1
|
Boccadifuoco U, Cheminet G, Morino B, Arlet JB. [Extramedullary hematopoiesis, a rare complication of sickle cell disease: A six-case series and literature review]. Rev Med Interne 2025; 46:193-203. [PMID: 39779438 DOI: 10.1016/j.revmed.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/20/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION Extramedullary hematopoiesis (EMH) is very rarely described during sickle cell disease (SCD). Our aim was to describe six cases of EMH occurring in adult SCD patients and to conduct a literature review. METHODS Retrospective, descriptive, and monocentric study, identifying all cases of EMH recorded in our cohort of adult SCD patients, up to April 2024. A literature review via PubMed included thirty-five articles (44 patients). RESULTS Six patients (4 men, 83.3% with SS genotype [n=5], 1 SC), with a median age of 22 (range 12-64) years at the time of EMH diagnosis were included. Four patients (66.7%) had an aseptic osteonecrosis of the hip. The localization of EMH varied: paravertebral (n=3), peri-articular in the hip (n=1), adrenal (n=1), hepatic (n=1), splenic (n=1) and was similar to the localizations reported in the literature. EMH was symptomatic at diagnosis in half of the cases. The diagnosis was established by histology (n=3/3) and/or typic magnetic resonance imaging (MRI) (n=4/4). The median baseline hemoglobin was 9.1 (extremes 5.8-10.9) g/dL. A watch-and-wait approach was primarily observed. CONCLUSION EMH in SCD patients appears to be rare, with varied localizations. Its diagnosis is made with MRI and/or biopsy, and its treatment is not consensual.
Collapse
Affiliation(s)
- Ugo Boccadifuoco
- Service de médecine interne, centre national de référence des syndromes drépanocytaires majeurs de l'adulte, hôpital européen Georges-Pompidou, AP-HP, 20, rue Leblanc, 75015 Paris, France
| | - Geoffrey Cheminet
- Service de médecine interne, centre national de référence des syndromes drépanocytaires majeurs de l'adulte, hôpital européen Georges-Pompidou, AP-HP, 20, rue Leblanc, 75015 Paris, France; Inserm U1163, laboratoire « Mécanismes cellulaires et moléculaires des désordres hématologiques et implications thérapeutiques », institut Imagine, université Paris-Cité, 75015 Paris, France; Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| | - Benjamin Morino
- Service de radiologie diagnostique et interventionnelle, hôpital européen Georges-Pompidou, AP-HP, 75015 Paris, France
| | - Jean-Benoît Arlet
- Service de médecine interne, centre national de référence des syndromes drépanocytaires majeurs de l'adulte, hôpital européen Georges-Pompidou, AP-HP, 20, rue Leblanc, 75015 Paris, France; Inserm U1163, laboratoire « Mécanismes cellulaires et moléculaires des désordres hématologiques et implications thérapeutiques », institut Imagine, université Paris-Cité, 75015 Paris, France; Laboratoire d'Excellence GR-Ex, 75015 Paris, France; Faculté de santé, université Paris-Cité, 75006 Paris, France.
| |
Collapse
|
2
|
Sobrino S, Joseph L, Magrin E, Chalumeau A, Hebert N, Corsia A, Denis A, Roudaut C, Aussel C, Leblanc O, Brusson M, Felix T, Diana JS, Petrichenko A, El Etri J, Godard A, Tibi E, Manceau S, Treluyer JM, Mavilio F, Bushman FD, Marcais A, Castelle M, Neven B, Hermine O, Renolleau S, Magnani A, Asnafi V, El Nemer W, Bartolucci P, Six E, Semeraro M, Miccio A, Cavazzana M. Severe inflammation and lineage skewing are associated with poor engraftment of engineered hematopoietic stem cells in patients with sickle cell disease. Nat Commun 2025; 16:3137. [PMID: 40169559 PMCID: PMC11961595 DOI: 10.1038/s41467-025-58321-4] [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: 08/14/2024] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
In sickle cell disease (SCD), the β6Glu→Val substitution in the β-globin leads to red blood cell sickling. The transplantation of autologous, genetically modified hematopoietic stem and progenitor cells (HSPCs) is a promising treatment option for patients with SCD. We completed a Phase I/II open-label clinical trial (NCT03964792) for patients with SCD using a lentiviral vector (DREPAGLOBE) expressing a potent anti-sickling β-globin. The primary endpoint was to evaluate the short-term safety and secondary endpoints included the efficacy and the long-term safety. We report on the results after 18 to 36 months of follow-up. No drug-related adverse events or signs of clonal hematopoiesis were observed. Despite similar vector copy numbers in the drug product, gene-marking in peripheral blood mononuclear cells and correction of the clinical phenotype varied from one patient to another. Single-cell transcriptome analyses show that in the patients with poor engraftment, the most immature HSCs display an exacerbated inflammatory signature (via IL-1 or TNF-α and interferon signaling pathways). This signature is accompanied by a lineage bias in the HSCs. Our clinical data indicates that the DREPAGLOBE-based gene therapy (GT) is safe. However, its efficacy is variable and probably depends on the number of infused HSCs and intrinsic, engraftment-impairing inflammatory alterations in HSCs. Trial: NCT03964792.
Collapse
Affiliation(s)
- Steicy Sobrino
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Laure Joseph
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
| | - Elisa Magrin
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Anne Chalumeau
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Nicolas Hebert
- Univ Paris Est Créteil, IMRB, Laboratory of Excellence LABEX GRex, Créteil, France
- Etablissement Français du Sang, Créteil, France
| | - Alice Corsia
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Adeline Denis
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Cécile Roudaut
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Clotilde Aussel
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Olivia Leblanc
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Mégane Brusson
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Tristan Felix
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Jean-Sebastien Diana
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jana El Etri
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Auria Godard
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Eden Tibi
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
| | - Sandra Manceau
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Jean Marc Treluyer
- Centre d'Investigation Clinique-Unité de Recherche Clinique, Hôpital Universitaire Necker Enfants-Malades, GH Paris Centre, Paris, France
- Université Paris Cité, Paris, France
| | - Fulvio Mavilio
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ambroise Marcais
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Martin Castelle
- Pediatric Immunology and Hematology Department, Hôpital Necker Enfants-Malades, Paris, France
| | - Benedicte Neven
- Pediatric Immunology and Hematology Department, Hôpital Necker Enfants-Malades, Paris, France
| | - Olivier Hermine
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Sylvain Renolleau
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Alessandra Magnani
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Vahid Asnafi
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Wassim El Nemer
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Pablo Bartolucci
- Univ Paris Est Créteil, IMRB, Laboratory of Excellence LABEX GRex, Créteil, France
- Paris-East Créteil University, Henri Mondor University Hospitals, APHP, Sickle Cell Referral Center-UMGGR, Créteil, France
| | - Emmanuelle Six
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Michaela Semeraro
- Centre d'Investigation Clinique-Unité de Recherche Clinique, Hôpital Universitaire Necker Enfants-Malades, GH Paris Centre, Paris, France
- Université Paris Cité, Inserm, Pharmacologie et évaluations des thérapeutiques chez l'enfant et la femme enceinte, Paris, France
| | - Annarita Miccio
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Marina Cavazzana
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France.
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France.
| |
Collapse
|
3
|
Grenier JMP, Godard A, Seute R, Grimaldi A, Peyrard B, Chiaroni J, Mohandas N, El Nemer W, De Grandis M. A Novel Method for a Precise Identification of Human Erythroblast Subpopulations by Flow Cytometry. Am J Hematol 2025. [PMID: 40099790 DOI: 10.1002/ajh.27668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/20/2025]
Affiliation(s)
| | - Auria Godard
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Robert Seute
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | | | - Barbara Peyrard
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Jacques Chiaroni
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Narla Mohandas
- Research Laboratory of Red Cell Physiology, New York Blood Center, New York, New York, USA
| | - Wassim El Nemer
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Maria De Grandis
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| |
Collapse
|
4
|
Li J, Chen M, Zhao W, Lv A, Lin S, Zheng Y, Cai M, Lin N, Xu L, Huang H. The role of miR-129-5p in regulating γ-globin expression and erythropoiesis in β-thalassemia. Hum Mol Genet 2025; 34:291-303. [PMID: 39657657 DOI: 10.1093/hmg/ddae180] [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: 09/28/2024] [Revised: 11/04/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024] Open
Abstract
The regulation of γ-globin expression is crucial due to its beneficial effects on diseases like β-thalassemia and sickle cell disease. B-cell lymphoma/leukemia 11A (BCL11A) is a significant suppressor of γ-globin, and microRNAs (miRNAs) targeting BCL11A have been shown to alleviate this suppression. In our previous high-throughput sequencing, we identified an 11.32-fold increase in miR-129-5p expression in β-thalassemia patients. However, the regulatory mechanisms of miR-129-5p in the context of erythroid differentiation remain to be elucidated. Our study aimed to elucidate the role of miR-129-5p in γ-globin regulation and erythropoiesis. We measured miR-129-5p levels in peripheral blood from β-thalassemia major and intermedia patients. Fluorescence in situ hybridization, dual-luciferase reporter assays, miRNA pull down assays and western blot analyses were conducted to examine the effects of miR-129-5p on γ-globin expression and BCL11A repression. Cell proliferation, apoptosis, and erythroid differentiation were assessed using cell counting kit-8, Wright-Giemsa, and benzidine staining, and flow cytometry assays. The expression levels of miR-129-5p were significantly elevated in β-thalassemia patients and positively correlated with γ-globin synthesis while negatively correlating with liver damage. miR-129- 5p enhanced γ-globin gene expression in K562 and HUDEP-2 cells by effectively repressing BCL11A. Overexpression of miR-129-5p inhibited cell proliferation, induced cell cycle arrest at the G1/G0 phase, promoted apoptosis and stimulated erythroid differentiation and maturation. Conversely, inhibition of miR-129-5p produced opposite cellular effects. miR-129-5p acts as a positive regulator of erythroid differentiation and γ-globin synthesis. It offers a promising miRNA target for activating the γ-globin gene and reducing ineffective erythropoiesis in β-thalassemia patients.
Collapse
Affiliation(s)
- Jingmin Li
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Meihuan Chen
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Wantong Zhao
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Aixiang Lv
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Siyang Lin
- The School of Medical Technology and Engineering, Fujian Medical University, 1 Xuefu North Road, Minhou District, Fuzhou 350108, China
| | - Yanping Zheng
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Meiying Cai
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Na Lin
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| | - Hailong Huang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, 88 Jiaotong Road, Taijiang District, Fuzhou 350004, China
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, Fujian Key Laboratory for Prenatal Diagnosis and Birth Defect, 18 Daoshan Road, Gulou District, Fuzhou 350001, China
| |
Collapse
|
5
|
El Hoss S, Bazoum H. From old to new: Repurposed drugs in the battle towards curing sickle cell disease. Br J Haematol 2025; 206:795-797. [PMID: 39568202 PMCID: PMC11829130 DOI: 10.1111/bjh.19912] [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: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/22/2024]
Abstract
This commentary discusses the therapeutic potential of drug repurposing in sickle cell disease, highlighting the efficacy of hydroxyurea in enhancing fetal haemoglobin and the work of Raz et al. discussing the potential of using memantine for improving cognitive function, while emphasizing the need for further research. Commentary on: Raz et al. Memantine treatment in sickle cell disease: A 1-year study of its effects on cognitive functions and neural processing. Br J Haematol 2025; 206:689-702.
Collapse
Affiliation(s)
- Sara El Hoss
- Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic ImplicationsInstitut IMAGINE, INSERM UMR1163ParisFrance
| | - Haoua Bazoum
- Laboratory of Molecular Mechanisms of Hematological Disorders and Therapeutic ImplicationsInstitut IMAGINE, INSERM UMR1163ParisFrance
| |
Collapse
|
6
|
Joly A, Schott A, Phadke I, Gonzalez-Menendez P, Kinet S, Taylor N. Beyond ATP: Metabolite Networks as Regulators of Physiological and Pathological Erythroid Differentiation. Physiology (Bethesda) 2025; 40:0. [PMID: 39226028 DOI: 10.1152/physiol.00035.2024] [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: 07/15/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Hematopoietic stem cells (HSCs) possess the capacity for self-renewal and the sustained production of all mature blood cell lineages. It has been well established that a metabolic rewiring controls the switch of HSCs from a self-renewal state to a more differentiated state, but it is only recently that we have appreciated the importance of metabolic pathways in regulating the commitment of progenitors to distinct hematopoietic lineages. In the context of erythroid differentiation, an extensive network of metabolites, including amino acids, sugars, nucleotides, fatty acids, vitamins, and iron, is required for red blood cell (RBC) maturation. In this review, we highlight the multifaceted roles via which metabolites regulate physiological erythropoiesis as well as the effects of metabolic perturbations on erythroid lineage commitment and differentiation. Of note, the erythroid differentiation process is associated with an exceptional breadth of solute carrier (SLC) metabolite transporter upregulation. Finally, we discuss how recent research, revealing the critical impact of metabolic reprogramming in diseases of disordered and ineffective erythropoiesis, has created opportunities for the development of novel metabolic-centered therapeutic strategies.
Collapse
Affiliation(s)
- Axel Joly
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Arthur Schott
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Ira Phadke
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Pedro Gonzalez-Menendez
- Departamento de Morfologia y Biologia Celular, Instituto Universitario de Oncologia del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sandrina Kinet
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Naomi Taylor
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
7
|
Wilson SR, Mitin N, Miller VLA, Smitherman AB, Carden MA. Adolescents and young adults with sickle cell disease exhibit accelerated aging with elevated T-cell p16 INK4a expression. Aging (Albany NY) 2024; 16:13225-13236. [PMID: 39546497 PMCID: PMC11719104 DOI: 10.18632/aging.206152] [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: 06/06/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
People living with sickle cell disease (SCD) experience complications indicative of an accelerated aging phenotype typified by early decline in physical function and increased risk for age-related conditions. Cellular senescence, measured by expression of p16INK4a in peripheral T-lymphocytes, is recognized as one of the underlying contributors to organismal aging. To examine if cellular senescence is increased in SCD patients, we cross-sectionally measured and compared expression of p16 mRNA in peripheral blood T lymphocytes in 18 adolescents and young adults with SCD to 27 similarly aged individuals without SCD. Expression of p16 was dramatically higher in individuals with SCD vs. without SCD (10.1 vs. 8.7 log2 p16 units, respectively, p < 0.001) - a gap of 43 years in biological age - consistent with accelerated aging in the SCD population. Race was not associated with the increased p16 expression in the SCD group. These initial results suggest that individuals with SCD have a significantly higher cellular senescence burden which may contribute to premature aging, physiological decline, and excess morbidities. Additional longitudinal assessment and consideration for trials of senolytic therapies among individuals living with SCD and high p16 expression are warranted to improve their health span.
Collapse
Affiliation(s)
- Samuel R. Wilson
- Department of Medicine, Division of Hematology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalia Mitin
- Sapere Bio, Research Triangle Park, Raleigh-Durham-Chapel Hill, NC 27709, USA
| | - Vanessa L. Ayer Miller
- Department of Pharmaceutical and Clinical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Andrew B. Smitherman
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Department of Pharmaceutical and Clinical Sciences, College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC 27506, USA
| | - Marcus A. Carden
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Cogent Biosciences, Waltham, MA 02451, USA
| |
Collapse
|
8
|
Sanchez-Villalobos M, Campos Baños E, Martínez-Balsalobre E, Navarro-Ramirez V, Videla MAB, Pinilla M, Guillén-Navarro E, Salido-Fierrez E, Pérez-Oliva AB. Whole Blood Transcriptome Analysis in Congenital Anemia Patients. Int J Mol Sci 2024; 25:11706. [PMID: 39519257 PMCID: PMC11546763 DOI: 10.3390/ijms252111706] [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: 09/30/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Congenital anemias include a broad range of disorders marked by inherent abnormalities in red blood cells. These abnormalities include enzymatic, membrane, and congenital defects in erythropoiesis, as well as hemoglobinopathies such as sickle cell disease and thalassemia. These conditions range in presentation from asymptomatic cases to those requiring frequent blood transfusions, exhibiting phenotypic heterogeneity and different degrees of severity. Despite understanding their different etiologies, all of them have a common pathophysiological origin with congenital defects of erythropoiesis. We can find different types, from congenital sideroblastic anemia (CSA), which is a bone marrow failure anemia, to hemoglobinopathies as sickle cell disease and thalassemia, with a higher prevalence and clinical impact. Recent efforts have focused on understanding erythropoiesis dysfunction in these anemias but, so far, deep gene sequencing analysis comparing all of them has not been performed. Our study used Quant 3' mRNA-Sequencing to compare transcriptomic profiles of four sickle cell disease patients, ten thalassemia patients, and one rare case of SLC25A38 CSA. Our results showed clear differentiated gene map expressions in all of them with respect to healthy controls. Our study reveals that genes related to metabolic processes, membrane genes, and erythropoiesis are upregulated with respect to healthy controls in all pathologies studied except in the SLC25A38 CSA patient, who shows a unique gene expression pattern compared to the rest of the congenital anemias studied. Our analysis is the first that compares gene expression patterns across different congenital anemias to provide a broad spectrum of genes that could have clinical relevance in these pathologies.
Collapse
Affiliation(s)
- Maria Sanchez-Villalobos
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
- Hematology Service, Virgen de la Arrixaca University Hospital, 30120 Murcia, Spain
| | - Eulalia Campos Baños
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
| | - Elena Martínez-Balsalobre
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
| | - Veronica Navarro-Ramirez
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
| | | | - Miriam Pinilla
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
| | - Encarna Guillén-Navarro
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
- Medical Genetics Section and Pediatrics Service, Virgen de la Arrixaca University Hospital, University of Murcia, 30120 Murcia, Spain
- CIBERER-ISCIII, 28029 Madrid, Spain
| | - Eduardo Salido-Fierrez
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
- Hematology Service, Virgen de la Arrixaca University Hospital, 30120 Murcia, Spain
| | - Ana Belén Pérez-Oliva
- Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), 30120 Murcia, Spain (E.C.B.); (M.A.B.V.)
| |
Collapse
|
9
|
Igbineweka NE, van Loon JJWA. Gene-environmental influence of space and microgravity on red blood cells with sickle cell disease. NPJ Genom Med 2024; 9:44. [PMID: 39349487 PMCID: PMC11442622 DOI: 10.1038/s41525-024-00427-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/09/2024] [Indexed: 10/02/2024] Open
Abstract
A fundamental question in human biology and for hematological disease is how do complex gene-environment interactions lead to individual disease outcome? This is no less the case for sickle cell disease (SCD), a monogenic disorder of Mendelian inheritance, both clinical course, severity, and treatment response, is variable amongst affected individuals. New insight and discovery often lie between the intersection of seemingly disparate disciplines. Recently, opportunities for space medicine have flourished and have offered a new paradigm for study. Two recent Nature papers have shown that hemolysis and oxidative stress play key mechanistic roles in erythrocyte pathogenesis during spaceflight. This paper reviews existing genetic and environmental modifiers of the sickle cell disease phenotype. It reviews evidence for erythrocyte pathology in microgravity environments and demonstrates why this may be relevant for the unique gene-environment interaction of the SCD phenotype. It also introduces the hematology and scientific community to methodological tools for evaluation in space and microgravity research. The increasing understanding of space biology may yield insight into gene-environment influences and new treatment paradigms in SCD and other hematological disease phenotypes.
Collapse
Affiliation(s)
- Norris E Igbineweka
- Imperial College London, Centre for Haematology, Department of Immunology & Inflammation, Commonwealth Building, Hammersmith Campus, Du Cane, London, W12 0NN, UK.
- Department of Haematology, King's College Hospital NHS Foundation Trust Denmark Hill, SE5 9RS, London, UK.
| | - Jack J W A van Loon
- Dutch Experiment Support Center (DESC), Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam Bone Center (ABC), Amsterdam UMC Location VU University Medical Center (VUmc) & Academic Centre for Dentistry Amsterdam (ACTA), Gustav Mahlerlaan 3004, 1081, LA Amsterdam, The Netherlands
- European Space Agency (ESA), European Space Research and Technology Centre (ESTEC), TEC-MMG, Keplerlaan 1, 2201, AZ Noordwijk, The Netherlands
| |
Collapse
|
10
|
Ubaid S, Kashif M, Laiq Y, Nayak AK, Kumar V, Singh V. Targeting HIF-1α in sickle cell disease and cancer: unraveling therapeutic opportunities and risks. Expert Opin Ther Targets 2024; 28:357-373. [PMID: 38861226 DOI: 10.1080/14728222.2024.2367640] [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: 02/10/2024] [Accepted: 06/10/2024] [Indexed: 06/12/2024]
Abstract
INTRODUCTION HIF-1α, a key player in medical science, holds immense significance in therapeutic approaches. This review delves into its complex dynamics, emphasizing the delicate balance required for its modulation. HIF-1α stands as a cornerstone in medical research, its role extending to therapeutic strategies. This review explores the intricate interplay surrounding HIF-1α, highlighting its critical involvement and the necessity for cautious modulation. AREAS COVERED In sickle cell disease (SCD), HIF-1α's potential to augment fetal hemoglobin (HbF) production and mitigate symptoms is underscored. Furthermore, its role in cancer is examined, particularly its influence on survival in hypoxic tumor microenvironments, angiogenesis, and metastasis. The discussion extends to the intricate relationship between HIF-1α modulation and cancer risks in SCD patients, emphasizing the importance of balancing therapeutic benefits and potential hazards. EXPERT OPINION Managing HIF-1α modulation in SCD patients requires a nuanced approach, considering therapeutic potential alongside associated risks, especially in exacerbating cancer risks. An evolutionary perspective adds depth, highlighting adaptations in populations adapted to low-oxygen environments and aligning cancer cell metabolism with primitive cells. The role of HIF-1α as a therapeutic target is discussed within the context of complex cancer biology and metabolism, acknowledging varied responses across diverse cancers influenced by intricate evolutionary adaptations.
Collapse
Affiliation(s)
- Saba Ubaid
- Department of Biochemistry, King George's Medical University, Lucknow, India
| | - Mohammad Kashif
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Yusra Laiq
- Department of Biotechnology, Era University, Lucknow, India
| | | | - Vipin Kumar
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | - Vivek Singh
- Department of Biochemistry, King George's Medical University, Lucknow, India
| |
Collapse
|
11
|
Agbalalah T, Bur D, Nwonu EJ, Rowaiye AB. Gut Microbiota: Potential Therapeutic Target for Sickle Cell Disease Pain and Complications. Anemia 2024; 2024:5431000. [PMID: 38533265 PMCID: PMC10965282 DOI: 10.1155/2024/5431000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
Aim Sickle cell disease has witnessed a 41.4% surge from 2000 to 2021, significantly affecting morbidity and mortality rates, particularly in children from regions with elevated under-5 mortality rates. Gut microbiota dysbiosis is increasingly recognised in SCD, exacerbating complications, particularly chronic pain, marked by significant alterations of proinflammatory bacteria abundance. This review explores the therapeutic potential of Akkermansia muciniphila and Roseburia spp. in alleviating SCD-related complications, emphasising their roles in maintaining gut barrier integrity, reducing inflammation, and modulating immune responses. Method A literature search up to November 2023 using PubMed, MEDLINE, and Google Scholar databases explored SCD pathophysiology, gut microbiota composition, Akkermansia muciniphila and Roseburia spp. abundance, pain and gut dysbiosis in SCD, and butyrate therapy. Result A. muciniphila and Roseburia spp. supplementation shows promise in alleviating chronic pain by addressing gut dysbiosis, offering new avenues for sustainable SCD management. This approach holds the potential for reducing reliance on reactive treatments and improving overall quality of life. This research underscores the pivotal role of the gut microbiome in SCD, advocating for personalised treatment approaches. Conclusion Further exploration and clinical trials are needed to harness the full potential of these gut bacteria for individuals affected by this challenging condition.
Collapse
Affiliation(s)
- Tarimoboere Agbalalah
- Department of Anatomy, Faculty of Basic Medical Sciences, Baze University, Abuja, Nigeria
- Department of Medical Biotechnology, National Biotechnology Development Agency, Abuja, Nigeria
| | - Doofan Bur
- Department of Medical Biotechnology, National Biotechnology Development Agency, Abuja, Nigeria
| | | | | |
Collapse
|
12
|
Han Y, Gao C, Liu Y, Zhang H, Wang S, Zhao H, Bao W, Guo X, Vinchi F, Lobo C, Shi P, Mendelson A, Luchsinger L, Zhong H, Yazdanbakhsh K, An X. Hemolysis-driven IFNα production impairs erythropoiesis by negatively regulating EPO signaling in sickle cell disease. Blood 2024; 143:1018-1031. [PMID: 38127913 PMCID: PMC10950476 DOI: 10.1182/blood.2023021658] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/27/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023] Open
Abstract
ABSTRACT Disordered erythropoiesis is a feature of many hematologic diseases, including sickle cell disease (SCD). However, very little is known about erythropoiesis in SCD. Here, we show that although bone marrow (BM) erythroid progenitors and erythroblasts in Hbbth3/+ thalassemia mice were increased more than twofold, they were expanded by only ∼40% in Townes sickle mice (SS). We further show that the colony-forming ability of SS erythroid progenitors was decreased and erythropoietin (EPO)/EPO receptor (EPOR) signaling was impaired in SS erythroid cells. Furthermore, SS mice exhibited reduced responses to EPO. Injection of mice with red cell lysates or hemin, mimicking hemolysis in SCD, led to suppression of erythropoiesis and reduced EPO/EPOR signaling, indicating hemolysis, a hallmark of SCD, and could contribute to the impaired erythropoiesis in SCD. In vitro hemin treatment did not affect Stat5 phosphorylation, suggesting that hemin-induced erythropoiesis suppression in vivo is via an indirect mechanism. Treatment with interferon α (IFNα), which is upregulated by hemolysis and elevated in SCD, led to suppression of mouse BM erythropoiesis in vivo and human erythropoiesis in vitro, along with inhibition of Stat5 phosphorylation. Notably, in sickle erythroid cells, IFN-1 signaling was activated and the expression of cytokine inducible SH2-containing protein (CISH), a negative regulator of EPO/EPOR signaling, was increased. CISH deletion in human erythroblasts partially rescued IFNα-mediated impairment of cell growth and EPOR signaling. Knocking out Ifnar1 in SS mice rescued the defective BM erythropoiesis and improved EPO/EPOR signaling. Our findings identify an unexpected role of hemolysis on the impaired erythropoiesis in SCD through inhibition of EPO/EPOR signaling via a heme-IFNα-CISH axis.
Collapse
Affiliation(s)
- Yongshuai Han
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - Chengjie Gao
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - Yunfeng Liu
- Laboratory of Complement Biology, New York Blood Center, New York, NY
| | - Huan Zhang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - Shihui Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - Huizhi Zhao
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - Weili Bao
- Laboratory of Complement Biology, New York Blood Center, New York, NY
| | - Xinhua Guo
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| | - Francesca Vinchi
- Laboratory of Iron Research, New York Blood Center, New York, NY
| | - Cheryl Lobo
- Laboratory of Blood Borne Parasites, New York Blood Center, New York, NY
| | - Patricia Shi
- Sickle Cell Clinical Research Program, New York Blood Center, New York, NY
| | - Avital Mendelson
- Laboratory of Stem Cell Biology and Engineering Research, New York Blood Center, New York, NY
| | - Larry Luchsinger
- Laboratory of Stem Cell Regenerative Research, New York Blood Center, New York, NY
| | - Hui Zhong
- Laboratory of Immune Regulation, New York Blood Center, New York, NY
| | | | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY
| |
Collapse
|
13
|
Corbacioglu S, Frangoul H, Locatelli F, Hobbs W, Walters M. Defining curative endpoints for transfusion-dependent β-thalassemia in the era of gene therapy and gene editing. Am J Hematol 2024; 99:422-429. [PMID: 38100154 DOI: 10.1002/ajh.27166] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 02/15/2024]
Abstract
β-thalassemia is a monogenic disease that results in varying degrees of anemia. In the most severe form, known as transfusion-dependent β-thalassemia (TDT), the clinical hallmarks are ineffective erythropoiesis and a requirement of regular, life-long red blood cell transfusions, with the development of secondary clinical complications such as iron overload, end-organ damage, and a risk of early mortality. With the exception of allogeneic hematopoietic cell transplantation, current treatments for TDT address disease symptoms and not the underlying cause of disease. Recently, a growing number of gene addition and gene editing-based treatments for patients with TDT with the potential to provide a one-time functional cure have entered clinical trials. A key challenge in the design and evaluation of these trials is selecting endpoints to evaluate if these novel genetic therapies have a curative versus an ameliorative effect. Here, we present an overview of the pathophysiology of TDT, review emerging gene addition or gene editing therapeutic approaches for TDT currently in clinical trials, and identify a series of endpoints that can quantify therapeutic effects, including a curative outcome.
Collapse
Affiliation(s)
| | - Haydar Frangoul
- Sarah Cannon Research Institute and the Children's Hospital at TriStar Centennial, Nashville, Tennessee, USA
| | - Franco Locatelli
- IRCCS, Ospedale Pediatrico Bambino, Gesù Rome, Catholic University of the Sacred Heart, Rome, Italy
| | - William Hobbs
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| | - Mark Walters
- Department of Pediatrics, UCSF Benioff Children's Hospital Oakland, Oakland, California, USA
| |
Collapse
|
14
|
Lin S, Zheng Y, Chen M, Xu L, Huang H. The interactions between ineffective erythropoiesis and ferroptosis in β-thalassemia. Front Physiol 2024; 15:1346173. [PMID: 38468700 PMCID: PMC10925657 DOI: 10.3389/fphys.2024.1346173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
In Guangxi, Hainan, and Fujian Province in southern China, β-thalassemia is a frequent monogenic hereditary disorder that is primarily defined by hemolytic anemia brought on by inefficient erythropoiesis. It has been found that ineffective erythropoiesis in β-thalassemia is closely associated with a high accumulation of Reactive oxygen species, a product of oxidative stress, in erythroid cells. During recent years, ferroptosis is an iron-dependent lipid peroxidation that involves abnormalities in lipid and iron metabolism as well as reactive oxygen species homeostasis. It is a recently identified kind of programmed cell death. β-thalassemia patients experience increased iron release from reticuloendothelial cells and intestinal absorption of iron, ultimately resulting in iron overload. Additionally, the secretion of Hepcidin is inhibited in these patients. What counts is both ineffective erythropoiesis and ferroptosis in β-thalassemia are intricately linked to the iron metabolism and Reactive oxygen species homeostasis. Consequently, to shed further light on the pathophysiology of β-thalassemia and propose fresh ideas for its therapy, this paper reviews ferroptosis, ineffective erythropoiesis, and the way they interact.
Collapse
Affiliation(s)
- Siyang Lin
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
| | - Yanping Zheng
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Meihuan Chen
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| | - Liangpu Xu
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| | - Hailong Huang
- Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- The School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, China
- Fujian Clinical Research Center for Maternal-Fetal Medicine, Fuzhou, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, China
| |
Collapse
|
15
|
Godard A, Seute R, Grimaldi A, Granier T, Chiaroni J, El Nemer W, De Grandis M. A comparative study of two routinely used protocols for ex vivo erythroid differentiation. Blood Cells Mol Dis 2024; 106:102829. [PMID: 38278056 DOI: 10.1016/j.bcmd.2024.102829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Erythropoiesis is a complex developmental process in which a hematopoietic stem cell undergoes serial divisions and differentiates through well-defined stages to give rise to red blood cells. Over the last decades, several protocols have been developed to perform ex vivo erythroid differentiation, allowing investigation into erythropoiesis and red cell production in health and disease. RESULTS In the current study, we compared the two commonly used protocols by assessing the differentiation kinetics, synchronisation, and cellular yield, using molecular and cellular approaches. Peripheral blood CD34+ cells were cultured in a two-phase (2P) or a four-phase (4P) liquid culture (LC) and monitored for 20 days. Both protocols could recapitulate all stages of erythropoiesis and generate reticulocytes, although to different extents. Higher proliferation and viability rates were achieved in the 4P-LC, with a higher degree of terminal differentiation and enucleation, associated with higher levels of the erythroid-specific transcription factors GATA-1, KLF-1, and TAL-1. Although the 2P-LC protocol was less efficient regarding terminal erythroid differentiation and maturation, it showed a higher yield of erythroid progenitors in the erythropoietin (EPO)-free expansion phase. CONCLUSIONS We provide data supporting the use of one protocol or the other to study the biological processes occurring in the early or late stages of erythroid differentiation, depending on the physiological process or pathological defect under investigation in a given study.
Collapse
Affiliation(s)
- Auria Godard
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Robert Seute
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Alexandra Grimaldi
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Thomas Granier
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Jacques Chiaroni
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Wassim El Nemer
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Maria De Grandis
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France.
| |
Collapse
|
16
|
Bouyssou I, El Hoss S, Doderer-Lang C, Schoenhals M, Rasoloharimanana LT, Vigan-Womas I, Ratsimbasoa A, Abate A, Golassa L, Mabilotte S, Kessler P, Guillotte-Blisnick M, Martinez FJ, Chitnis CE, Strouboulis J, Ménard D. Unveiling P. vivax invasion pathways in Duffy-negative individuals. Cell Host Microbe 2023; 31:2080-2092.e5. [PMID: 38056460 PMCID: PMC10727064 DOI: 10.1016/j.chom.2023.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/01/2023] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
Vivax malaria has long been thought to be absent from sub-Saharan Africa owing to the high proportion of individuals lacking the Duffy antigen receptor for chemokines (DARC) in their erythrocytes. The interaction between P. vivax Duffy-binding protein (PvDBP) and DARC is assumed to be the main pathway used by merozoites to invade reticulocytes. However, the increasing number of reports of vivax malaria cases in genotypically Duffy-negative (DN) individuals has raised questions regarding the P. vivax invasion pathway(s). Here, we show that a subset of DN erythroblasts transiently express DARC during terminal erythroid differentiation and that P. vivax merozoites, irrespective of their origin, can invade DARC+ DN erythroblasts. These findings reveal that a large number of DN individuals may represent a silent reservoir of deep P. vivax infections at the sites of active erythropoiesis with low or no parasitemia, and it may represent an underestimated biological problem with potential clinical consequences in sub-Saharan Africa.
Collapse
Affiliation(s)
- Isabelle Bouyssou
- Malaria Genetics and Resistance Unit, INSERM U1201, Institut Pasteur, Université Paris Cité, 75015 Paris, France; École Doctorale ED515 "Complexité du Vivant", Sorbonne Université, 75005 Paris, France; Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Sara El Hoss
- Red Cell Haematology Laboratory, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London SE5 9NU, UK.
| | - Cécile Doderer-Lang
- Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, Université de Strasbourg, 67000 Strasbourg, France
| | - Matthieu Schoenhals
- Immunology of Infectious Diseases, Institut Pasteur of Madagascar, Antananarivo 101, Madagasca
| | | | | | - Arsène Ratsimbasoa
- Faculté de Médecine, Université de Fianarantsoa, Fianarantsoa 301, Madagascar
| | - Andargie Abate
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, PO Box 1176, Addis Ababa, Ethiopia
| | - Lemu Golassa
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, PO Box 1176, Addis Ababa, Ethiopia
| | - Solenne Mabilotte
- Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, Université de Strasbourg, 67000 Strasbourg, France
| | - Pascal Kessler
- Centre de Recherche en Biomédecine de Strasbourg, Université de Strasbourg, 67000 Strasbourg, France
| | | | - Francisco J Martinez
- Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Chetan E Chitnis
- Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - John Strouboulis
- Red Cell Haematology Laboratory, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London SE5 9NU, UK.
| | - Didier Ménard
- Malaria Genetics and Resistance Unit, INSERM U1201, Institut Pasteur, Université Paris Cité, 75015 Paris, France; Malaria Parasite Biology and Vaccines Unit, Institut Pasteur, Université Paris Cité, 75015 Paris, France; Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, Université de Strasbourg, 67000 Strasbourg, France; Laboratory of Parasitology and Medical Mycology, CHU Strasbourg, 67000 Strasbourg, France.
| |
Collapse
|
17
|
Bartolucci P. Exploration de l’hémolyse associée à la drépanocytose et perspectives thérapeutiques spécifiques. Rev Med Interne 2023; 44:4S7-4S11. [PMID: 38049244 DOI: 10.1016/s0248-8663(23)01303-6] [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] [Indexed: 12/06/2023]
Abstract
Sickle Cell Anemia is a disease with a strong vascular tropism. Beyond anemia, the pathophysiological mechanisms responsible for hemolysis, directly affect both acute and chronic vascular damages, thus resulting in a systemic disease. Understanding the different types of hemolysis underline the need for novel specific biomarkers. Targeted therapeutic approaches for these pathophysiological pathways are necessary to improve Sickle Cell patients' prognosis. Finally, given its complexity, Sickle Cell Disease is often used as a "proof of concept" for other pathologies. It seems likely that the rapidly evolving knowledge in this field will also benefit other diseases. © 2023 Société nationale française de médecine interne (SNFMI). Published by Elsevier Masson SAS. All rights reserved.
Collapse
Affiliation(s)
- P Bartolucci
- Recherche IH, EFS Créteil-Mondor, Créteil, France; IMRB Inserm 955, équipe Pirenne, Créteil, France; Centre de références des syndromes drépanocytaires majeures, hôpital Henri-Mondor, Créteil, France; Université Paris-Est Créteil, Créteil, France.
| |
Collapse
|
18
|
Ginzburg Y, An X, Rivella S, Goldfarb A. Normal and dysregulated crosstalk between iron metabolism and erythropoiesis. eLife 2023; 12:e90189. [PMID: 37578340 PMCID: PMC10425177 DOI: 10.7554/elife.90189] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023] Open
Abstract
Erythroblasts possess unique characteristics as they undergo differentiation from hematopoietic stem cells. During terminal erythropoiesis, these cells incorporate large amounts of iron in order to generate hemoglobin and ultimately undergo enucleation to become mature red blood cells, ultimately delivering oxygen in the circulation. Thus, erythropoiesis is a finely tuned, multifaceted process requiring numerous properly timed physiological events to maintain efficient production of 2 million red blood cells per second in steady state. Iron is required for normal functioning in all human cells, the erythropoietic compartment consuming the majority in light of the high iron requirements for hemoglobin synthesis. Recent evidence regarding the crosstalk between erythropoiesis and iron metabolism sheds light on the regulation of iron availability by erythroblasts and the consequences of insufficient as well as excess iron on erythroid lineage proliferation and differentiation. In addition, significant progress has been made in our understanding of dysregulated iron metabolism in various congenital and acquired malignant and non-malignant diseases. Finally, we report several actual as well as theoretical opportunities for translating the recently acquired robust mechanistic understanding of iron metabolism regulation to improve management of patients with disordered erythropoiesis, such as anemia of chronic inflammation, β-thalassemia, polycythemia vera, and myelodysplastic syndromes.
Collapse
Affiliation(s)
- Yelena Ginzburg
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Xiuli An
- LFKRI, New York Blood CenterNew YorkUnited States
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Cell and Molecular Biology affinity group (CAMB), University of PennsylvaniaPhiladelphiaUnited States
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics at the Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Penn Center for Musculoskeletal Disorders at the Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at University of PennsylvaniaPhiladelphiaUnited States
- RNA Institute at University of PennsylvaniaPhiladelphiaUnited States
| | - Adam Goldfarb
- Department of Pathology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
19
|
Zhang X, Song J, Shah BN, Han J, Hassan T, Miasniakova G, Sergueeva A, Nekhai S, Machado RF, Gladwin MT, Saraf SL, Prchal JT, Gordeuk VR. Gene expression changes in sickle cell reticulocytes and their clinical associations. Sci Rep 2023; 13:12864. [PMID: 37553354 PMCID: PMC10409856 DOI: 10.1038/s41598-023-40039-2] [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: 05/05/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023] Open
Abstract
Transcriptional changes in compensatory erythropoiesis in sickle cell anemia (SCA) and their disease modulation are unclear. We detected 1226 differentially expressed genes in hemoglobin SS reticulocytes compared to non-anemic hemoglobin AA controls. Assessing developmental expression changes in hemoglobin AA erythroblasts for these genes suggests heightened terminal differentiation in early erythroblasts in SCA that diminishes toward the polychromatic to orthochromatic stage transition. Comparison of reticulocyte gene expression changes in SCA with that in Chuvash erythrocytosis, a non-anemic disorder of increased erythropoiesis due to constitutive activation of hypoxia inducible factors, identified 453 SCA-specific changes attributable to compensatory erythropoiesis. Peripheral blood mononuclear cells (PBMCs) in SCA contain elevated proportions of erythroid progenitors due to heightened erythropoiesis. Deconvolution analysis in PBMCs from 131 SCA patients detected 54 genes whose erythroid expression correlated with erythropoiesis efficiency, which were enriched with SCA-specific changes (OR = 2.9, P = 0.00063) and annotation keyword "ubiquitin-dependent protein catabolic process", "protein ubiquitination", and "protein polyubiquitination" (OR = 4.2, P = 7.5 × 10-5). An erythroid expression quantitative trait locus of one of these genes, LNX2 encoding an E3 ubiquitin ligase, associated with severe pain episodes in 774 SCA patients (OR = 1.7, P = 3.9 × 10-5). Thus, erythroid gene transcription responds to unique conditions within SCA erythroblasts and these changes potentially correspond to vaso-occlusive manifestations.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| | - Jihyun Song
- Department of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Binal N Shah
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jin Han
- College of Pharmacy, University of Illinois at Chicago, Chicago, IL, USA
| | - Taif Hassan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | - Sergei Nekhai
- Center for Sickle Cell Disease, Howard University, Washington, DC, USA
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Mark T Gladwin
- Division of Pulmonary, Allergy, and Critical Care Medicine, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Santosh L Saraf
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Josef T Prchal
- Department of Medicine, University of Utah, Salt Lake City, UT, USA.
| | - Victor R Gordeuk
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
20
|
Albahout KS, Yunus M, Mohammad YG, Almalki AF, Alduailej SK, Alanazi BZ. Correlation of Transfusion Dependence and Its Associated Sequelae to Hematological and Biochemical Parameters in Patients With Sickle Cell Disease and Beta Thalassemia Major in Khobar: A Retrospective Study. Cureus 2023; 15:e42151. [PMID: 37602131 PMCID: PMC10438923 DOI: 10.7759/cureus.42151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Sickle cell disease (SCD) and beta thalassemia major (βTM) are multisystemic, genetically inherited diseases. They are caused by mutations of hemoglobin, which ultimately cause abnormal functioning of the red blood cells. The morbidity and mortality rates of these diseases are significant, as they may result in severe complications, some of which are quite fatal; hence, early diagnosis and treatment are crucial. The purpose of this study is to collect patients' data in terms of their manifestations and overall clinical picture and correlate them to the laboratory parameters with emphasis on their transfusion dependence and its sequelae in King Fahd Hospital of the University (KFHU), Al-Khobar, Saudi Arabia. After obtaining ethical approval from the institutional review board and in collaboration with the blood bank, patients' data were retrospectively collected from the hospital's database and categorized into two disease groups. Accordingly, data related to the biological and demographic information, clinical picture pattern, laboratory investigations, and therapeutic measures, with emphasis on blood transfusion as a treatment option, were gathered and analyzed. Eventually, the aforementioned data aspects were assessed for the probability of correlations, which were proven to be present to some level as an answer to our cohort study's question. Such findings, which will be depicted later in this study, might represent a ground for having a more comprehensive and extensive approach in terms of the general evaluation of patients with SCD and βTM based on the established level of correlation. During the course of conducting our research, we encountered some limitations, including the sample size and scarce data available during the process of data collection.
Collapse
Affiliation(s)
- Khaled S Albahout
- General Surgery, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | - Mohammed Yunus
- Pathology, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | | | - Adnan F Almalki
- Medicine, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| | | | - Basel Z Alanazi
- Medicine, Imam Abdulrahman Bin Faisal University, Dammam, SAU
| |
Collapse
|
21
|
Shangaris P, El Hoss S. Fetal microchimerism and beyond: a new player in regenerative medicine. Haematologica 2023; 108:1731-1733. [PMID: 36700405 PMCID: PMC10316241 DOI: 10.3324/haematol.2022.282244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Affiliation(s)
- Panicos Shangaris
- Women and Children's Health, School of Life Course and Population Sciences, Kings College London, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London.
| | - Sara El Hoss
- Red Cell Hematology Lab, Comprehensive Cancer Center, School of Cancer and Pharmaceutical Sciences, King's College London
| |
Collapse
|
22
|
Venkatesan V, Christopher AC, Rhiel M, Azhagiri MKK, Babu P, Walavalkar K, Saravanan B, Andrieux G, Rangaraj S, Srinivasan S, Karuppusamy KV, Jacob A, Bagchi A, Pai AA, Nakamura Y, Kurita R, Balasubramanian P, Pai R, Marepally SK, Mohankumar KM, Velayudhan SR, Boerries M, Notani D, Cathomen T, Srivastava A, Thangavel S. Editing the core region in HPFH deletions alters fetal and adult globin expression for treatment of β-hemoglobinopathies. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:671-688. [PMID: 37215154 PMCID: PMC10197010 DOI: 10.1016/j.omtn.2023.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 04/24/2023] [Indexed: 05/24/2023]
Abstract
Reactivation of fetal hemoglobin (HbF) is a commonly adapted strategy to ameliorate β-hemoglobinopathies. However, the continued production of defective adult hemoglobin (HbA) limits HbF tetramer production affecting the therapeutic benefits. Here, we evaluated deletional hereditary persistence of fetal hemoglobin (HPFH) mutations and identified an 11-kb sequence, encompassing putative repressor region (PRR) to β-globin exon-1 (βE1), as the core deletion that ablates HbA and exhibits superior HbF production compared with HPFH or other well-established targets. PRR-βE1-edited hematopoietic stem and progenitor cells (HSPCs) retained their genome integrity and their engraftment potential to repopulate for long-term hematopoiesis in immunocompromised mice producing HbF positive cells in vivo. Furthermore, PRR-βE1 gene editing is feasible without ex vivo HSPC culture. Importantly, the editing induced therapeutically significant levels of HbF to reverse the phenotypes of both sickle cell disease and β-thalassemia major. These findings imply that PRR-βE1 gene editing of patient HSPCs could lead to improved therapeutic outcomes for β-hemoglobinopathy gene therapy.
Collapse
Affiliation(s)
- Vigneshwaran Venkatesan
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
- Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Abisha Crystal Christopher
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
| | - Manuel Rhiel
- Institute for Transfusion Medicine and Gene Therapy, Medical Center – University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany
| | - Manoj Kumar K. Azhagiri
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
- Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Prathibha Babu
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
- Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Kaivalya Walavalkar
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
| | - Bharath Saravanan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Faculty of Medicine & Medical Center - University of Freiburg, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sumathi Rangaraj
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
| | - Saranya Srinivasan
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
| | - Karthik V. Karuppusamy
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
- Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Annlin Jacob
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
| | - Abhirup Bagchi
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
| | - Aswin Anand Pai
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research Center, Ibaraki 3050074, Japan
| | - Ryo Kurita
- Cell Engineering Division, RIKEN BioResource Research Center, Ibaraki 3050074, Japan
| | | | - Rekha Pai
- Department of Pathology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Srujan Kumar Marepally
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
| | | | - Shaji R. Velayudhan
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Faculty of Medicine & Medical Center - University of Freiburg, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dimple Notani
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center – University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany
| | - Alok Srivastava
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
- Department of Hematology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Saravanabhavan Thangavel
- Centre for Stem Cell Research (CSCR), A Unit of InStem Bengaluru, Christian Medical College Campus, Vellore, Tamil Nadu 632002, India
| |
Collapse
|
23
|
Gotardo ÉMF, Brito PL, Gushiken LFS, Chweih H, Leonardo FC, Costa FF, Conran N. Molecular and cellular effects of in vivo chronic intravascular hemolysis and anti-inflammatory therapeutic approaches. Vascul Pharmacol 2023; 150:107176. [PMID: 37116732 DOI: 10.1016/j.vph.2023.107176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
Intravascular hemolysis (IVH) occurs in numerous inherited and acquired disorders, including sickle cell disease (SCD), malaria and sepsis. These diseases display unique symptoms, but often share complications, such as vasomotor dysfunction and pulmonary hypertension. Consequently, in vivo models are needed to study the effects of continuous intravascular hemolytic processes, independently of the molecular or extrinsic alteration that leads to erythrocyte destruction. We gave twice-weekly low-dose phenylhydrazine (LDPHZ) to C57BL/6 J mice for 4 weeks, and measured parameters indicative of anemia, hemoglobin-clearance pathways, inflammation and iron turnover, comparing these to those of a murine model of SCD, which displays associated IVH. LDPHZ administration provoked discreet anemia in mice and significant reticulocytosis, in association with hemoglobin/heme-clearance pathway protein depletion. Mice subjected to chronic hemolysis displayed elevated leukocyte counts and plasma levels of interleukin (IL)-1β, TNF-α, IL-6, soluble ICAM-1, endothelin-1 and anti-inflammatory IL-10, closely emulating alterations indicative of systemic inflammatory and endothelial activation in SCD, and confirming chronic IVH in itself as a serious complication. Discreet accelerations in hepatic and splenic iron turnover also occurred in LDPHZ mice, without alterations in liver damage markers. Examining the effects of two therapies on hemolysis-induced inflammation, the administration of hydroxyurea (and to a lesser extent, l-glutamine) significantly abrogated hemolytic inflammation in mice, without apparent inhibition of hemolysis. In conclusion, the isolation of chronic IVH, a common disease mechanism, using this model, may allow the study of hemolysis-specific sequelae at the cellular and systemic level, and the investigation of candidate agents that could potentially counter hemolytic inflammation.
Collapse
Affiliation(s)
- Érica M F Gotardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| | - Pâmela L Brito
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Lucas F S Gushiken
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Hanan Chweih
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Flavia C Leonardo
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Fernando F Costa
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - Nicola Conran
- Hematology and Transfusion Center, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
24
|
Lins CF, Salmon CEG, Amorim de Souza L, Quesado RCS, de Souza Moraes R, Silva-Pinto AC, Matos MA, Nogueira-Barbosa MH. Quantitative MRI evaluation of bone marrow in sickle cell disease: relationship with haemolysis and clinical severity. Clin Radiol 2023; 78:e268-e278. [PMID: 36623977 DOI: 10.1016/j.crad.2022.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/06/2022] [Accepted: 11/17/2022] [Indexed: 12/27/2022]
Abstract
AIM To evaluate bone marrow fat fraction using the Dixon technique (FFDix) of magnetic resonance imaging (MRI) as a potential biomarker of haemolysis and clinical severity in the overall assessment and follow-up of sickle cell disease (SCD) patients. MATERIAL AND METHODS The present study was a cross-sectional study in which healthy individuals and SCD patients (matched for age, sex, and weight) were subjected to MRI of the lumbar spine and pelvis to quantify FFDix in the bone marrow using the Dixon technique. SCD severity was analysed by clinical and laboratory data, and an online calculator. A high degree of haemolysis was defined using the cut-off values haemoglobin (Hb) ≤10 g/dl, lactate dehydrogenase (LDH) ≥325 U/l, reticulocytes ≥3% and total bilirubin (TB) ≥1.2 mg/dl. Pearson's correlation, receiver operating characteristic (ROC) curve and binary logistic regression analysis were performed. RESULTS Forty-eight SCD patients (26 homozygous: HbSS and 22 compound heterozygous: HbSC) and 48 healthy individuals participated in the study. FFDix was lower in SCD patients than in the control group, showing even lower values in the HbSS subtype and patients with a higher degree of haemolysis. HbSC patients with a higher degree of haemolysis using hydroxyurea (medium dosage 9.8 mg/kg/day) had lower FFDix. ROC curves and odds ratios for detecting patients with a higher degree of haemolysis at the different FFDix measurement sites demonstrated excellent performance: iliac bones (cut-off ≤16.75%, AUC = 0.824, p<0.001), femoral heads (cut-off ≤46.7%, AUC = 0.775, p=0.001), lumbar vertebrae (cut-off ≤7.8%, AUC = 0.755, p=0.002). CONCLUSION Decreased FFDix is indicative of higher degree of haemolysis and SCD severity with great potential as a non-invasive biomarker contributing to the overall assessment and follow-up of SCD patients.
Collapse
Affiliation(s)
- C Freitas Lins
- Bahiana School of Medicine and Public Health (EBMSP), Av. Dom João VI, 275, Brotas, Salvador, Bahia, Brazil; Clínica Delfin Medicina Diagnóstica, Av. Antônio Carlos Magalhães, 442, Pituba, Salvador, Bahia, Brazil; Ribeirão Preto Medical School, USP Ribeirão Preto, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Campus Universitário s/n - Monte Alegre, Ribeirão Preto, SP, Brazil; Ribeirão Preto Medical School Musculoskeletal Imaging Research Laboratory, Brazil.
| | - C E Garrido Salmon
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, São Paulo, Brazil
| | - L Amorim de Souza
- Bahiana School of Medicine and Public Health (EBMSP), Av. Dom João VI, 275, Brotas, Salvador, Bahia, Brazil
| | - R C Saldanha Quesado
- Bahiana School of Medicine and Public Health (EBMSP), Av. Dom João VI, 275, Brotas, Salvador, Bahia, Brazil
| | - R de Souza Moraes
- Clínica Delfin Medicina Diagnóstica, Av. Antônio Carlos Magalhães, 442, Pituba, Salvador, Bahia, Brazil
| | - A C Silva-Pinto
- Ribeirão Preto Medical School, USP Ribeirão Preto, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Campus Universitário s/n - Monte Alegre, Ribeirão Preto, SP, Brazil
| | - M Almeida Matos
- Bahiana School of Medicine and Public Health (EBMSP), Av. Dom João VI, 275, Brotas, Salvador, Bahia, Brazil
| | - M H Nogueira-Barbosa
- Ribeirão Preto Medical School, USP Ribeirão Preto, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, Campus Universitário s/n - Monte Alegre, Ribeirão Preto, SP, Brazil; Ribeirão Preto Medical School Musculoskeletal Imaging Research Laboratory, Brazil; Department of Orthopedic Surgery, University of Missouri Health Care, Columbia, MO, USA
| |
Collapse
|
25
|
Screen "play" for drug discovery. Proc Natl Acad Sci U S A 2022; 119:e2215625119. [PMID: 36264843 PMCID: PMC9659400 DOI: 10.1073/pnas.2215625119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
26
|
Feng R, Mayuranathan T, Huang P, Doerfler PA, Li Y, Yao Y, Zhang J, Palmer LE, Mayberry K, Christakopoulos GE, Xu P, Li C, Cheng Y, Blobel GA, Simon MC, Weiss MJ. Activation of γ-globin expression by hypoxia-inducible factor 1α. Nature 2022; 610:783-790. [PMID: 36224385 PMCID: PMC9773321 DOI: 10.1038/s41586-022-05312-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 09/02/2022] [Indexed: 12/24/2022]
Abstract
Around birth, globin expression in human red blood cells (RBCs) shifts from γ-globin to β-globin, which results in fetal haemoglobin (HbF, α2γ2) being gradually replaced by adult haemoglobin (HbA, α2β2)1. This process has motivated the development of innovative approaches to treat sickle cell disease and β-thalassaemia by increasing HbF levels in postnatal RBCs2. Here we provide therapeutically relevant insights into globin gene switching obtained through a CRISPR-Cas9 screen for ubiquitin-proteasome components that regulate HbF expression. In RBC precursors, depletion of the von Hippel-Lindau (VHL) E3 ubiquitin ligase stabilized its ubiquitination target, hypoxia-inducible factor 1α (HIF1α)3,4, to induce γ-globin gene transcription. Mechanistically, HIF1α-HIF1β heterodimers bound cognate DNA elements in BGLT3, a long noncoding RNA gene located 2.7 kb downstream of the tandem γ-globin genes HBG1 and HBG2. This was followed by the recruitment of transcriptional activators, chromatin opening and increased long-range interactions between the γ-globin genes and their upstream enhancer. Similar induction of HbF occurred with hypoxia or with inhibition of prolyl hydroxylase domain enzymes that target HIF1α for ubiquitination by the VHL E3 ubiquitin ligase. Our findings link globin gene regulation with canonical hypoxia adaptation, provide a mechanism for HbF induction during stress erythropoiesis and suggest a new therapeutic approach for β-haemoglobinopathies.
Collapse
Affiliation(s)
- Ruopeng Feng
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Peng Huang
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip A Doerfler
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Yichao Li
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Yu Yao
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Jingjing Zhang
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lance E Palmer
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Kalin Mayberry
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Peng Xu
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Chunliang Li
- Department of Tumor Cell Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Yong Cheng
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Gerd A Blobel
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mitchell J Weiss
- Department of Hematology, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
27
|
Venugopal J, Wang J, Guo C, Eitzman DT. Amiodarone improves anemia in a murine model of sickle cell disease and is associated with increased erythrocyte bis(monoacylglycerol) phosphate. Sci Rep 2022; 12:16437. [PMID: 36180774 PMCID: PMC9525675 DOI: 10.1038/s41598-022-20955-5] [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: 03/11/2022] [Accepted: 09/21/2022] [Indexed: 11/26/2022] Open
Abstract
Sickle cell disease (SCD) is associated with altered plasma and erythrocyte lipid profiles. In a previous study, SCD mice with deficiency of proprotein convertase subtilisin/kexin type 9 (PCSK9) were observed to have more severe anemia and increased sickling compared to control SCD mice. Although PCSK9 affects circulating low density lipoprotein (LDL) by regulation of the LDL receptor, the effect of PCSK9 on anemia was independent of LDL receptor expression. In the current study, erythrocyte metabolomics were performed and revealed altered erythrocyte lipid species between SCD mice with and without PCSK9. Of particular interest, the late endosome-specific lipid bis(mono)acylglycerol phosphate (BMP) 44:12 was markedly decreased in erythrocytes from SCD mice deficient in PCSK9 mice relative to control SCD mice. Incubation of sickle erythrocytes with a neutralizing antibody to BMP increased erythrocyte sickling in vitro. In vitro treatment of SCD erythrocytes with amiodarone (1.5 μM) or medroxyprogesterone (6.75 μM), two pharmacologic compounds known to increase BMP, resulted in reduced erythrocyte sickling. Treatment of SCD mice with amiodarone (10 mg/kg) for 2 weeks resulted in increased BMP, improvement in anemia with reduced reticulocytosis, and decreased ex vivo sickling. In conclusion, severity of anemia in SCD is improved with amiodarone treatment, an effect which may be mediated through increased erythrocyte BMP.
Collapse
Affiliation(s)
- Jessica Venugopal
- Cardiovascular Research Center, University of Michigan Internal Medicine-Cardiology Division, 7301A MSRB III, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-0644, USA
| | - Jintao Wang
- Cardiovascular Research Center, University of Michigan Internal Medicine-Cardiology Division, 7301A MSRB III, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-0644, USA
| | - Chiao Guo
- Cardiovascular Research Center, University of Michigan Internal Medicine-Cardiology Division, 7301A MSRB III, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-0644, USA
| | - Daniel T Eitzman
- Cardiovascular Research Center, University of Michigan Internal Medicine-Cardiology Division, 7301A MSRB III, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-0644, USA.
| |
Collapse
|
28
|
Abstract
Sickle cell disease (SCD) is characterized by variable clinical outcomes, with some patients suffering life-threatening complications during childhood, and others living relatively symptom-free into old age. Because of this variability, there is an important potential role for precision medicine, in which particular different treatments are selected for different groups of patients. However, the application of precision medicine in SCD is limited by difficulties in identifying different prognostic groups and the small number of available treatments. The main genetic determinant of outcomes in SCD is the underlying β-globin genotype, with sickle cell anemia (HbSS) and hemoglobin SC disease (HbSC) forming the 2 major forms of the disease in most populations of African origin. Although there are clear differences in clinical outcomes between these conditions, treatments approaches are very similar, with little evidence on how to treat HbSC in particular. Other genomic information, such as the co-inheritance of α-thalassemia, or high fetal hemoglobin (HbF) levels, is of some prognostic value but insufficient to determine treatments. Precision medicine is further limited by the fact that the 2 main drugs used in SCD, penicillin and hydroxyurea, are currently recommended for all patients. Newer treatments, such as crizanlizumab and voxelotor, raise the possibility that groups will emerge who respond best to particular drugs or combinations. Perhaps the best current example of precision medicine in SCD is the selective use of blood transfusions as primary stroke prevention in children with evidence of cerebral vasculopathy. More precise treatments may emerge as we understand more about the pathology of SCD, including problems with erythropoiesis.
Collapse
|
29
|
Meloni A, Barbuto L, Pistoia L, Positano V, Renne S, Peritore G, Fina P, Spasiano A, Allò M, Messina G, Casini T, Massa A, Romano L, Pepe A, Cademartiri F. Frequency, pattern, and associations of renal iron accumulation in sickle/β-thalassemia patients. Ann Hematol 2022; 101:1941-1950. [PMID: 35821343 DOI: 10.1007/s00277-022-04915-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022]
Abstract
We evaluated frequency, pattern, and associations of renal iron accumulation in sickle/β-thalassemia. Thirty-three sickle/β-thalassemia patients (36.5 ± 14.7 years; 13 females), 14 homozygous sickle cell disease (SCD) patients, and 71 thalassemia major (TM) patients, enrolled in the E-MIOT Network, underwent magnetic resonance imaging. Iron overload (IO) was quantified by the T2* technique. Sickle/β-thalassemia patients had a significantly lower frequency of renal IO (T2* < 31 ms) than homozygous SCD patients (9.1% vs. 57.1%; P = 0.001), besides having similar hepatic, cardiac and pancreatic IO. Kidney T2* values were comparable between regularly transfused sickle/β-thalassemia and TM patients but were significantly lower in regularly transfused homozygous SCD patients than in the other two groups. In sickle/β-thalassemia patients, global renal T2* values were not associated with age, gender, splenectomy, and presence of regular transfusions or chelation. No correlation was detected between renal T2* values and serum ferritin levels or iron load in the other organs. Global renal T2* values were not associated with serum creatinine levels but showed a significant inverse correlation with serum lactate dehydrogenase (R = - 0.709; P < 0.0001) and indirect bilirubin (R = - 0.462; P = 0.012). Renal IO is not common in sickle/β-thalassemia patients, with a prevalence significantly lower compared to that of homozygous SCD patients, but with a similar underlying mechanism due to the chronic hemolysis.
Collapse
Affiliation(s)
- Antonella Meloni
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy.,U.O.C. Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Luigi Barbuto
- U.O.C. Radiologia Generale E Di Pronto Soccorso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Naples, Italy
| | - Laura Pistoia
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy
| | - Vincenzo Positano
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy.,U.O.C. Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Stefania Renne
- Struttura Complessa Di Cardioradiologia-UTIC, Presidio Ospedaliero "Giovanni Paolo II", Lamezia Terme, Italy
| | - Giuseppe Peritore
- Unità Operativa Complessa Di Radiologia, "ARNAS" Civico, Di Cristina Benfratelli, Palermo, Italy
| | - Priscilla Fina
- Unità Operativa Complessa Diagnostica Per Immagini, Ospedale "Sandro Pertini", Rome, Italy
| | - Anna Spasiano
- U.O.S.D. Malattie Rare del Globulo Rosso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Naples, Italy
| | - Massimo Allò
- Ematologia Microcitemia, Ospedale San Giovanni di Dio - ASP Crotone, Crotone, Italy
| | - Giuseppe Messina
- Centro Microcitemie, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Tommaso Casini
- Centro Talassemie Ed Emoglobinopatie, Ospedale "Meyer", Florence, Italy
| | - Antonella Massa
- Servizio Trasfusionale, Ospedale "Giovanni Paolo II", Olbia, Italy
| | - Luigia Romano
- U.O.C. Radiologia Generale E Di Pronto Soccorso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Naples, Italy
| | - Alessia Pepe
- Institute of Radiology, Department of Medicine, University of Padua, Padua, Italy
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy.
| |
Collapse
|
30
|
Prime Editor 3 Mediated Beta-Thalassemia Mutations of the HBB Gene in Human Erythroid Progenitor Cells. Int J Mol Sci 2022; 23:ijms23095002. [PMID: 35563395 PMCID: PMC9099916 DOI: 10.3390/ijms23095002] [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: 03/23/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/17/2022] Open
Abstract
Recently developed Prime Editor 3 (PE3) has been implemented to induce genome editing in various cell types but has not been proven in human hematopoietic stem and progenitor cells. Using PE3, we successfully installed the beta-thalassemia (beta-thal) mutations in the HBB gene in the erythroid progenitor cell line HUDEP-2. We inserted the mCherry reporter gene cassette into editing plasmids, each including the prime editing guide RNA (pegRNA) and nick sgRNA. The plasmids were electroporated into HUDEP-2 cells, and the PE3 modified cells were identified by mCherry expression and collected using fluorescence-activated cell sorting (FACS). Sanger sequencing of the positive cells confirmed that PE3 induced precise beta-thal mutations with editing ratios from 4.55 to 100%. Furthermore, an off-target analysis showed no unintentional edits occurred in the cells. The editing ratios and parameters of pegRNA and nick sgRNA were also analyzed and summarized and will contribute to enhanced PE3 design in future studies. The characterization of the HUDEP-2 beta-thal cells showed typical thalassemia phenotypes, involving ineffective erythropoiesis, abnormal erythroid differentiation, high apoptosis rate, defective alpha-globin colocalization, cell viability deterioration, and ROS resisting deficiency. These HUDEP-2 beta-thal cells could provide ideal models for future beta-thal gene therapy studies.
Collapse
|
31
|
Ribeil J. Primary myelofibrosis in untreated sickle cell disease: Are adult patients at higher risk for developing hematological myeloid neoplasms? Am J Hematol 2022; 97:4-6. [PMID: 34626435 DOI: 10.1002/ajh.26371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Jean‐Antoine Ribeil
- Section of Hematology/Oncology, Boston University and Boston Medical Center; Sickle Cell Center, Boston University School of Medicine Boston Massachusetts USA
| |
Collapse
|
32
|
Pincez T, Lee SSK, Ilboudo Y, Preuss M, Pham Hung d'Alexandry d'Orengiani AL, Bartolucci P, Galactéros F, Joly P, Bauer DE, Loos RJF, Lindsley RC, Lettre G. Clonal hematopoiesis in sickle cell disease. Blood 2021; 138:2148-2152. [PMID: 34521115 PMCID: PMC8617438 DOI: 10.1182/blood.2021011121] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 08/20/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Thomas Pincez
- Montreal Heart Institute, Montréal, QC, Canada
- Faculté de Médecine and
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Charles-Bruneau Cancer Center, Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Simon S K Lee
- The Charles Bronfman Institute for Personalized Medicine, Icahn Mount Sinai School of Medicine, New York, NY
| | - Yann Ilboudo
- Montreal Heart Institute, Montréal, QC, Canada
- Faculté de Médecine and
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn Mount Sinai School of Medicine, New York, NY
| | - Anne-Laure Pham Hung d'Alexandry d'Orengiani
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est, Institut Mondor de Recherche Biomédicale (IMRB), Unité 955, Equipe 2, Créteil, France
| | - Pablo Bartolucci
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est, Institut Mondor de Recherche Biomédicale (IMRB), Unité 955, Equipe 2, Créteil, France
| | - Frédéric Galactéros
- Red Cell Genetic Disease Unit, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est, Institut Mondor de Recherche Biomédicale (IMRB), Unité 955, Equipe 2, Créteil, France
| | - Philippe Joly
- Unité Fonctionnelle 34445 "Biochimie des Pathologies Érythrocytaires,' Laboratoire de Biochimie et Biologie Moléculaire Grand Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
- Laboratoire Inter-Universitaire de Biologie de la Motricité EA7424, Equipe "Biologie Vasculaire et du Globule Rouge," Université Claude Bernard Lyon 1, Comité d'Universités et d'Etablissements, Lyon, France
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Department of Pediatrics, Harvard Medical School, Boston, MA; and
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn Mount Sinai School of Medicine, New York, NY
| | - R Coleman Lindsley
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Guillaume Lettre
- Montreal Heart Institute, Montréal, QC, Canada
- Faculté de Médecine and
| |
Collapse
|
33
|
Brewin J, El Hoss S, Strouboulis J, Rees D. A novel index to evaluate ineffective erythropoiesis in hematological diseases offers insights into sickle cell disease. Haematologica 2021; 107:338-341. [PMID: 34670360 PMCID: PMC8719095 DOI: 10.3324/haematol.2021.279623] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Indexed: 11/09/2022] Open
Abstract
Not available.
Collapse
Affiliation(s)
- John Brewin
- Molecular Haematology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom; Department of Haematological Medicine, King's College Hospital, London, United Kingdom
| | - Sara El Hoss
- Molecular Haematology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom.
| | - John Strouboulis
- Molecular Haematology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom
| | - David Rees
- Molecular Haematology, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom; Department of Haematological Medicine, King's College Hospital, London, United Kingdom
| |
Collapse
|
34
|
Ineffective erythropoiesis in sickle cell disease: new insights and future implications. Curr Opin Hematol 2021; 28:171-176. [PMID: 33631786 DOI: 10.1097/moh.0000000000000642] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW Sickle cell disease (SCD) is a hemolytic anemia caused by a point mutation in the β globin gene leading to the expression of an abnormal hemoglobin (HbS) that polymerizes under hypoxic conditions driving red cell sickling. Circulating red cells have been extensively characterized in SCD, as their destruction and removal from peripheral blood are the major contributors to anemia. However, few reports showed cellular abnormalities during erythropoiesis in SCD, suggesting that anemia could also be influenced by defects of central origin. RECENT FINDINGS El Hoss et al. demonstrated ineffective erythropoiesis (IE) in SCD and deciphered the molecular mechanism underlying cell death during the hemoglobin synthesis phase of terminal differentiation. They showed that HbS polymerization induces apoptosis of differentiating erythroblasts and that fetal hemoglobin rescues these cells through its antipolymerization function. SUMMARY IE is the major cause of anemia in β-thalassemia patients, and it is generally surmised that it contributes little to anemia of SCD. Recent reports demonstrate the occurrence of IE in SCD patients and show important alterations in the hematopoietic and erythroid niches, both in SCD patients and in the humanized Townes SCD mouse model. This implies that therapeutic strategies initially designed to improve red cell survival in the circulation of SCD patients would also positively impact erythropoiesis and bone marrow cellularity.
Collapse
|
35
|
Vasseur C, Domingues-Hamdi E, Pakdaman S, Galactéros F, Baudin-Creuza V. Alpha haemoglobin-stabilising protein concentration in the red blood cells of patients with sickle cell anaemia with and without hydroxycarbamide treatment. Br J Haematol 2021; 196:183-192. [PMID: 34378186 DOI: 10.1111/bjh.17728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/02/2021] [Accepted: 07/12/2021] [Indexed: 11/27/2022]
Abstract
Alpha haemoglobin-stabilising protein (AHSP) is a key chaperone synthesised in red blood cell (RBC) precursors. Many studies have reported AHSP as a potential biomarker of various diseases. AHSP gene expression has been studied in detail, but little is known about AHSP protein levels in RBCs. We investigated the AHSP concentration of RBC lysates from control subjects (n = 10) and patients with sickle cell anaemia (SCA) with (n = 10) and without (n = 12) hydroxycarbamide (HC) treatment, to evaluate the clinical relevance of AHSP in SCA. We developed a sandwich enzyme-linked immunosorbent assay method, with which we were able, for the first time, to determine the mean AHSP concentration in control RBC lysates (0·82 µg/ml). The AHSP concentration (2·23 µg/ml) was significantly higher in untreated patients with the SS genotype than in controls. The AHSP concentration decreased significantly on HC treatment (1·50 µg/ml) but remained significantly higher than that in controls. A strong positive correlation was observed between the AHSP concentration and the α-haemoglobin pool with the three groups of subjects pooled into a single group. Our present findings indicate that AHSP concentration can be considered a candidate biomarker for monitoring HC responses in patients with SCA and suggest a role for AHSP in various RBC diseases.
Collapse
Affiliation(s)
- Corinne Vasseur
- Inserm U955-IMRB Team Pirenne, Univ Paris Est Créteil, Créteil, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Elisa Domingues-Hamdi
- Inserm U955-IMRB Team Pirenne, Univ Paris Est Créteil, Créteil, France.,Laboratory of Excellence GR-Ex, Paris, France
| | - Sadaf Pakdaman
- Inserm U955-IMRB Team Pirenne, Univ Paris Est Créteil, Créteil, France.,Laboratory of Excellence GR-Ex, Paris, France.,Etablissement Français du Sang (EFS) Ile de France-Mondor, Créteil, France
| | - Frédéric Galactéros
- Inserm U955-IMRB Team Pirenne, Univ Paris Est Créteil, Créteil, France.,Laboratory of Excellence GR-Ex, Paris, France.,Unité des Maladies Génétiques du Globule Rouge, Hôpital Universitaire Henri Mondor (AP-HP), Créteil, France
| | - Véronique Baudin-Creuza
- Inserm U955-IMRB Team Pirenne, Univ Paris Est Créteil, Créteil, France.,Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|