synthesis and its utilization is highly regulated and cell-specific. In this review, we have attempted to describe how the heme synthesis machinery is regulated by mitochondrial homeostasis as a means of coupling heme synthesis to its utilization and to the metabolic requirements of the cell. We have focused on discussing the regulation of mitochondrial heme synthesis enzymes by housekeeping proteins, transport of heme intermediates, and regulation of heme synthesis by macromolecular complex formation and mitochondrial metabolism. Recently discovered mechanisms are discussed in the context of the model organisms in which they were identified, while more established work is discussed in light of technological advancements.
Collapse
|
41
|
Next generation sequencing for diagnosis of hereditary anemia: Experience in a Spanish reference center. Clin Chim Acta 2022; 531:112-119. [DOI: 10.1016/j.cca.2022.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/19/2022]
|
42
|
Congenital sideroblastic anemia model due to ALAS2 mutation is susceptible to ferroptosis. Sci Rep 2022; 12:9024. [PMID: 35637209 PMCID: PMC9151922 DOI: 10.1038/s41598-022-12940-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 05/18/2022] [Indexed: 11/20/2022] Open
Abstract
X-linked sideroblastic anemia (XLSA), the most common form of congenital sideroblastic anemia, is caused by a germline mutation in the erythroid-specific 5-aminolevulinate synthase (ALAS2) gene. In XLSA, defective heme biosynthesis leads to ring sideroblast formation because of excess mitochondrial iron accumulation. In this study, we introduced ALAS2 missense mutations on human umbilical cord blood-derived erythroblasts; hereafter, we refer to them as XLSA clones. XLSA clones that differentiated into mature erythroblasts showed an increased frequency of ring sideroblast formation with impaired hemoglobin biosynthesis. The expression profiling revealed significant enrichment of genes involved in ferroptosis, which is a form of regulated cell death induced by iron accumulation and lipid peroxidation. Notably, treatment with erastin, a ferroptosis inducer, caused a higher proportion of cell death in XLSA clones. XLSA clones exhibited significantly higher levels of intracellular lipid peroxides and enhanced expression of BACH1, a regulator of iron metabolism and potential accelerator of ferroptosis. In XLSA clones, BACH1 repressed genes involved in iron metabolism and glutathione synthesis. Collectively, defective heme biosynthesis in XLSA clones could confer enhanced BACH1 expression, leading to increased susceptibility to ferroptosis. The results of our study provide important information for the development of novel therapeutic targets for XLSA.
Collapse
|
43
|
The mutual crosstalk between iron and erythropoiesis. Int J Hematol 2022; 116:182-191. [PMID: 35618957 DOI: 10.1007/s12185-022-03384-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 04/26/2022] [Accepted: 05/06/2022] [Indexed: 02/08/2023]
Abstract
Iron homeostasis and erythropoiesis are strongly interconnected. On one side iron is essential to terminal erythropoiesis for hemoglobin production, on the other erythropoiesis may increase iron absorption through the production of erythroferrone, the erythroid hormone that suppresses hepcidin expression Also erythropoietin production is modulated by iron through the iron regulatory proteins-iron responsive elements that control the hypoxia inducible factor 2-α. The second transferrin receptor, an iron sensor both in the liver and in erythroid cells modulates erythropoietin sensitivity and is a further link between hepcidin and erythropoiesis. When erythropoietin is decreased in iron deficiency the erythropoietin sensitivity is increased because the second transferrin receptor is removed from cell surface. A deranged balance between erythropoiesis and iron/hepcidin may lead to anemia, as in the case of iron deficiency, defective iron uptake and erythroid utilization or subnormal recycling. Defective control of hepcidin production may cause iron deficiency, as in the recessive disorder iron refractory iron deficiency anemia or in anemia of inflammation, or in iron loading anemias, which are characterized by excessive but ineffective erythropoiesis. The elucidation of the mechanisms that regulates iron homeostasis and erythropoiesis is leading to the development of drugs for the benefit of both iron and erythropoiesis disorders.
Collapse
|
44
|
Girelli D, Busti F, Brissot P, Cabantchik I, Muckenthaler MU, Porto G. Hemochromatosis classification: update and recommendations by the BIOIRON Society. Blood 2022; 139:3018-3029. [PMID: 34601591 PMCID: PMC11022970 DOI: 10.1182/blood.2021011338] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022] Open
Abstract
Hemochromatosis (HC) is a genetically heterogeneous disorder in which uncontrolled intestinal iron absorption may lead to progressive iron overload (IO) responsible for disabling and life-threatening complications such as arthritis, diabetes, heart failure, hepatic cirrhosis, and hepatocellular carcinoma. The recent advances in the knowledge of pathophysiology and molecular basis of iron metabolism have highlighted that HC is caused by mutations in at least 5 genes, resulting in insufficient hepcidin production or, rarely, resistance to hepcidin action. This has led to an HC classification based on different molecular subtypes, mainly reflecting successive gene discovery. This scheme was difficult to adopt in clinical practice and therefore needs revision. Here we present recommendations for unambiguous HC classification developed by a working group of the International Society for the Study of Iron in Biology and Medicine (BIOIRON Society), including both clinicians and basic scientists during a meeting in Heidelberg, Germany. We propose to deemphasize the use of the molecular subtype criteria in favor of a classification addressing both clinical issues and molecular complexity. Ferroportin disease (former type 4a) has been excluded because of its distinct phenotype. The novel classification aims to be of practical help whenever a detailed molecular characterization of HC is not readily available.
Collapse
Affiliation(s)
- Domenico Girelli
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Pierre Brissot
- INSERM, Univ-Rennes, Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1241, Institut NuMeCan, Rennes, France
| | - Ioav Cabantchik
- Alexander Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel
| | - Martina U. Muckenthaler
- Department of Pediatric Oncology, Hematology, and Immunology and Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Mannheim, Germany
| | - Graça Porto
- Institute for Molecular and Cell Biology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Clinical Hematology, Santo António Hospital, Porto University, Porto, Portugal
| | - on behalf of the Nomenclature Committee of the International Society for the Study of Iron in Biology and Medicine (BIOIRON Society)
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
- INSERM, Univ-Rennes, Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1241, Institut NuMeCan, Rennes, France
- Alexander Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel
- Department of Pediatric Oncology, Hematology, and Immunology and Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Mannheim, Germany
- Institute for Molecular and Cell Biology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Clinical Hematology, Santo António Hospital, Porto University, Porto, Portugal
| |
Collapse
|
45
|
Clough CA, Pangallo J, Sarchi M, Ilagan JO, North K, Bergantinos R, Stolla MC, Naru J, Nugent P, Kim E, Stirewalt DL, Subramaniam AR, Abdel-Wahab O, Abkowitz JL, Bradley RK, Doulatov S. Coordinated missplicing of TMEM14C and ABCB7 causes ring sideroblast formation in SF3B1-mutant myelodysplastic syndrome. Blood 2022; 139:2038-2049. [PMID: 34861039 PMCID: PMC8972092 DOI: 10.1182/blood.2021012652] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 11/22/2021] [Indexed: 11/20/2022] Open
Abstract
SF3B1 splicing factor mutations are near-universally found in myelodysplastic syndromes (MDS) with ring sideroblasts (RS), a clonal hematopoietic disorder characterized by abnormal erythroid cells with iron-loaded mitochondria. Despite this remarkably strong genotype-to-phenotype correlation, the mechanism by which mutant SF3B1 dysregulates iron metabolism to cause RS remains unclear due to an absence of physiological models of RS formation. Here, we report an induced pluripotent stem cell model of SF3B1-mutant MDS that for the first time recapitulates robust RS formation during in vitro erythroid differentiation. Mutant SF3B1 induces missplicing of ∼100 genes throughout erythroid differentiation, including proposed RS driver genes TMEM14C, PPOX, and ABCB7. All 3 missplicing events reduce protein expression, notably occurring via 5' UTR alteration, and reduced translation efficiency for TMEM14C. Functional rescue of TMEM14C and ABCB7, but not the non-rate-limiting enzyme PPOX, markedly decreased RS, and their combined rescue nearly abolished RS formation. Our study demonstrates that coordinated missplicing of mitochondrial transporters TMEM14C and ABCB7 by mutant SF3B1 sequesters iron in mitochondria, causing RS formation.
Collapse
Affiliation(s)
- Courtnee A Clough
- Molecular and Cellular Biology Program and
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
| | - Joseph Pangallo
- Molecular and Cellular Biology Program and
- Computational Biology Program, Public Health Sciences Division and
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Martina Sarchi
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Janine O Ilagan
- Computational Biology Program, Public Health Sciences Division and
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Khrystyna North
- Computational Biology Program, Public Health Sciences Division and
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Rochelle Bergantinos
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
| | - Massiel C Stolla
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
| | - Jasmine Naru
- Seattle Cancer Care Alliance, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Patrick Nugent
- Molecular and Cellular Biology Program and
- Computational Biology Program, Public Health Sciences Division and
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Eunhee Kim
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY
- Department of Biological Sciences, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology, South Korea
| | - Derek L Stirewalt
- Seattle Cancer Care Alliance, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Arvind R Subramaniam
- Computational Biology Program, Public Health Sciences Division and
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York; and
| | - Janis L Abkowitz
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
- Department of Genome Sciences, University of Washington, Seattle, WA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle WA
| | - Robert K Bradley
- Computational Biology Program, Public Health Sciences Division and
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Sergei Doulatov
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
- Department of Genome Sciences, University of Washington, Seattle, WA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle WA
| |
Collapse
|
46
|
Vishwanathan V, D’Silva P. Loss of Function of mtHsp70 Chaperone Variants Leads to Mitochondrial Dysfunction in Congenital Sideroblastic Anemia. Front Cell Dev Biol 2022; 10:847045. [PMID: 35252210 PMCID: PMC8888832 DOI: 10.3389/fcell.2022.847045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Congenital Sideroblastic Anemias (CSA) is a group of rare genetic disorders characterized by the abnormal accumulation of iron in erythrocyte precursors. A common hallmark underlying these pathological conditions is mitochondrial dysfunction due to altered protein homeostasis, heme biosynthesis, and oxidative phosphorylation. A clinical study on congenital sideroblastic anemia has identified mutations in mitochondrial Hsp70 (mtHsp70/Mortalin). Mitochondrial Hsp70 plays a critical role in maintaining mitochondrial function by regulating several pathways, including protein import and folding, and iron-sulfur cluster synthesis. Owing to the structural and functional homology between human and yeast mtHsp70, we have utilized the yeast system to delineate the role of mtHsp70 variants in the etiology of CSA’s. Analogous mutations in yeast mtHsp70 exhibited temperature-sensitive growth phenotypes under non-respiratory and respiratory conditions. In vivo analyses indicate a perturbation in mitochondrial mass and functionality accompanied by an alteration in the organelle network and cellular redox levels. Preliminary in vitro biochemical studies of mtHsp70 mutants suggest impaired import function, altered ATPase activity and substrate interaction. Together, our findings suggest the loss of chaperone activity to be a pivotal factor in the pathophysiology of congenital sideroblastic anemia.
Collapse
|
47
|
Vobugari N, Chaturvedi M, Schlam-Camhi IM, Smith HP. Sideroblastic anaemia in a patient with sickle cell disease. BMJ Case Rep 2022; 15:15/2/e246623. [PMID: 35135795 PMCID: PMC8830102 DOI: 10.1136/bcr-2021-246623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Sideroblastic anaemia is a rare condition. We report a unique case of concomitant sideroblastic anaemia in a patient with sickle cell disease with long-standing blood transfusion history. Due to a low prevalence of sideroblastic anaemia, the diagnosis of sideroblastic anaemia is often difficult, especially when coexisting with common types of anaemia, including sickle cell disease. This case highlights the detrimental effects of anchoring bias. Rare causes of refractory anaemia should be considered in patients with haemoglobin disorders as the therapeutic approaches for these conditions are different. High suspicion on the part of the clinician and low threshold for workup of anaemia often aids in the diagnosis of coexisting conditions such as sideroblastic anaemia. Early diagnosis and treatment of sideroblastic anaemia improves patient outcomes and prevents long-term complications.
Collapse
Affiliation(s)
- Nikitha Vobugari
- Internal Medicine, MedStar Washington Hospital Center, Washington, DC, USA
| | - Mansi Chaturvedi
- Internal Medicine, MedStar Washington Hospital Center, Washington, DC, USA
| | - Ilana Miriam Schlam-Camhi
- Hematology/Oncology, Tufts Medical Center, Boston, Massachusetts, USA
- Hematology/Oncology, MedStar Washington Hospital Center, Washington, DC, USA
| | - Hedy Patricia Smith
- Hematology/Oncology, MedStar Washington Hospital Center, Washington, DC, USA
| |
Collapse
|
48
|
Beecher G, Fleming MD, Liewluck T. Hereditary myopathies associated with hematological abnormalities. Muscle Nerve 2022; 65:374-390. [PMID: 34985130 DOI: 10.1002/mus.27474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 01/19/2023]
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.
Collapse
Affiliation(s)
- Grayson Beecher
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Teerin Liewluck
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
49
|
Azacitidine is a potential therapeutic drug for pyridoxine-refractory female X-linked sideroblastic anemia. Blood Adv 2021; 6:1100-1114. [PMID: 34781359 PMCID: PMC8864662 DOI: 10.1182/bloodadvances.2021005664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/04/2021] [Indexed: 01/19/2023] Open
Abstract
A patient-derived iPSC model recapitulates defective erythroid maturation in female XLSA. Azacitidine reactivates the silent wild-type ALAS2 allele and ameliorates inefficient erythropoiesis in iPSC-derived HPCs from female XLSA.
X-linked sideroblastic anemia (XLSA) is associated with mutations in the erythroid-specific δ-aminolevulinic acid synthase (ALAS2) gene. Treatment of XLSA is mainly supportive, except in patients who are pyridoxine responsive. Female XLSA often represents a late onset of severe anemia, mostly related to the acquired skewing of X chromosome inactivation. In this study, we successfully generated active wild-type and mutant ALAS2-induced pluripotent stem cell (iPSC) lines from the peripheral blood cells of an affected mother and 2 daughters in a family with pyridoxine-resistant XLSA related to a heterozygous ALAS2 missense mutation (R227C). The erythroid differentiation potential was severely impaired in active mutant iPSC lines compared with that in active wild-type iPSC lines. Most of the active mutant iPSC-derived erythroblasts revealed an immature morphological phenotype, and some showed dysplasia and perinuclear iron deposits. In addition, globin and HO-1 expression and heme biosynthesis in active mutant erythroblasts were severely impaired compared with that in active wild-type erythroblasts. Furthermore, genes associated with erythroblast maturation and karyopyknosis showed significantly reduced expression in active mutant erythroblasts, recapitulating the maturation defects. Notably, the erythroid differentiation ability and hemoglobin expression of active mutant iPSC-derived hematopoietic progenitor cells (HPCs) were improved by the administration of δ-aminolevulinic acid, verifying the suitability of the cells for drug testing. Administration of a DNA demethylating agent, azacitidine, reactivated the silent, wild-type ALAS2 allele in active mutant HPCs and ameliorated the erythroid differentiation defects, suggesting that azacitidine is a potential novel therapeutic drug for female XLSA. Our patient-specific iPSC platform provides novel biological and therapeutic insights for XLSA.
Collapse
|
50
|
Goldenstein H, Shrem SB, Weiss O, Zeligson S, Segel R, Mory A, Weiss K. A rare rearrangement of 5q31.2 in a child with a neurodevelopmental syndrome. Clin Dysmorphol 2021; 30:181-185. [PMID: 34232135 DOI: 10.1097/mcd.0000000000000380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Hagit Goldenstein
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa
| | - Sara Beni Shrem
- Pediatric Neurodevelopment Service, Meuchedet Health Services
| | - Omri Weiss
- Medical Genetics Institute, Shaare Zedek Medical Center and Hebrew University-Hadassah School of Medicine, Jerusalem
| | - Sharon Zeligson
- Medical Genetics Institute, Shaare Zedek Medical Center and Hebrew University-Hadassah School of Medicine, Jerusalem
| | - Reeval Segel
- Medical Genetics Institute, Shaare Zedek Medical Center and Hebrew University-Hadassah School of Medicine, Jerusalem
| | - Adi Mory
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Karin Weiss
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|