1
|
Qin H, He Y, Xie Z. A 9-year-old child presenting with anemia accompanied by abnormal red blood cell morphology. Pract Lab Med 2025; 45:e00459. [PMID: 40236604 PMCID: PMC11997398 DOI: 10.1016/j.plabm.2025.e00459] [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: 09/25/2024] [Accepted: 02/26/2025] [Indexed: 04/17/2025] Open
Abstract
Cases of anemia presenting with abnormal erythrocyte morphology often pose diagnostic challenges, particularly in patients with refractory anemia. Here, we present the case of a 9-year-old male patient under investigation for anemia, who had a history of anemia and received a blood transfusion at birth. Despite the absence of obvious clinical manifestations related to anemia thereafter, his condition was not given due consideration. The patient experienced a sudden onset of illness and was initially suspected to have thalassemia. However, subsequent pertinent examinations, notably bone marrow aspiration and genetic testing, led to the diagnosis of hereditary sideroblastic anemia alongside chronic atrophic gastritis. This case illustrates the diagnostic journey of anemia characterized by abnormal red blood cell morphology, aiming to facilitate early and accurate diagnosis, as well as prompt treatment, for such patients in clinical practice.
Collapse
Affiliation(s)
- Huijun Qin
- Department of Clinical Laboratory, Sichuan Provincial People's Hospital, Chengdu, 610000, PR China
- Department of Clinical Laboratory, Dazhou Central Hospital, Dazhou, 635000, PR China
| | - Yuan He
- Department of Clinical Laboratory, Sichuan Provincial People's Hospital, Chengdu, 610000, PR China
| | - Zaixiang Xie
- Department of Clinical Laboratory, Qianjiang District Hospital of Traditional, Chinese Medicine, Chongqing, 409000, PR China
| |
Collapse
|
2
|
Jové-Solavera D, Rámila M, Ferrer-Cortés X, Olivella M, Venturi V, Morado M, Hernández-Rodríguez I, Khan A, Pérez-Montero S, Tornador C, Germing U, Gattermann N, Sanchez M. The role of genetic testing in accurate diagnosis of X-linked sideroblastic anemia: novel ALAS2 mutations and the impact of X-chromosome inactivation. Sci Rep 2025; 15:11843. [PMID: 40195342 PMCID: PMC11977005 DOI: 10.1038/s41598-025-95590-x] [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: 01/07/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025] Open
Abstract
X-linked sideroblastic anemia (XLSA) is a hereditary disorder affecting heme biosynthesis, caused by mutations in the ALAS2 gene, which encodes the erythroid-specific enzyme 5-aminolevulinate synthase. This enzyme, which requires pyridoxal 5'-phosphate (PLP) as a cofactor, catalyzes the first and rate-limiting step of heme synthesis in erythroid cells. XLSA is characterized by hypochromic microcytic anemia and ring sideroblasts in bone marrow, with most patients showing variable degrees of response to pyridoxine supplementation; however, female carriers of ALAS2 mutations often present a distinct clinical phenotype. A comprehensive review of the literature reveals over 100 distinct ALAS2 mutations linked to XLSA in more than 240 families. Here, we report seven new patients (four female cases) initially diagnosed with various conditions, later confirmed to have X-linked Sideroblastic Anemia due to ALAS2 mutations through genetic analysis. Among these, five represent novel ALAS2 mutations, including the first ever reported stop-loss mutation in ALAS2 associated with XLSA rather than X-linked dominant protoporphyria (XLDPP). Computational modelling of six reported cases revealed that four mutations significantly impact protein structure, conformation and cofactor interaction, consistent with our enzymatic assays demonstrating reduced ALAS2 activity. Furthermore, X-chromosome studies in female probands revealed a pronounced skewing of X-chromosome, which may provide an explanation for their distinct clinical manifestations in females.
Collapse
Affiliation(s)
- Daniel Jové-Solavera
- Iron Metabolism: Regulation and Diseases, Department of Biomedical Sciences, Universitat Internacional de Catalunya (UIC), 08195, Sant Cugat del Vallès, Spain
| | - Marta Rámila
- Iron Metabolism: Regulation and Diseases, Department of Biomedical Sciences, Universitat Internacional de Catalunya (UIC), 08195, Sant Cugat del Vallès, Spain
| | - Xènia Ferrer-Cortés
- Iron Metabolism: Regulation and Diseases, Department of Biomedical Sciences, Universitat Internacional de Catalunya (UIC), 08195, Sant Cugat del Vallès, Spain
- BloodGenetics S.L. Diagnostics in Inherited Blood Diseases, 08950, Esplugues de Llobregat, Spain
| | - Mireia Olivella
- Biosciences Department, Faculty of Sciences and Technology, University of Vic - Central University of Catalonia, Vic, Spain
| | - Veronica Venturi
- Iron Metabolism: Regulation and Diseases, Department of Biomedical Sciences, Universitat Internacional de Catalunya (UIC), 08195, Sant Cugat del Vallès, Spain
| | - Marta Morado
- Service of Hematology, Hospital La Paz, 28046, Madrid, Spain
| | - Ines Hernández-Rodríguez
- Hematology Department, ICO-Hospital Germans Trias i Pujol, Institut de Recerca Josep Carreras, Badalona, Barcelona, Spain
| | - Aneal Khan
- M.A.G.I.C. (Metabolics and Genetics in Canada), Calgary, AB, Canada
| | - Santiago Pérez-Montero
- BloodGenetics S.L. Diagnostics in Inherited Blood Diseases, 08950, Esplugues de Llobregat, Spain
| | - Cristian Tornador
- BloodGenetics S.L. Diagnostics in Inherited Blood Diseases, 08950, Esplugues de Llobregat, Spain
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Norbert Gattermann
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Mayka Sanchez
- Iron Metabolism: Regulation and Diseases, Department of Biomedical Sciences, Universitat Internacional de Catalunya (UIC), 08195, Sant Cugat del Vallès, Spain.
- BloodGenetics S.L. Diagnostics in Inherited Blood Diseases, 08950, Esplugues de Llobregat, Spain.
| |
Collapse
|
3
|
Dong Y, Zheng M, Ding W, Guan H, Xiao J, Li F. Nrf2 activators for the treatment of rare iron overload diseases: From bench to bedside. Redox Biol 2025; 81:103551. [PMID: 39965404 PMCID: PMC11876910 DOI: 10.1016/j.redox.2025.103551] [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: 01/08/2025] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
Iron overload and related oxidative damage are seen in many rare diseases, due to mutation of iron homeostasis-related genes. As a core regulator on cellular antioxidant reaction, Nrf2 can also decrease systemic and cellular iron levels by regulating iron-related genes and pathways, making Nrf2 activators very good candidates for the treatment of iron overload disorders. Successful examples include the clinical use of omaveloxolone for Friedreich's Ataxia and dimethyl fumarate for relapsing-remitting multiple sclerosis. Despite these uses, the therapeutic potentials of Nrf2 activators for iron overload disorders may be overlooked in clinical practice. Therefore, this study talks about the potential use, possible mechanisms, and precautions of Nrf2 activators in treating rare iron overload diseases. In addition, a combination therapy with Nrf2 activators and iron chelators is proposed for clinical reference, aiming to facilitate the clinical use of Nrf2 activators for more iron overload disorders.
Collapse
Affiliation(s)
- Yimin Dong
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Zheng
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weizhong Ding
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hanfeng Guan
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jun Xiao
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Feng Li
- Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Gradim M, Ferreira F. [Congenital Sideroblastic Anemia and Iron Overload in Older Age]. ACTA MEDICA PORT 2025; 38:279-280. [PMID: 40185132 DOI: 10.20344/amp.22803] [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: 12/31/2024] [Accepted: 02/10/2025] [Indexed: 04/07/2025]
Affiliation(s)
- Mariana Gradim
- Serviço de Imuno-Hemoterapia. Instituto Português de Oncologia do Porto Francisco Gentil. Porto. Portugal
| | - Fátima Ferreira
- Serviço de Hematologia Clínica. Unidade Local de Saúde de São João. Porto. Portugal
| |
Collapse
|
5
|
Rekaya S, Ben Fraj I, Hamdi R, Ben Taieb A, Merdassi A, Jouini H, Zarrouk H, Zaiter I, Kouki R, Bejaoui M, Mellouli F, Ben Khaled M, Ouederni M. Sideroblastic anemia in children: challenges in diagnosis and management in three cases. Ann Hematol 2025; 104:2537-2543. [PMID: 40042629 PMCID: PMC12052921 DOI: 10.1007/s00277-025-06266-5] [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: 08/15/2024] [Accepted: 02/15/2025] [Indexed: 05/06/2025]
Abstract
Sideroblastic anemias (SAs) represent a heterogeneous group of rare hematological disorders characterized by iron accumulation in mitochondria of erythroblasts with ineffective erythropoiesis. SAs are categorized into acquired and congenital forms. Acquired, secondary, and clonal, SA is rare in pediatric populations. Congenital SA (CSA) is classified into syndromic and non-syndromic forms. Herein, we describe three cases of pediatric patients with SA. The diagnosis of SA was based on the presence of type 3 sideroblasts on BM aspirate smear (greater than 15%) and genetic tests. In the first case, the diagnosis of myelodysplastic syndrome with ring sideroblasts (MDS-RS) with somatic SF3B1 mutation was made at the age of 11 years. A whole exome sequencing did not reveal any germinal predisposition for MDS. A wait-and-see strategy was adopted. After one year- of follow-up, no blood transfusion was needed and no further cytopenia occurred. The two other children had presented anemia at an early age and were diagnosed with CSA. The first case was a girl with SCL25A38 gene mutation. For the second one, the diagnosis of aminolevulinic acid synthase 2 deficiency was considered the most plausible given the family history and the favourable response to pyridoxine. Iron overload occurred in both patients with CSA, requiring chelation therapy. In conclusion, Perls' stain remains a valuable tool for guiding the diagnosis of unexplained anemia in pediatric patients. Genetic testing is crucial for the characterization of congenital sideroblastic anemias. The incidence of myeloid neoplasms with ring sideroblasts is exceptional in children, and the long-term prognosis remains undefined.
Collapse
Affiliation(s)
- Samia Rekaya
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia.
- Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia.
| | - Ilhem Ben Fraj
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
- Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia
| | - Rym Hamdi
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
| | - Aicha Ben Taieb
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
- Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia
| | - Amani Merdassi
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
| | - Hamida Jouini
- Laboratory of Hematology, Children Hospital Béchir Hamza, Tunis, Tunisia
| | - Hajer Zarrouk
- Laboratory of Hematology, Children Hospital Béchir Hamza, Tunis, Tunisia
| | - Ikram Zaiter
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
| | - Ridha Kouki
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
| | - Mohamed Bejaoui
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
- Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia
| | - Fethi Mellouli
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
- Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia
| | - Monia Ben Khaled
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
- Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia
| | - Monia Ouederni
- Department of Pediatrics: Immuno-Hematology and Stem Cell Transplantation, Bone Marrow Transplantation Center, Rue Djebel Lakhdhar, 1006, Tunis, Tunisia
- Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
6
|
Ammar M, Kmiha S, Maalej M, Felhi R, Kharrat M, Alila-Fersi O, Chouchen J, Maaloul I, Mkaouar-Rebai E, Kammoun T, Tlili A, Fakhfakh F. Identification of a novel truncated pathogenic variant in PUS1 gene in two siblings of consanguineous Tunisian family: intrafamilial phenotypic variability related to mtDNA copy number. Ann Hematol 2025; 104:943-952. [PMID: 39961824 PMCID: PMC11971136 DOI: 10.1007/s00277-025-06259-4] [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: 02/15/2024] [Accepted: 02/11/2025] [Indexed: 04/05/2025]
Abstract
Congenital sideroblastic anemia (CSA) is a rare genetic disorder caused by defects on heme biosynthesis and mitochondrial energy production. This disease is characterized by the presence of ring sideroblasts in the bone marrow caused by excessive iron accumulation in mitochondria of erythroblasts and by anemia of varying severity. In addition to its clinical variability, CSA is also characterized by genetic heterogeneity which required next-generation sequencing technologies to identify responsible gene. In the present study, whole-exome sequencing followed by Sanger sequencing were performed on a consanguineous family including two patients with congenital sideroblastic anemia. Mitochondrial DNA deletion and copy number were tested respectively by long PCR and QPCR. Subsequent bioinformatic investigations were performed using several programs. WES and Sanger sequencing results revealed a novel pathogenic variant c.579-580insT (p.N194X) in the PUS1 gene. Several bioinformatic tools supported that this variant was disease-causing. This variation leads to an incomplete catalytic site which will be probably non-functional and could disturb heme biosynthesis. In addition, no mtDNA deletion was detected in the two patients whereas mtDNA quantification revealed a decrease of mtDNA copy number in P2 and its increasing in P1. The increase in mtDNA copy number, which is most likely connected to a compensatory mechanism, may be the cause of the moderate phenotypic severity observed in P1 with the same p.N194X variant with P2. In conclusion, we identified a novel truncated pathogenic variant in PUS1 gene in two siblings of consanguineous family with intrafamilial phenotypic variability related to mtDNA copy number.
Collapse
Affiliation(s)
- Marwa Ammar
- Laboratory of Molecular and Functional Genetics, Faculty of Sciences of Sfax, University of Sfax, Route Soukra. Km 3., Sfax, Tunisia.
| | - Sana Kmiha
- Department of Pediatrics, CHU Hedi Chaker, Sfax, Tunisia
| | - Marwa Maalej
- Laboratory of Molecular and Functional Genetics, Faculty of Sciences of Sfax, University of Sfax, Route Soukra. Km 3., Sfax, Tunisia
| | - Rahma Felhi
- Laboratory of Molecular and Functional Genetics, Faculty of Sciences of Sfax, University of Sfax, Route Soukra. Km 3., Sfax, Tunisia
| | - Marwa Kharrat
- Laboratory of Molecular and Functional Genetics, Faculty of Sciences of Sfax, University of Sfax, Route Soukra. Km 3., Sfax, Tunisia
| | - Olfa Alila-Fersi
- Laboratory of Molecular and Functional Genetics, Faculty of Sciences of Sfax, University of Sfax, Route Soukra. Km 3., Sfax, Tunisia
| | - Jihen Chouchen
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ines Maaloul
- Department of Pediatrics, CHU Hedi Chaker, Sfax, Tunisia
| | - Emna Mkaouar-Rebai
- Laboratory of Molecular and Functional Genetics, Faculty of Sciences of Sfax, University of Sfax, Route Soukra. Km 3., Sfax, Tunisia
| | | | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Faiza Fakhfakh
- Laboratory of Molecular and Functional Genetics, Faculty of Sciences of Sfax, University of Sfax, Route Soukra. Km 3., Sfax, Tunisia.
| |
Collapse
|
7
|
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
|
8
|
Boucher AA, Dayton VJ, Pratt AR, Nassar NN, Elgammal Y, Kalfa TA. Three-generation female cohort with macrocytic anemia and iron overload. Am J Hematol 2025; 100:133-138. [PMID: 39329459 PMCID: PMC11625981 DOI: 10.1002/ajh.27489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Alexander A. Boucher
- Division of Pediatric Hematology/Oncology, Department of PediatricsUniversity of MinnesotaMinneapolisMinnesotaUSA
- Division of Hematology, Oncology, and Transplantation, Department of MedicineUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Vanessa J. Dayton
- Laboratory Medicine and Pathology, Hennepin County Medical CenterHennepin Healthcare Research InstituteMinneapolisMinnesotaUSA
| | - Annaliisa R. Pratt
- Laboratory Medicine and Pathology, Hennepin County Medical CenterHennepin Healthcare Research InstituteMinneapolisMinnesotaUSA
| | - Nicolas N. Nassar
- Cancer and Blood Diseases InstituteCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati Medical SchoolCincinnatiOhioUSA
| | - Yasmin Elgammal
- Cancer and Blood Diseases InstituteCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Theodosia A. Kalfa
- Cancer and Blood Diseases InstituteCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati Medical SchoolCincinnatiOhioUSA
| |
Collapse
|
9
|
Santoro A, De Santis S, Palmieri F, Vozza A, Agrimi G, Andolfo I, Russo R, Palazzo A, Storlazzi CT, Ferrucci A, Jun YW, Kool ET, Fiermonte G, Iolascon A, Paradies E, Marobbio CMT, Palmieri L. P2 Receptor Antagonists Rescue Defective Heme Content in an In Vitro SLC25A38-Associated Congenital Sideroblastic Anemia Cell Model. Int J Mol Sci 2024; 25:13314. [PMID: 39769087 PMCID: PMC11679772 DOI: 10.3390/ijms252413314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/30/2025] Open
Abstract
Mutations in the SLC25A38 gene are responsible for the second most common form of congenital sideroblastic anemia (CSA), a severe condition for which no effective treatment exists. We developed and characterized a K562 erythroleukemia cell line with markedly reduced expression of the SLC25A38 protein (A38-low cells). This model successfully recapitulated the main features of CSA, including reduced heme content and mitochondrial respiration, increase in mitochondrial iron, ROS levels and sensitivity to oxidative stress. Notably, our study uncovered a new role for extracellular pyridoxal 5'-phosphate (PLP) and other P2 receptor antagonists in rescuing the altered parameters of A38-low cells (for example, the heme content of the A38-low cells was increased from about 50% to about 80% by the P2 receptor antagonists treatment compared with the value of the controls). These findings suggest that targeting P2 receptors could represent a promising therapeutic approach for SLC25A38-associated CSA.
Collapse
Affiliation(s)
- Antonella Santoro
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Silvia De Santis
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Ferdinando Palmieri
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70126 Bari, Italy;
- Center of Excellence in Comparative Genomics, University of Bari Aldo Moro, 70125 Bari, Italy
| | - Angelo Vozza
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Gennaro Agrimi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Immacolata Andolfo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (I.A.); (R.R.); (A.I.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Roberta Russo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (I.A.); (R.R.); (A.I.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Antonio Palazzo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Arianna Ferrucci
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70126 Bari, Italy;
| | - Yong Woong Jun
- Department of Chemistry, Korea Advanced Institute Science and Technology (KAIST), Daejeon 34141, Republic of Korea;
| | - Eric T. Kool
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA;
- Sarafan ChEM-H Institute, Stanford University, Stanford, CA 94305, USA
| | - Giuseppe Fiermonte
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy; (I.A.); (R.R.); (A.I.)
- CEINGE-Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy
| | - Eleonora Paradies
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70126 Bari, Italy;
| | - Carlo Marya Thomas Marobbio
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
| | - Luigi Palmieri
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.S.); (S.D.S.); (F.P.); (A.V.); (G.A.); (A.P.); (C.T.S.); (G.F.); (L.P.)
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), 70126 Bari, Italy;
- Center of Excellence in Comparative Genomics, University of Bari Aldo Moro, 70125 Bari, Italy
| |
Collapse
|
10
|
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 PMCID: PMC11830978 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
|
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
|
Moutapam-Ngamby-Adriaansen Y, Maillot F, Labarthe F, Lioger B. Blood cytopenias as manifestations of inherited metabolic diseases: a narrative review. Orphanet J Rare Dis 2024; 19:65. [PMID: 38355710 PMCID: PMC10865644 DOI: 10.1186/s13023-024-03074-4] [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: 11/03/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Inherited Metabolic Diseases (IMD) encompass a diverse group of rare genetic conditions that, despite their individual rarity, collectively affect a substantial proportion, estimated at as much as 1 in 784 live births. Among their wide-ranging clinical manifestations, cytopenia stands out as a prominent feature. Consequently, IMD should be considered a potential diagnosis when evaluating patients presenting with cytopenia. However, it is essential to note that the existing scientific literature pertaining to the link between IMD and cytopenia is limited, primarily comprising case reports and case series. This paucity of data may contribute to the inadequate recognition of the association between IMD and cytopenia, potentially leading to underdiagnosis. In this review, we synthesize our findings from a literature analysis along with our clinical expertise to offer a comprehensive insight into the clinical presentation of IMD cases associated with cytopenia. Furthermore, we introduce a structured diagnostic approach underpinned by decision-making algorithms, with the aim of enhancing the early identification and management of IMD-related cytopenia.
Collapse
Affiliation(s)
- Yannick Moutapam-Ngamby-Adriaansen
- Service de Médecine Interne, CHRU de Tours, Tours Cedex 1, France.
- Service de Médecine Interne Et Polyvalente, 2, Centre Hospitalier de Blois, Mail Pierre Charlot, 41000, Blois, France.
| | - François Maillot
- Service de Médecine Interne, CHRU de Tours, Tours Cedex 1, France
- Reference Center for Inborn Errors of Metabolism ToTeM, CHRU de Tours, Hôpital Clocheville, 49 Bd Béranger, 37000, Tours, France
- INSERM U1253, iBrain, Université François Rabelais de Tours, 10 Boulevard Tonnellé, 37000, Tours, France
- INSERM U1069, Nutrition, Croissance et Cancer, Faculté de Médecine, Université François Rabelais de Tours, 10 Boulevard Tonnellé, 37000, Tours, France
| | - François Labarthe
- Reference Center for Inborn Errors of Metabolism ToTeM, CHRU de Tours, Hôpital Clocheville, 49 Bd Béranger, 37000, Tours, France
- INSERM U1069, Nutrition, Croissance et Cancer, Faculté de Médecine, Université François Rabelais de Tours, 10 Boulevard Tonnellé, 37000, Tours, France
- Service de Pédiatrie, CHRU de Tours, Tours Cedex 1, France
| | - Bertrand Lioger
- Service de Médecine Interne Et Polyvalente, 2, Centre Hospitalier de Blois, Mail Pierre Charlot, 41000, Blois, France
| |
Collapse
|
13
|
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
|
14
|
Toufiq M, Rinchai D, Bettacchioli E, Kabeer BSA, Khan T, Subba B, White O, Yurieva M, George J, Jourde-Chiche N, Chiche L, Palucka K, Chaussabel D. Harnessing large language models (LLMs) for candidate gene prioritization and selection. J Transl Med 2023; 21:728. [PMID: 37845713 PMCID: PMC10580627 DOI: 10.1186/s12967-023-04576-8] [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] [Received: 08/28/2023] [Accepted: 09/28/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Feature selection is a critical step for translating advances afforded by systems-scale molecular profiling into actionable clinical insights. While data-driven methods are commonly utilized for selecting candidate genes, knowledge-driven methods must contend with the challenge of efficiently sifting through extensive volumes of biomedical information. This work aimed to assess the utility of large language models (LLMs) for knowledge-driven gene prioritization and selection. METHODS In this proof of concept, we focused on 11 blood transcriptional modules associated with an Erythroid cells signature. We evaluated four leading LLMs across multiple tasks. Next, we established a workflow leveraging LLMs. The steps consisted of: (1) Selecting one of the 11 modules; (2) Identifying functional convergences among constituent genes using the LLMs; (3) Scoring candidate genes across six criteria capturing the gene's biological and clinical relevance; (4) Prioritizing candidate genes and summarizing justifications; (5) Fact-checking justifications and identifying supporting references; (6) Selecting a top candidate gene based on validated scoring justifications; and (7) Factoring in transcriptome profiling data to finalize the selection of the top candidate gene. RESULTS Of the four LLMs evaluated, OpenAI's GPT-4 and Anthropic's Claude demonstrated the best performance and were chosen for the implementation of the candidate gene prioritization and selection workflow. This workflow was run in parallel for each of the 11 erythroid cell modules by participants in a data mining workshop. Module M9.2 served as an illustrative use case. The 30 candidate genes forming this module were assessed, and the top five scoring genes were identified as BCL2L1, ALAS2, SLC4A1, CA1, and FECH. Researchers carefully fact-checked the summarized scoring justifications, after which the LLMs were prompted to select a top candidate based on this information. GPT-4 initially chose BCL2L1, while Claude selected ALAS2. When transcriptional profiling data from three reference datasets were provided for additional context, GPT-4 revised its initial choice to ALAS2, whereas Claude reaffirmed its original selection for this module. CONCLUSIONS Taken together, our findings highlight the ability of LLMs to prioritize candidate genes with minimal human intervention. This suggests the potential of this technology to boost productivity, especially for tasks that require leveraging extensive biomedical knowledge.
Collapse
Affiliation(s)
- Mohammed Toufiq
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Eleonore Bettacchioli
- INSERM UMR1227, Lymphocytes B et Autoimmunité, Université de Bretagne Occidentale, Brest, France
- Service de Rhumatologie, CHU de Brest, Brest, France
| | | | - Taushif Khan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Bishesh Subba
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Olivia White
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Laurent Chiche
- Service de Médecine Interne, Hôpital Européen, Marseille, France
| | - Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | |
Collapse
|
15
|
Tam PCK, Daniel S, Lim J, Beligaswatte A. Pristinamycin-induced sideroblastic anaemia. J Antimicrob Chemother 2023:dkad180. [PMID: 37311214 DOI: 10.1093/jac/dkad180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023] Open
Affiliation(s)
- Patrick C K Tam
- Department of Infectious Diseases, Flinders Medical Centre, Adelaide, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Santhosh Daniel
- Department of Infectious Diseases, Flinders Medical Centre, Adelaide, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Jonathan Lim
- Department of Haematology, Flinders Medical Centre, Adelaide, SA, Australia
| | - Ashanka Beligaswatte
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
- Department of Haematology, Flinders Medical Centre, Adelaide, SA, Australia
| |
Collapse
|
16
|
Salam MAE, Salama K, Selim YMM, Saad M, Rady R, Alawbathani S, Schroeder S, Elmonem MA, Elkhateeb N. Three siblings with variable degrees of neuromuscular involvement and congenital sideroblastic anemia: A peculiar phenotype and a surprise genotypic explanation. Ann Hum Genet 2023. [PMID: 36916508 DOI: 10.1111/ahg.12505] [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: 10/07/2022] [Revised: 02/22/2023] [Accepted: 02/26/2023] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Congenital sideroblastic anemias (CSAs) are a group of inherited bone-marrow disorders manifesting with erythroid hyperplasia and ineffective erythropoiesis. METHODS We describe a detailed clinical and genetic characterization of three siblings with CSA. RESULTS Two of them had limb-girdle myopathy and global developmental delay. The two elder siblings performed allogenic hematopoietic stem-cell transplantation 5 and 3 years prior with stabilization of the hematological features. Exome sequencing in the non-transplanted sibling revealed a novel homozygous nonsense variant in SLC25A38 gene NM_017875.2:c.559C > T; p.(Arg187*) causing autosomal-recessive sideroblastic anemia type-2, and a second homozygous pathogenic previously reported variant in GMPPB gene NM_013334.3:c.458C > T; p.(Thr153Ile) causing autosomal-recessive muscular dystrophy-dystroglycanopathy type B14. With the established diagnosis, hematopoietic stem cell transplantation is now being scheduled for the youngest sibling, and a trial therapy with acetylcholine esterase inhibitors was started for the two neurologically affected patients with partial clinical improvement. CONCLUSION This family emphasizes the importance of whole-exome sequencing for familial cases with complex phenotypes and vague neurological manifestations.
Collapse
Affiliation(s)
- Mai Abd El Salam
- Department of Pediatrics (Pediatric Hematology Unit), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Khaled Salama
- Department of Pediatrics (Pediatric Hematology Unit), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Yasmeen M M Selim
- Department of Pediatrics (Pediatric Hematology Unit), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mariam Saad
- Department of Pediatrics (Pediatric Hematology Unit), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rasha Rady
- Department of Pediatrics (Pediatric Hematology Unit), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | - Mohamed A Elmonem
- Department of Clinical and Chemical Pathology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.,Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Nour Elkhateeb
- Department of Pediatrics (Pediatric Neurology and Metabolic Medicine Unit), Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.,Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
17
|
Scaramellini N, Fischer D, Agarvas AR, Motta I, Muckenthaler MU, Mertens C. Interpreting Iron Homeostasis in Congenital and Acquired Disorders. Pharmaceuticals (Basel) 2023; 16:ph16030329. [PMID: 36986429 PMCID: PMC10054723 DOI: 10.3390/ph16030329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Mammalian cells require iron to satisfy their metabolic needs and to accomplish specialized functions, such as hematopoiesis, mitochondrial biogenesis, energy metabolism, or oxygen transport. Iron homeostasis is balanced by the interplay of proteins responsible for iron import, storage, and export. A misbalance of iron homeostasis may cause either iron deficiencies or iron overload diseases. The clinical work-up of iron dysregulation is highly important, as severe symptoms and pathologies may arise. Treating iron overload or iron deficiency is important to avoid cellular damage and severe symptoms and improve patient outcomes. The impressive progress made in the past years in understanding mechanisms that maintain iron homeostasis has already changed clinical practice for treating iron-related diseases and is expected to improve patient management even further in the future.
Collapse
Affiliation(s)
- Natalia Scaramellini
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milano, Italy
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Dania Fischer
- Department of Anesthesiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Anand R. Agarvas
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Immunology, and Hematology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany
| | - Irene Motta
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milano, Italy
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Martina U. Muckenthaler
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Immunology, and Hematology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Side, 69120 Heidelberg, Germany
| | - Christina Mertens
- Center for Translational Biomedical Iron Research, Department of Pediatric Oncology, Immunology, and Hematology, University of Heidelberg, INF 350, 69120 Heidelberg, Germany
- Molecular Medicine Partnership Unit, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221564582; Fax: +49-6221564580
| |
Collapse
|
18
|
Chen PH, Gorshein E, Tormey C, Siddon AJ, Perincheri S. "Double-hit" ineffective erythropoiesis-concurrent β-thalassemia with α-gene triplication and myelodysplastic syndrome with SF3B1 mutation. Am J Hematol 2023; 98:984-988. [PMID: 36688500 DOI: 10.1002/ajh.26856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023]
Affiliation(s)
- Po-Han Chen
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Elan Gorshein
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christopher Tormey
- Department of Laboratory Medicine, Yale New Haven Hospital, New Haven, Connecticut, USA
| | - Alexa J Siddon
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Laboratory Medicine, Yale New Haven Hospital, New Haven, Connecticut, USA
| | - Sudhir Perincheri
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
19
|
Mohamed S, Ibrahim F, Alasafar MN, Alshurafa A, Akiki S, Soliman D, Kohla S, Amer A, Qasim H, Cherif H. Recurrent sideroblastic anemia during pregnancy. Clin Case Rep 2023; 11:e6814. [PMID: 36644616 PMCID: PMC9834147 DOI: 10.1002/ccr3.6814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 11/01/2022] [Accepted: 12/14/2022] [Indexed: 01/15/2023] Open
Abstract
Sideroblastic anemia is a heterogeneous group of disorders typified by the presence of ring sideroblasts in the bone marrow and has congenital and acquired types. Sideroblastic anemia is a rare event in pregnancy. We report a case of a 32-year-old female patient, gravida 4 para 3, 27th weeks pregnant, who presented to the emergency department complaining of palpitation and generalized weakness for 2 weeks. She was found to have severe normochromic normocytic anemia, with hemoglobin of 4.2 g/dl, and low reticulocytes count of 13 × 103/μl. She gave a history of recurrent anemia, which had only occurred during pregnancy. Her bone marrow aspirate showed many ring sideroblasts concluding the diagnosis of sideroblastic anemia (SA). Further investigation revealed a significantly low pyridoxine level (vitamin B6) of (8 nmol/L). The Hb level improved with vitamin B6 replacement, without any transfusion support.
Collapse
Affiliation(s)
- Shehab Mohamed
- Department of HematologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| | - Firyal Ibrahim
- Department of Laboratory Medicine and PathologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| | | | - Awni Alshurafa
- Department of HematologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| | - Susanna Akiki
- Department of Laboratory Medicine and PathologyHamad Medical CorporationDohaQatar
- Weill Cornell Medicine‐QatarDohaQatar
| | - Dina Soliman
- Department of Laboratory Medicine and PathologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| | - Samah Kohla
- Department of Laboratory Medicine and PathologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| | - Aliaa Amer
- Department of Laboratory Medicine and PathologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| | - Hana Qasim
- Department of HematologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| | - Honar Cherif
- Department of HematologyNational Center for Cancer Care and Research, Hamad Medical CorporationDohaQatar
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
Brasil S, Allocca M, Magrinho SCM, Santos I, Raposo M, Francisco R, Pascoal C, Martins T, Videira PA, Pereira F, Andreotti G, Jaeken J, Kantautas KA, Perlstein EO, Ferreira VDR. Systematic Review: Drug Repositioning for Congenital Disorders of Glycosylation (CDG). Int J Mol Sci 2022; 23:8725. [PMID: 35955863 PMCID: PMC9369176 DOI: 10.3390/ijms23158725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/02/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
Advances in research have boosted therapy development for congenital disorders of glycosylation (CDG), a group of rare genetic disorders affecting protein and lipid glycosylation and glycosylphosphatidylinositol anchor biosynthesis. The (re)use of known drugs for novel medical purposes, known as drug repositioning, is growing for both common and rare disorders. The latest innovation concerns the rational search for repositioned molecules which also benefits from artificial intelligence (AI). Compared to traditional methods, drug repositioning accelerates the overall drug discovery process while saving costs. This is particularly valuable for rare diseases. AI tools have proven their worth in diagnosis, in disease classification and characterization, and ultimately in therapy discovery in rare diseases. The availability of biomarkers and reliable disease models is critical for research and development of new drugs, especially for rare and heterogeneous diseases such as CDG. This work reviews the literature related to repositioned drugs for CDG, discovered by serendipity or through a systemic approach. Recent advances in biomarkers and disease models are also outlined as well as stakeholders' views on AI for therapy discovery in CDG.
Collapse
Affiliation(s)
- Sandra Brasil
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Mariateresa Allocca
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Institute of Biomolecular Chemistry, National Research Council of Italy, 80078 Pozzuoli, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | - Salvador C. M. Magrinho
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- LAQV-Requimte, Chemistry Department, School of Science and Technology, Nova University of Lisbon, 2819-516 Caparica, Portugal
| | - Inês Santos
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Sci and Volunteer Program from School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Madalena Raposo
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Sci and Volunteer Program from School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Rita Francisco
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Carlota Pascoal
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Tiago Martins
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Sci and Volunteer Program from School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Paula A. Videira
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| | - Florbela Pereira
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- LAQV-Requimte, Chemistry Department, School of Science and Technology, Nova University of Lisbon, 2819-516 Caparica, Portugal
| | - Giuseppina Andreotti
- Institute of Biomolecular Chemistry, National Research Council of Italy, 80078 Pozzuoli, Italy
| | - Jaak Jaeken
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Center for Metabolic Diseases, Department of Pediatrics, KU Leuven, 3000 Leuven, Belgium
| | | | | | - Vanessa dos Reis Ferreira
- UCIBIO—Applied Molecular Biosciences Unit, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Nova University of Lisbon, 2829-516 Caparica, Portugal
- CDG & Allies PPAIN—Professionals and Patient Associations International Network, Department of Life Sciences, School of Science and Technology, NOVA University of Lisbon, 2819-516 Caparica, Portugal
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Taylor JL, Brown BL. Structural basis for dysregulation of aminolevulinic acid synthase in human disease. J Biol Chem 2022; 298:101643. [PMID: 35093382 PMCID: PMC8892079 DOI: 10.1016/j.jbc.2022.101643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/19/2023] Open
Abstract
Heme is a critical biomolecule that is synthesized in vivo by several organisms such as plants, animals, and bacteria. Reflecting the importance of this molecule, defects in heme biosynthesis underlie several blood disorders in humans. Aminolevulinic acid synthase (ALAS) initiates heme biosynthesis in α-proteobacteria and nonplant eukaryotes. Debilitating and painful diseases such as X-linked sideroblastic anemia and X-linked protoporphyria can result from one of more than 91 genetic mutations in the human erythroid-specific enzyme ALAS2. This review will focus on recent structure-based insights into human ALAS2 function in health and how it dysfunctions in disease. We will also discuss how certain genetic mutations potentially result in disease-causing structural perturbations. Furthermore, we use thermodynamic and structural information to hypothesize how the mutations affect the human ALAS2 structure and categorize some of the unique human ALAS2 mutations that do not respond to typical treatments, that have paradoxical in vitro activity, or that are highly intolerable to changes. Finally, we will examine where future structure-based insights into the family of ALA synthases are needed to develop additional enzyme therapeutics.
Collapse
Affiliation(s)
- Jessica L Taylor
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Breann L Brown
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
27
|
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: 8] [Impact Index Per Article: 2.7] [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
|
28
|
Oakley JH, Campagna DR, Sun L, Rockowitz S, Sliz P, Boudreaux J, Woods G, Fleming MD. A synonymous coding variant that alters ALAS2 splicing and causes X-linked sideroblastic anemia. Pediatr Blood Cancer 2022; 69:e29309. [PMID: 34411431 DOI: 10.1002/pbc.29309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 11/08/2022]
Affiliation(s)
- Jamie Heather Oakley
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Dean R Campagna
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Liang Sun
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Shira Rockowitz
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Piotr Sliz
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jeanne Boudreaux
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Gary Woods
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
29
|
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
|
30
|
Marku A, Galli A, Marciani P, Dule N, Perego C, Castagna M. Iron Metabolism in Pancreatic Beta-Cell Function and Dysfunction. Cells 2021; 10:2841. [PMID: 34831062 PMCID: PMC8616520 DOI: 10.3390/cells10112841] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/26/2022] Open
Abstract
Iron is an essential element involved in a variety of physiological functions. In the pancreatic beta-cells, being part of Fe-S cluster proteins, it is necessary for the correct insulin synthesis and processing. In the mitochondria, as a component of the respiratory chain, it allows the production of ATP and reactive oxygen species (ROS) that trigger beta-cell depolarization and potentiate the calcium-dependent insulin release. Iron cellular content must be finely tuned to ensure the normal supply but also to prevent overloading. Indeed, due to the high reactivity with oxygen and the formation of free radicals, iron excess may cause oxidative damage of cells that are extremely vulnerable to this condition because the normal elevated ROS production and the paucity in antioxidant enzyme activities. The aim of the present review is to provide insights into the mechanisms responsible for iron homeostasis in beta-cells, describing how alteration of these processes has been related to beta-cell damage and failure. Defects in iron-storing or -chaperoning proteins have been detected in diabetic conditions; therefore, the control of iron metabolism in these cells deserves further investigation as a promising target for the development of new disease treatments.
Collapse
Affiliation(s)
| | | | | | | | - Carla Perego
- Department of Excellence Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Trentacoste, 22134 Milano, Italy; (A.M.); (A.G.); (P.M.); (N.D.)
| | - Michela Castagna
- Department of Excellence Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Trentacoste, 22134 Milano, Italy; (A.M.); (A.G.); (P.M.); (N.D.)
| |
Collapse
|
31
|
Heeney MM, Berhe S, Campagna DR, Oved JH, Kurre P, Shaw PJ, Teo J, Shanap MA, Hassab HM, Glader BE, Shah S, Yoshimi A, Ameri A, Antin JH, Boudreaux J, Briones M, Dickerson KE, Fernandez CV, Farah R, Hasle H, Keel SB, Olson TS, Powers JM, Rose MJ, Shimamura A, Bottomley SS, Fleming MD. SLC25A38 congenital sideroblastic anemia: Phenotypes and genotypes of 31 individuals from 24 families, including 11 novel mutations, and a review of the literature. Hum Mutat 2021; 42:1367-1383. [PMID: 34298585 DOI: 10.1002/humu.24267] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/10/2021] [Accepted: 07/21/2021] [Indexed: 01/19/2023]
Abstract
The congenital sideroblastic anemias (CSAs) are a heterogeneous group of inherited disorders of erythropoiesis characterized by pathologic deposits of iron in the mitochondria of developing erythroblasts. Mutations in the mitochondrial glycine carrier SLC25A38 cause the most common recessive form of CSA. Nonetheless, the disease is still rare, there being fewer than 70 reported families. Here we describe the clinical phenotype and genotypes of 31 individuals from 24 families, including 11 novel mutations. We also review the spectrum of reported mutations and genotypes associated with the disease, describe the unique localization of missense mutations in transmembrane domains and account for the presence of several alleles in different populations.
Collapse
Affiliation(s)
- Matthew M Heeney
- Division of Hematology, Dana-Farber Boston Children's Cancer and Blood Disorders Center and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Simon Berhe
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dean R Campagna
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph H Oved
- Cellular Therapy and Transplant Section, Division of Oncology and Comprehensive Bone Marrow Failure Center, Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Philadelphia, USA
| | - Peter Kurre
- Pediatric Comprehensive Bone Marrow Failure Center, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Peter J Shaw
- BMT Services, Children's Hospital at Westmead; Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Juliana Teo
- Department of Haematology, Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | | | - Hoda M Hassab
- Department of Paediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Bertil E Glader
- Division of Hematology-Oncology, Lucille Packard Children's Hospital, Stanford, California, USA
| | - Sanjay Shah
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Ayami Yoshimi
- Department of Paediatrics and Adolescent Medicine, Division of Paediatric Haematology and Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Afshin Ameri
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Augusta University, Augusta, Georgia, USA
| | - Joseph H Antin
- Hematopoietic Stem Cell Transplantation Program, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, Massachusetts, USA
| | - Jeanne Boudreaux
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Michael Briones
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Kathryn E Dickerson
- Department of Pediatrics, University of Texas Southwestern, Dallas, Texas, USA
| | - Conrad V Fernandez
- Division of Hematology-Oncology, IWH Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Roula Farah
- Department of Pediatrics, Lebanese American University Medical Center, Beirut, Lebanon
| | - Henrik Hasle
- Department of Pediatrics, Aarhus University Hospital, Aarhus University, Aarhus, Denmark
| | - Sioban B Keel
- Division of Hematology, University of Washington and Seattle Cancer Care Alliance, Seattle, Washington, USA
| | - Timothy S Olson
- Cellular Therapy and Transplant Section, Division of Oncology and Comprehensive Bone Marrow Failure Center, Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jacquelyn M Powers
- Texas Children's Hospital and Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Melissa J Rose
- Division of Hematology & Oncology, Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Akiko Shimamura
- Division of Hematology, Dana-Farber Boston Children's Cancer and Blood Disorders Center and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Sylvia S Bottomley
- Hematology-Oncology Section, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Han X, Wen Q, Liu X, Wan K, Yan HJ, Zhang C, Zhang X. [New mutation of congenital sideroblastic anemia: a case report and literature review]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2021; 42:603-605. [PMID: 34455750 PMCID: PMC8408491 DOI: 10.3760/cma.j.issn.0253-2727.2021.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Indexed: 11/05/2022]
Affiliation(s)
- X Han
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, PLA Blood Disease Center, Chongqing Key Discipline of Medicine, Chongqing 400037, China
| | - Q Wen
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, PLA Blood Disease Center, Chongqing Key Discipline of Medicine, Chongqing 400037, China
| | - X Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, PLA Blood Disease Center, Chongqing Key Discipline of Medicine, Chongqing 400037, China
| | - K Wan
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, PLA Blood Disease Center, Chongqing Key Discipline of Medicine, Chongqing 400037, China
| | - H J Yan
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, PLA Blood Disease Center, Chongqing Key Discipline of Medicine, Chongqing 400037, China
| | - C Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, PLA Blood Disease Center, Chongqing Key Discipline of Medicine, Chongqing 400037, China
| | - X Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, PLA Blood Disease Center, Chongqing Key Discipline of Medicine, Chongqing 400037, China
| |
Collapse
|
33
|
Steinberg-Shemer O, Tamary H. Impact of Next-Generation Sequencing on the Diagnosis and Treatment of Congenital Anemias. Mol Diagn Ther 2021; 24:397-407. [PMID: 32557003 DOI: 10.1007/s40291-020-00478-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Congenital anemias are a wide spectrum of diseases including hypoproliferative anemia syndromes, dyserythropoietic anemias, sideroblastic anemias, red blood cell membrane and enzymatic defects, hemoglobinopathies, and thalassemia syndromes. The various congenital anemia syndromes may have similar clinical and laboratory presentations, making the diagnosis challenging. The traditional work-up, which includes a complete blood count, blood smears, bone marrow studies, flow cytometry, and the osmotic fragility test, does not always lead to the diagnosis. Specialized tests such as red blood cell enzyme activity and ektacytometry are not widely available. In addition, red blood cell transfusions may mask some of the laboratory characteristics. Therefore, genetic testing is crucial for accurate diagnosis of patients with congenital anemias. However, gene-by-gene testing is labor intensive because of the large number of genes involved. Thus, targeted next-generation sequencing using custom-made gene panels has been increasingly utilized, with a high success rate of diagnosis. Accurate genetic diagnosis is important for determining specific therapeutic modalities, as well as for avoiding splenectomy when contraindicated. In addition, molecular diagnosis can allow for genetic counseling and prenatal diagnosis in severe cases. We suggest a work-up scheme for patients with congenital anemias, including early incorporation of targeted next-generation sequencing panels.
Collapse
Affiliation(s)
- Orna Steinberg-Shemer
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Hematology Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Hannah Tamary
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Pediatric Hematology Laboratory, Felsenstein Medical Research Center, Petach Tikva, Israel.
| |
Collapse
|
34
|
Maio N, Zhang DL, Ghosh MC, Jain A, SantaMaria AM, Rouault TA. Mechanisms of cellular iron sensing, regulation of erythropoiesis and mitochondrial iron utilization. Semin Hematol 2021; 58:161-174. [PMID: 34389108 DOI: 10.1053/j.seminhematol.2021.06.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022]
Abstract
To maintain an adequate iron supply for hemoglobin synthesis and essential metabolic functions while counteracting iron toxicity, humans and other vertebrates have evolved effective mechanisms to conserve and finely regulate iron concentration, storage, and distribution to tissues. At the systemic level, the iron-regulatory hormone hepcidin is secreted by the liver in response to serum iron levels and inflammation. Hepcidin regulates the expression of the sole known mammalian iron exporter, ferroportin, to control dietary absorption, storage and tissue distribution of iron. At the cellular level, iron regulatory proteins 1 and 2 (IRP1 and IRP2) register cytosolic iron concentrations and post-transcriptionally regulate the expression of iron metabolism genes to optimize iron availability for essential cellular processes, including heme biosynthesis and iron-sulfur cluster biogenesis. Genetic malfunctions affecting the iron sensing mechanisms or the main pathways that utilize iron in the cell cause a broad range of human diseases, some of which are characterized by mitochondrial iron accumulation. This review will discuss the mechanisms of systemic and cellular iron sensing with a focus on the main iron utilization pathways in the cell, and on human conditions that arise from compromised function of the regulatory axes that control iron homeostasis.
Collapse
Affiliation(s)
- Nunziata Maio
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - De-Liang Zhang
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Manik C Ghosh
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Anshika Jain
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Anna M SantaMaria
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Tracey A Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
35
|
Sideroblastic Anemia Associated With Isoniazid Prophylaxis in a Person Living With HIV. Am J Ther 2021; 27:e409-e410. [PMID: 31833877 DOI: 10.1097/mjt.0000000000000962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Ryan P, Kinirons P. Nutritional neuropathy postoesophagogastrectomy. BMJ Case Rep 2021; 14:e242716. [PMID: 34039553 PMCID: PMC8160179 DOI: 10.1136/bcr-2021-242716] [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] [Accepted: 05/10/2021] [Indexed: 11/03/2022] Open
Abstract
We report a case of a patient who presented complaining of a 1-week history of progressive lower limb weakness and decreased sensation bilaterally suggestive of a peripheral neuropathy, with vague associated symptoms of fluctuating concentration. Clinically, we suspected a Guillain-Barré variant. However, her functioning continued to decline despite intravenous immunoglobulin therapy, and she had normal spinal imaging studies and CSF analysis. Of note, she had a subtotal oesophagectomy and proximal gastrectomy 20 months previously for oesophageal cancer. We found her to be deficient in vitamin D, vitamin E and copper. She was treated with nutritional supplementation of these vitamins and infusion of trace elements, resulting in a gradual improvement in lower limb power, sensation and coordination, as well as improved cognition and mentation. Monthly outpatient neurology follow-up shows continued improvement in symptoms and return towards baseline functioning with regular infusions of nutritional elements and monitoring of blood levels.
Collapse
Affiliation(s)
- Paul Ryan
- Anaesthesia/Intensive Care, Tallaght University Hospital, Dublin, Ireland
| | - Peter Kinirons
- Neurology, Bon Secours Hospital Hospital Cork, Cork, Munster, Ireland
| |
Collapse
|
37
|
Juchniewicz P, Piotrowska E, Kloska A, Podlacha M, Mantej J, Węgrzyn G, Tukaj S, Jakóbkiewicz-Banecka J. Dosage Compensation in Females with X-Linked Metabolic Disorders. Int J Mol Sci 2021; 22:ijms22094514. [PMID: 33925963 PMCID: PMC8123450 DOI: 10.3390/ijms22094514] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 01/19/2023] Open
Abstract
Through the use of new genomic and metabolomic technologies, our comprehension of the molecular and biochemical etiologies of genetic disorders is rapidly expanding, and so are insights into their varying phenotypes. Dosage compensation (lyonization) is an epigenetic mechanism that balances the expression of genes on heteromorphic sex chromosomes. Many studies in the literature have suggested a profound influence of this phenomenon on the manifestation of X-linked disorders in females. In this review, we summarize the clinical and genetic findings in female heterozygotic carriers of a pathogenic variant in one of ten selected X-linked genes whose defects result in metabolic disorders.
Collapse
Affiliation(s)
- Patrycja Juchniewicz
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (P.J.); (A.K.); (J.J.-B.)
| | - Ewa Piotrowska
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.P.); (J.M.); (G.W.); (S.T.)
- Correspondence: ; Tel.: +48-58-523-6040
| | - Anna Kloska
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (P.J.); (A.K.); (J.J.-B.)
| | - Magdalena Podlacha
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.P.); (J.M.); (G.W.); (S.T.)
| | - Jagoda Mantej
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.P.); (J.M.); (G.W.); (S.T.)
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.P.); (J.M.); (G.W.); (S.T.)
| | - Stefan Tukaj
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (M.P.); (J.M.); (G.W.); (S.T.)
| | - Joanna Jakóbkiewicz-Banecka
- Department of Medical Biology and Genetics, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdańsk, Poland; (P.J.); (A.K.); (J.J.-B.)
| |
Collapse
|
38
|
Huang J, Ge M, Shao Y, Wang M, Jin P, Huo J, Li X, Zhang J, Nie N, Zheng Y. A hemizygous p.R204Q mutation in the ALAS2 gene underlies X-linked sideroblastic anemia in an adult Chinese Han man. BMC Med Genomics 2021; 14:107. [PMID: 33858445 PMCID: PMC8048311 DOI: 10.1186/s12920-021-00950-x] [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: 11/11/2020] [Accepted: 03/23/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND X-linked sideroblastic anemia (XLSA) is the most common form of congenital sideroblastic anemia (CSA), and is associated with the mutations in the 5-aminolevulinate synthase 2 (ALAS2). The genetic basis of more than 40% of CSA cases remains unknown. METHODS A two-generation Chinese family with XLSA was studied by next-generation sequencing to identify the underlying CSA-related mutations. RESULTS In the study, we identified a missense ALAS2 R204Q mutation in a hemizygous Chinese Han man and in his heterozygous daughter. The male proband presented clinical manifestations at 38 years old and had a good response to pyridoxine. CONCLUSIONS XLSA, as a hereditary disease, can present clinical manifestations later in lives, for adult male patients with ringed sideroblasts and hypochromic anemia, it should be evaluated with gene analyses to exclude CSA.
Collapse
Affiliation(s)
- Jinbo Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Meili Ge
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China.
| | - Yingqi Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Min Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Peng Jin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Jiali Huo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Xingxin Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Neng Nie
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| | - Yizhou Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 288 Nanjing Road, Tianjin, 300020, People's Republic of China
| |
Collapse
|
39
|
Uminski K, Houston DS, Hartley JN, Liu J, Cuvelier GDE, Israels SJ. Clinical characterization and hematopoietic stem cell transplant outcomes for congenital sideroblastic anemia caused by a novel pathogenic variant in SLC25A38. Pediatr Blood Cancer 2020; 67:e28623. [PMID: 32790119 DOI: 10.1002/pbc.28623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 01/19/2023]
Abstract
BACKGROUND Congenital sideroblastic anemia (CSA) constitutes an uncommon category of inherited anemia often associated with pathologic iron accumulation. Pathogenic variants in several genes have been identified as causative for CSA. Autosomal recessive pathogenic variants in the mitochondrial glycine transporter SLC25A38 have been implicated in a subset of patients with CSA. PROCEDURE We describe seven individuals of Canadian Cree descent with a known or inferred homozygous novel founder missense variant in SLC25A38 (c.560G>A, p.Arg187Gln). RESULTS All individuals presented as young children (median age 6 months) with severe microcytic, hypochromic anemia associated with pretransfusion iron overload, requiring red cell transfusion support and iron chelation. Six individuals received pyridoxine supplementation; two demonstrating transient partial responses. Three individuals underwent allogeneic hematopoietic stem cell transplantation (HSCT). One individual with significant iron loading died in the posttransplant period due to complications of sepsis. The other two individuals remain transfusion-free following HSCT. CONCLUSIONS Despite a common genetic etiology, phenotypic variability was noted in this cohort. A transient response to pyridoxine was noted in two individuals but should not be considered a long-term therapeutic strategy. HSCT was curative when performed before significant iron loading occurred. Early identification of CSA and timely HSCT can result in excellent long-term outcomes.
Collapse
Affiliation(s)
- Kelsey Uminski
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Donald S Houston
- Department of Internal Medicine, Section of Hematology and Medical Oncology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jessica N Hartley
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jing Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Geoffrey D E Cuvelier
- Departments of Pediatric Hematology-Oncology, Cancer Care Manitoba, and Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sara J Israels
- Departments of Pediatric Hematology-Oncology, Cancer Care Manitoba, and Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
40
|
Abu-Zeinah G, DeSancho MT. Understanding Sideroblastic Anemia: An Overview of Genetics, Epidemiology, Pathophysiology and Current Therapeutic Options. J Blood Med 2020; 11:305-318. [PMID: 33061728 PMCID: PMC7524202 DOI: 10.2147/jbm.s232644] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/18/2020] [Indexed: 01/19/2023] Open
Abstract
Sideroblastic anemia (SA) consists of a group of inherited and acquired anemias of ineffective erythropoiesis characterized by the accumulation of ring sideroblasts in the bone marrow due to disrupted heme biosynthesis. Congenital sideroblastic anemia (CSA) is rare and has three modes of inheritance: X-linked (XLSA), autosomal recessive (ARCSA), and maternal. Acquired SA is more common and can be a result of myelodysplastic syndromes (MDS) or other, generally reversible causes. The diagnostic approach to SA includes a work-up for reversible causes and genetic testing for CSA based on clinical suspicion, family history and genetic pedigree. The treatment of SA depends on the underlying etiology but remains primarily supportive with vitamin B6 supplementation for select cases of XLSA, thiamine for thiamine-responsive megaloblastic anemia subtype, red blood cell transfusions for symptomatic patients and iron chelation therapy for iron overload. The management of anemia in MDS subtypes with ring sideroblasts remains unique and includes the recently approved erythroid maturation agent, Luspatercept. Although there is currently no curative therapy for CSA, anecdotal reports of hematopoietic stem cell transplant demonstrate remissions in selective, non-syndromic cases. This review summarizes the genetics, pathophysiology, diagnosis and treatment of SA for general practitioners and clinical hematologists.
Collapse
Affiliation(s)
- Ghaith Abu-Zeinah
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, USA
| | - Maria T DeSancho
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, USA
| |
Collapse
|
41
|
Chiabrando D, Bertino F, Tolosano E. Hereditary Ataxia: A Focus on Heme Metabolism and Fe-S Cluster Biogenesis. Int J Mol Sci 2020; 21:ijms21113760. [PMID: 32466579 PMCID: PMC7312568 DOI: 10.3390/ijms21113760] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Heme and Fe-S clusters regulate a plethora of essential biological processes ranging from cellular respiration and cell metabolism to the maintenance of genome integrity. Mutations in genes involved in heme metabolism and Fe-S cluster biogenesis cause different forms of ataxia, like posterior column ataxia and retinitis pigmentosa (PCARP), Friedreich's ataxia (FRDA) and X-linked sideroblastic anemia with ataxia (XLSA/A). Despite great efforts in the elucidation of the molecular pathogenesis of these disorders several important questions still remain to be addressed. Starting with an overview of the biology of heme metabolism and Fe-S cluster biogenesis, the review discusses recent progress in the understanding of the molecular pathogenesis of PCARP, FRDA and XLSA/A, and highlights future line of research in the field. A better comprehension of the mechanisms leading to the degeneration of neural circuity responsible for balance and coordinated movement will be crucial for the therapeutic management of these patients.
Collapse
|
42
|
La P, Oved JH, Ghiaccio V, Rivella S. Mitochondria Biogenesis Modulates Iron-Sulfur Cluster Synthesis to Increase Cellular Iron Uptake. DNA Cell Biol 2020; 39:756-765. [PMID: 32282232 DOI: 10.1089/dna.2019.5123] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Iron-sulfur (Fe-S) clusters are required for mitochondrial function. Fe-S cluster synthesis occurs in the mitochondria and iron uptake is required for mitochondrial biogenesis. However, Fe-S clusters inhibit the expression of the iron importer transferrin receptor 1 (TfR1), whereas lack of the Fe-S cluster stimulates TfR1 expression. Yet, it is unclear whether Fe-S cluster synthesis increases with mitochondria biogenesis and, in turn, whether this negatively modulates TfR1 expression. We manipulated peroxisome proliferator-activated receptor-gamma coactivator-1α expression to control mitochondrial biogenesis in a variety of cell types, including erythroid cells. We demonstrated that Fe-S cluster synthesis increases with mitochondria biogenesis but does not interfere with increasing TfR1 expression. In fact, TfR1 expression is stimulated through alternative means to meet iron requirement for mitochondria biogenesis. Furthermore, under enhanced mitochondria biogenesis, increased Fe-S cluster synthesis inhibits the function of iron-regulating protein (IRP)1 and hence stimulates the expression of 5'-aminolevulinate synthase 2 (ALAS2), a target of IRP1 and rate-limiting enzyme in erythroid heme biogenesis. Increased ALAS2 expression leads to enhanced heme production, hemoglobinization, and erythropoiesis. Therefore, our study also provides a mechanism to link mitochondrial biogenesis with erythropoiesis and has a potential therapeutic value in the treatment of blood disorders.
Collapse
Affiliation(s)
- Ping La
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph H Oved
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Cell Therapy and Transplant Section, Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Valentina Ghiaccio
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stefano Rivella
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Cell and Molecular Biology Affinity Group (CAMB)-Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Penn Center for Musculoskeletal Disorders, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
43
|
van Vuren AJ, van Wijk R, van Beers EJ, Marx JJ. Liver Iron Retention Estimated from Utilization of Oral and Intravenous Radioiron in Various Anemias and Hemochromatosis in Humans. Int J Mol Sci 2020; 21:ijms21031077. [PMID: 32041196 PMCID: PMC7037197 DOI: 10.3390/ijms21031077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 01/19/2023] Open
Abstract
Patients with hereditary hemochromatosis and non-transfusion-dependent hereditary anemia develop predominantly liver iron-overload. We present a unique method allowing quantification of liver iron retention in humans during first-pass of 59Fe-labeled iron through the portal system, using standard ferrokinetic techniques measuring red cell iron uptake after oral and intravenous 59Fe administration. We present data from patients with iron deficiency (ID; N = 47), hereditary hemochromatosis (HH; N = 121) and non-transfusion-dependent hereditary anemia (HA; N = 40). Mean mucosal iron uptake and mucosal iron transfer (±SD) were elevated in patients with HH (59 ± 18%, 80 ± 15% respectively), HA (65 ± 17%, 74 ± 18%) and ID (84 ± 14%, 94 ± 6%) compared to healthy controls (43 ± 19%, 64 ± 18%) (p < 0.05) resulting in increased iron retention after 14 days compared to healthy controls in all groups (p < 0.01). The fraction of retained iron utilized for red cell production was 0.37 ± 0.17 in untreated HA, 0.55 ± 0.20 in untreated HH and 0.99 ± 0.22 in ID (p < 0.01). Interestingly, compared to red blood cell iron utilization after oral iron administration, red blood cell iron utilization was higher after injection of transferrin-bound iron in HA and HH. Liver iron retention was considerably higher in HH and HA compared to ID. We hypothesize that albumin serves as a scavenger of absorbed Fe(II) for delivering albumin-bound Fe(III) to hepatocytes.
Collapse
Affiliation(s)
- Annelies J. van Vuren
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Richard van Wijk
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Eduard J. van Beers
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-88-755-84-50
| | - Joannes J.M. Marx
- Departments of Haematology and Internal Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
44
|
Mohan V, Patil O, Ravikumar K, Gopi S, Raman T, Radha V. Thiamine-responsive megaloblastic anemia syndrome: A case report. JOURNAL OF DIABETOLOGY 2020. [DOI: 10.4103/jod.jod_51_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
45
|
Peoc'h K, Nicolas G, Schmitt C, Mirmiran A, Daher R, Lefebvre T, Gouya L, Karim Z, Puy H. Regulation and tissue-specific expression of δ-aminolevulinic acid synthases in non-syndromic sideroblastic anemias and porphyrias. Mol Genet Metab 2019; 128:190-197. [PMID: 30737140 DOI: 10.1016/j.ymgme.2019.01.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/21/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
Recently, new genes and molecular mechanisms have been identified in patients with porphyrias and sideroblastic anemias (SA). They all modulate either directly or indirectly the δ-aminolevulinic acid synthase (ALAS) activity. ALAS, is encoded by two genes: the erythroid-specific (ALAS2), and the ubiquitously expressed (ALAS1). In the liver, ALAS1 controls the rate-limiting step in the production of heme and hemoproteins that are rapidly turned over in response to metabolic needs. Several heme regulatory targets have been identified as regulators of ALAS1 activity: 1) transcriptional repression via a heme-responsive element, 2) post-transcriptional destabilization of ALAS1 mRNA, 3) post-translational inhibition via a heme regulatory motif, 4) direct inhibition of the activity of the enzyme and 5) breakdown of ALAS1 protein via heme-mediated induction of the protease Lon peptidase 1. In erythroid cells, ALAS2 is a gatekeeper of production of very large amounts of heme necessary for hemoglobin synthesis. The rate of ALAS2 synthesis is transiently increased during the period of active heme synthesis. Its gene expression is determined by trans-activation of nuclear factor GATA1, CACC box and NF-E2-binding sites in the promoter areas. ALAS2 mRNA translation is also regulated by the iron-responsive element (IRE)/iron regulatory proteins (IRP) binding system. In patients, ALAS enzyme activity is affected in most of the mutations causing non-syndromic SA and in several porphyrias. Decreased ALAS2 activity results either directly from loss-of-function ALAS2 mutations as seen in X-linked sideroblastic anemia (XLSA) or from defect in the availability of one of its two mitochondrial substrates: glycine in SLC25A38 mutations and succinyl CoA in GLRX5 mutations. Moreover, ALAS2 gain of function mutations is responsible for X-linked protoporphyria and increased ALAS1 activity lead to acute attacks of hepatic porphyrias. A missense dominant mutation in the Walker A motif of the ATPase binding site in the gene coding for the mitochondrial protein unfoldase CLPX also contributes to increasing ALAS and subsequently protoporphyrinemia. Altogether, these recent data on human ALAS have informed our understanding of porphyrias and sideroblastic anemias pathogeneses and may contribute to new therapeutic strategies.
Collapse
Affiliation(s)
- Katell Peoc'h
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Gaël Nicolas
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Caroline Schmitt
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| | - Arienne Mirmiran
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Raed Daher
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Thibaud Lefebvre
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| | - Laurent Gouya
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| | - Zoubida Karim
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France.
| | - Hervé Puy
- INSERM U1149, CNRS ERL 8252, Centre de Recherche sur l'inflammation, Université Paris Diderot, site Bichat, Sorbonne Paris Cité, France, 16 rue Henri Huchard, 75018 Paris, France; Laboratory of Excellence, GR-Ex, Paris, France; AP-HP, HUPNVS, Centre Français des Porphyries, Hôpital Louis Mourier, Colombes, France.
| |
Collapse
|
46
|
Fouquet C, Le Rouzic M, Leblanc T, Fouyssac F, Leverger G, Hessissen L, Marlin S, Bourrat E, Fahd M, Raffoux E, Vannier J, Jäkel N, Knoefler R, Triolo V, Pasquet M, Bayart S, Thuret I, Lutz P, Vermylen C, Touati M, Rose C, Matthes T, Isidor B, Kannengiesser C, Ducassou S. Genotype/phenotype correlations of childhood‐onset congenital sideroblastic anaemia in a European cohort. Br J Haematol 2019; 187:530-542. [DOI: 10.1111/bjh.16100] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/08/2019] [Indexed: 01/19/2023]
Affiliation(s)
| | | | | | | | - Guy Leverger
- CHU de Paris Hôpital Armand Trousseau Paris France
| | | | | | | | - Mony Fahd
- CHU de Paris Hôpital Robert Debré Paris France
| | | | | | - Nadja Jäkel
- Department für Hämatologie Onkologie und Hämostaseologie Leipzig Germany
| | - Ralf Knoefler
- Department of Pediatric Haemostaseology University Hospital Carl Gustav Carus Dresden Germany
| | | | | | | | | | - Patrick Lutz
- CHU de Strasbourg Hôpital de Hautepierre Strasbourg France
| | - Christiane Vermylen
- Université Catholique de Louvain, Cliniques universitaires Saint‐Luc Brussels Belgium
| | | | | | - Thomas Matthes
- Geneva University Hospital, Hematology Service Geneva Switzerland
| | | | - Caroline Kannengiesser
- Assistance Publique des Hôpitaux de Paris, Département de Génétique Hôpital Bichat, Université Paris VII Paris France
| | | |
Collapse
|
47
|
Comont T, Delavigne K, Cougoul P, Bertoli S, Delabesse E, Fenaux P, Beyne-Rauzy O. [Management of myelodysplastic syndromes in 2019: An update]. Rev Med Interne 2019; 40:581-589. [PMID: 31054780 DOI: 10.1016/j.revmed.2019.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/10/2019] [Indexed: 01/04/2023]
Abstract
Myelodysplastic syndromes are a heterogeneous group of clonal myeloid disorders characterized by peripheral cytopenias and an increased risk of progression to acute myeloid leukemia. Inflammatory, auto-immune or syndromic symptoms can make the diagnosis difficult. Diagnosis is currently based on bone marrow cytology but cytogenetics and molecular features are currently overpassing their initial prognostic function (allowing early diagnosis and prediction of therapeutic response). The prognostic classification is based on the Revised International Prognostic Scoring System, which also provides guidance for therapeutic management. The treatment of low-risk myelodysplastic syndromes is based on the correction of cytopenias (erythropoiesis stimulating agents, transfusions, lenalidomide, etc.), whereas in high-risk group, the goal is the control of the leukemic clone (hypomethylating agents, allograft of hematopoietic stem cell transplantation). Other molecules are used to manage complications of cytopenias or transfusion (anti-infectious prophylaxis and treatments, martial chelation). New molecules are being studied with some interesting results (luspatercept, venetoclax). This article aims to provide an update on the knowledge that an internist should know for the practical management of myelodysplastic syndromes in 2019.
Collapse
Affiliation(s)
- T Comont
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France; UFR Purpan, université Toulouse III Paul Sabatier, 31400 Toulouse, France; UMR1037-Inserm, ERL5294 CNRS, centre de recherche en cancérologie de Toulouse, 31100 Toulouse, France.
| | - K Delavigne
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France
| | - P Cougoul
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France
| | - S Bertoli
- UFR Purpan, université Toulouse III Paul Sabatier, 31400 Toulouse, France; UMR1037-Inserm, ERL5294 CNRS, centre de recherche en cancérologie de Toulouse, 31100 Toulouse, France; Service d'hématologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France
| | - E Delabesse
- Laboratoire d'hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopôle, 31100 Toulouse, France
| | - P Fenaux
- Service d'hématologie, hôpital Saint-Louis, AP-HP, 75010 Paris, France
| | - O Beyne-Rauzy
- Service de médecine interne et immunopathologie, institut universitaire du Cancer de Toulouse Oncopôle, centre hospitalier universitaire de Toulouse, 31100 Toulouse, France; UFR Purpan, université Toulouse III Paul Sabatier, 31400 Toulouse, France; UMR1037-Inserm, ERL5294 CNRS, centre de recherche en cancérologie de Toulouse, 31100 Toulouse, France
| |
Collapse
|
48
|
Generation and Molecular Characterization of Human Ring Sideroblasts: a Key Role of Ferrous Iron in Terminal Erythroid Differentiation and Ring Sideroblast Formation. Mol Cell Biol 2019; 39:MCB.00387-18. [PMID: 30670569 DOI: 10.1128/mcb.00387-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Ring sideroblasts are a hallmark of sideroblastic anemia, although little is known about their characteristics. Here, we first generated mutant mice by disrupting the GATA-1 binding motif at the intron 1 enhancer of the ALAS2 gene, a gene responsible for X-linked sideroblastic anemia (XLSA). Although heterozygous female mice showed an anemic phenotype, ring sideroblasts were not observed in their bone marrow. We next established human induced pluripotent stem cell-derived proerythroblast clones harboring the same ALAS2 gene mutation. Through coculture with sodium ferrous citrate, mutant clones differentiated into mature erythroblasts and became ring sideroblasts with upregulation of metal transporters (MFRN1, ZIP8, and DMT1), suggesting a key role for ferrous iron in erythroid differentiation. Interestingly, holo-transferrin (holo-Tf) did not induce erythroid differentiation as well as ring sideroblast formation, and mutant cells underwent apoptosis. Despite massive iron granule content, ring sideroblasts were less apoptotic than holo-Tf-treated undifferentiated cells. Microarray analysis revealed upregulation of antiapoptotic genes in ring sideroblasts, a profile partly shared with erythroblasts from a patient with XLSA. These results suggest that ring sideroblasts exert a reaction to avoid cell death by activating antiapoptotic programs. Our model may become an important tool to clarify the pathophysiology of sideroblastic anemia.
Collapse
|
49
|
Tesarova M, Vondrackova A, Stufkova H, Veprekova L, Stranecky V, Berankova K, Hansikova H, Magner M, Galoova N, Honzik T, Vodickova E, Stary J, Zeman J. Sideroblastic anemia associated with multisystem mitochondrial disorders. Pediatr Blood Cancer 2019; 66:e27591. [PMID: 30588737 DOI: 10.1002/pbc.27591] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/18/2018] [Accepted: 11/29/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Sideroblastic anemia represents a heterogeneous group of inherited or acquired diseases with disrupted erythroblast iron utilization, ineffective erythropoiesis, and variable systemic iron overload. In a cohort of 421 patients with multisystem mitochondrial diseases, refractory anemia was found in 8 children. RESULTS Five children had sideroblastic anemia with increased numbers of ring sideroblasts >15%. Two of the children had a fatal course of MLASA1 syndrome (mitochondrial myopathy, lactic acidosis, and sideroblastic anemia [SA]) due to a homozygous, 6-kb deletion in the PUS1 gene, part of the six-member family of pseudouridine synthases (pseudouridylases). Large homozygous deletions represent a novel cause of presumed PUS1-loss-of-function phenotype. The other three children with SA had Pearson syndrome (PS) due to mtDNA deletions of 4 to 8 kb; two of these children showed early onset of PS and died due to repeated sepsis; the other child had later onset of PS and survived as the hematological parameters normalized and the disease transitioned to Kearns-Sayre syndrome. In addition, anemia without ring sideroblasts was found in three other patients with mitochondrial disorders, including two children with later onset of PS and one child with failure to thrive, microcephaly, developmental delay, hypertrophic cardiomyopathy, and renal tubular acidosis due to the heterozygous mutations c.610A>G (p.Asn204Asp) and c.674C>T (p.Pro225Leu) in the COX10 gene encoding the cytochrome c oxidase assembly factor. CONCLUSIONS Sideroblastic anemia was found in fewer than 1.2% of patients with multisystem mitochondrial disease, and it was usually associated with an unfavorable prognosis.
Collapse
Affiliation(s)
- Marketa Tesarova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Alzbeta Vondrackova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Hana Stufkova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Lenka Veprekova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Viktor Stranecky
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Kamila Berankova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Hana Hansikova
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Magner
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Natalia Galoova
- Department of Paediatric Oncology and Haematology, Children's University Hospital, Kosice, Slovakia
| | - Tomas Honzik
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Elena Vodickova
- Department of Haematology, Motol University Hospital, Prague, Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jiri Zeman
- Department of Paediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
50
|
Fujiwara T, Harigae H. Molecular pathophysiology and genetic mutations in congenital sideroblastic anemia. Free Radic Biol Med 2019; 133:179-185. [PMID: 30098397 DOI: 10.1016/j.freeradbiomed.2018.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/02/2018] [Accepted: 08/04/2018] [Indexed: 01/19/2023]
Abstract
Sideroblastic anemia is a heterogeneous congenital and acquired disorder characterized by anemia and the presence of ring sideroblasts in the bone marrow. Congenital sideroblastic anemia (CSA) is a rare disease caused by mutations in genes involved in the heme biosynthesis, iron-sulfur [Fe-S] cluster biosynthesis, and mitochondrial protein synthesis. The most prevalent form of CSA is X-linked sideroblastic anemia, caused by mutations in the erythroid-specific δ-aminolevulinate synthase (ALAS2), which is the first enzyme of the heme biosynthesis pathway in erythroid cells. To date, a remarkable number of genetically undefined CSA cases remain, but a recent application of the next-generation sequencing technology has recognized novel causative genes for CSA. However, in most instances, the detailed molecular mechanisms of how defects of each gene result in the abnormal mitochondrial iron accumulation remain unclear. This review aims to cover the current understanding of the molecular pathophysiology of CSA.
Collapse
Affiliation(s)
- Tohru Fujiwara
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai 980-8575, Japan.
| |
Collapse
|