1
|
Yang H, Han S, Xu J, He S, Lu Q, Luo T, Chen S, Dang L, Wang G, Li J, Huang M, Liao Y, He Y, Cai N, Huang L, Zhou M, Mo Y, Zhu W, Wu Z, Zhou G, Chen J. Effects of Thalidomide on Metabolism and Lifespan of Red Blood Cell in Patients With β-Thalassemia Major: A Post Hoc Analysis of a Randomized Controlled Trial. Clin Ther 2025; 47:252-260. [PMID: 39971661 DOI: 10.1016/j.clinthera.2025.01.008] [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: 08/12/2024] [Revised: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
PURPOSE Recent studies have shown the thalidomide's therapeutic potential in treatment of patients with β-thalassemia major. However, the effect of thalidomide on metabolism and lifespan of red blood cells (RBCs) is rarely reported. METHODS This study was a post hoc analysis of a randomized controlled trial (Chinese Clinical Trial Registry, ChiCTR1800015702). One hundred patients with β-thalassemia major were randomly assigned 1:1 to treatment with a placebo or thalidomide. The primary outcomes were the differences in RBC lifespan, reticulocyte count, and peripheral nucleated RBC count of patients after treatment of 12 weeks. Other indicators of hemolytic reaction were also analyzed. FINDINGS Compared with the placebo group after treatment of 12 weeks, the thalidomide group showed a longer RBC lifespan (16.29 ± 6.42 vs 12.90 ± 4.98 days; P = 0.004), smaller mean corpuscular volume (68.34 ± 7.79 vs 78.01 ± 6.33 fl; P < 0.001), smaller mean corpuscular hemoglobin (21.62 ± 2.85 vs 24.68 ± 2.69 pg; P < 0.001), and lower lactate dehydrogenase (190.00 [148.00 - 305.00] vs 251.00 [199.20 - 327.80]; P = 0.014). Meanwhile, thalidomide significantly increased the RBC lifespan at 24 weeks (21.24 ± 8.30 days; P < 0.001) and 48 weeks (23.21 ± 8.42 days; P < 0.001) when compared with baseline (12.8 ± 6.0 days). IMPLICATIONS Thalidomide increases the RBC lifespan and reduces hemolytic reactions in patients with β-thalassemia major. Chinese Clinical Trial Registry identifier: ChiCTR1800015702.
Collapse
Affiliation(s)
- Huanju Yang
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Sichong Han
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianquan Xu
- Department of Hematology, Yulin Guinan Hospital, Yulin, Guangxi, China
| | - Sheng He
- Guangxi Key Laboratory of Basic Research on Birth Defects Prevention and Treatment, Guangxi Zhuang Autonomous Region Women and Children Health Care Hospital, Nanning, Guangxi, China
| | - Qiyang Lu
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Tianying Luo
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Shuying Chen
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Lujie Dang
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Guizhen Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinyan Li
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Minjie Huang
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Yangdong Liao
- Department of Blood Transfusion, Wuzhou Gongren Hospital, Wuzhou, Guangxi, China
| | - Yanfang He
- Wuzhou Central Blood Station, Wuzhou, Guangxi, China
| | - Ning Cai
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Lan Huang
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Meiguang Zhou
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Yongquan Mo
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China
| | - Weijian Zhu
- Department of Hematology, Zhuhai People's Hospital, Zhuhai, Guangdong, China
| | - Zhengwei Wu
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China.
| | - Guangbiao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jiangming Chen
- Department of Hematology, Wuzhou Gongren Hospital (The Seventh Clinical Medical College of Guangxi Medical University), Wuzhou, Guangxi, China.
| |
Collapse
|
2
|
Han Y, Gudmundsdottir B, Gudmundsson KO, Roy KR, Tisdale J, Du Y. MLL1 complex is a critical regulator of fetal hemoglobin repression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645036. [PMID: 40196665 PMCID: PMC11974897 DOI: 10.1101/2025.03.24.645036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Increasing fetal-type hemoglobin (HbF) expression in adult erythroid cells holds promise in the treatment of sickle cell disease (SCD) and β-thalassemia. We have identified MLL1 complex as a critical regulator of fetal and embryonic hemoglobin repression. Knockdowns of MEN1 and KMT2A, encoding essential components of the complex, caused a significant downregulation of BCL11A expression and a substantial increase in γ- and ε-globin mRNA levels in HUDEP-2 cells. Significant binding of MEN1 and KMT2A were readily detected at the promoter and a critical enhancer of BCL11A in HUDEP-2 cells, suggesting that BCL11A is a direct transcriptional target of MLL1 complex. Consistent with these results, MEN1 or KMT2A knockdown in normal human CD34 + hematopoietic stem and progenitor cells (HSPCs) induced to undergo erythroid differentiation also significantly decreased their BCL11A expression and increased their γ- and ε-globin expression and the production of F cells in the culture. Treatment of these cells with MENIN inhibitors yielded similar results and promoted erythroid differentiation with minimal effects on their growth. These findings underscore a critical role of MLL1 complex in regulating fetal and embryonic hemoglobin expression and suggest that MENIN inhibitors could offer a promising therapeutic approach for sickle cell disease and β-thalassemia.
Collapse
|
3
|
Shah AP, Majeti KR, Ekman FK, Selvaraj S, Sharma D, Sinha R, Soupene E, Chati P, Luna SE, Charlesworth CT, McCreary T, Lesch BJ, Tran T, Chu SN, Porteus MH, Kyle Cromer M. Engineering synthetic signaling receptors to enable erythropoietin-free erythropoiesis. Nat Commun 2025; 16:1140. [PMID: 39880867 PMCID: PMC11779867 DOI: 10.1038/s41467-025-56239-5] [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: 10/18/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025] Open
Abstract
Blood transfusion plays a vital role in modern medicine, but frequent shortages occur. Ex vivo manufacturing of red blood cells (RBCs) from universal donor cells offers a potential solution, yet the high cost of recombinant cytokines remains a barrier. Erythropoietin (EPO) signaling is crucial for RBC development, and EPO is among the most expensive media components. To address this challenge, we develop highly optimized small molecule-inducible synthetic EPO receptors (synEPORs) using design-build-test cycles and genome editing. By integrating synEPOR at the endogenous EPOR locus in O-negative induced pluripotent stem cells, we achieve equivalent erythroid differentiation, transcriptomic changes, and hemoglobin production using small molecules compared to EPO-supplemented cultures. This approach dramatically reduces culture media costs. Our strategy not only addresses RBC production challenges but also demonstrates how protein and genome engineering can introduce precisely regulated cellular behaviors, potentially improving scalable manufacturing of a wide range of clinically relevant cell types.
Collapse
Affiliation(s)
- Aadit P Shah
- School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Kiran R Majeti
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Freja K Ekman
- School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Sridhar Selvaraj
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Devesh Sharma
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Roshani Sinha
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eric Soupene
- Benioff Children's Hospital Oakland, University of California, San Francisco, San Francisco, CA, USA
| | - Prathamesh Chati
- Department of Biological & Medical Informatics, University of California, San Francisco, San Francisco, CA, USA
| | - Sofia E Luna
- School of Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | | - Travis McCreary
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin J Lesch
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Tammy Tran
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Simon N Chu
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - M Kyle Cromer
- Department of Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Bioengineering & Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Chu SN, Soupene E, Sharma D, Sinha R, McCreary T, Hernandez B, Shen H, Wienert B, Bowman C, Yin H, Lesch BJ, Jia K, Romero KA, Kostamo Z, Zhang Y, Tran T, Cordero M, Homma S, Hampton JP, Gardner JM, Conklin BR, MacKenzie TC, Sheehan VA, Porteus MH, Cromer MK. Dual α-globin-truncated erythropoietin receptor knockin restores hemoglobin production in α-thalassemia-derived erythroid cells. Cell Rep 2025; 44:115141. [PMID: 39754719 PMCID: PMC11837859 DOI: 10.1016/j.celrep.2024.115141] [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: 01/11/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 01/06/2025] Open
Abstract
The most severe form of α-thalassemia results from loss of all four copies of α-globin. Postnatally, patients face challenges similar to β-thalassemia, including severe anemia and erythrotoxicity due to the imbalance of β-globin and α-globin chains. Despite progress in genome editing treatments for β-thalassemia, there is no analogous curative option for α-thalassemia. To address this, we designed a Cas9/AAV6-mediated genome editing strategy that integrates a functional α-globin gene into the β-globin locus in α-thalassemia patient-derived hematopoietic stem and progenitor cells (HSPCs). Incorporation of a truncated erythropoietin receptor transgene into the α-globin integration cassette significantly increased erythropoietic output from edited HSPCs and led to the most robust production of α-globin, and consequently hemoglobin tetramers. By directing edited HSPCs toward increased production of clinically relevant erythroid cells, this approach has the potential to mitigate the limitations of current treatments for the hemoglobinopathies, including low genome editing and low engraftment rates.
Collapse
Affiliation(s)
- Simon N Chu
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric Soupene
- Department of Pediatrics, University of California, San Francisco, Oakland, CA 94609, USA
| | - Devesh Sharma
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Roshani Sinha
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Travis McCreary
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Britney Hernandez
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Huifeng Shen
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30329, USA
| | | | - Chance Bowman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering & Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Han Yin
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Benjamin J Lesch
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering & Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kun Jia
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kathleen A Romero
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Zachary Kostamo
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Yankai Zhang
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Tammy Tran
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marco Cordero
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shota Homma
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Jessica P Hampton
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - James M Gardner
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bruce R Conklin
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tippi C MacKenzie
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vivien A Sheehan
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - M Kyle Cromer
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering & Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
5
|
Ghosh A, Coffin M, Diaz DM, Barndt S, Schulz V, Gallagher P, Lo SH, Fowler VM. A novel isoform of Tensin1 promotes actin filament assembly for efficient erythroblast enucleation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628322. [PMID: 39763869 PMCID: PMC11702514 DOI: 10.1101/2024.12.13.628322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Mammalian red blood cells are generated via a terminal erythroid differentiation pathway culminating in cell polarization and enucleation. Actin filament polymerization is critical for enucleation, but the molecular regulatory mechanisms remain poorly understood. We utilized publicly available RNA-seq and proteomics datasets to mine for actin-binding proteins and actin-nucleation factors differentially expressed during human erythroid differentiation and discovered that a focal adhesion protein-Tensin-1-dramatically increases in expression late in differentiation. Remarkably, we found that differentiating human CD34+ cells express a novel truncated form of Tensin-1 (eTNS1; Mr ~125 kDa) missing the N-terminal half of the protein, due to an internal mRNA translation start site resulting in a unique exon 1. eTNS1 localized to the cytoplasm during terminal erythroid differentiation, with no apparent membrane association or focal adhesion formation. Knocking out eTNS1 had no effect on assembly of the spectrin membrane skeleton but led to impaired enucleation and absent or mis-localized actin filament foci in enucleating erythroblasts. We conclude that eTNS1 is a novel regulator of actin filament assembly during human erythroid terminal differentiation required for efficient enucleation.
Collapse
Affiliation(s)
- Arit Ghosh
- Department of Biological Sciences, University of Delaware, Newark, DE
- Delaware Biotechnology Institute, UD Flow Cytometry Core, Newark, DE
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE
| | - Megan Coffin
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Dimitri M. Diaz
- Department of Biological Sciences, University of Delaware, Newark, DE
| | - Sarah Barndt
- Department of Medical and Molecular Sciences, University of Delaware, Newark, DE
| | - Vincent Schulz
- Department of Pediatrics, Yale University, New Haven, CT
| | | | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, CA 95817, USA
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Molecular and Genomic Medicine, National Health of Research Institutes, Miaoli 35053, Taiwan
| | - Velia M. Fowler
- Department of Biological Sciences, University of Delaware, Newark, DE
| |
Collapse
|
6
|
Luna SE, Camarena J, Hampton JP, Majeti KR, Charlesworth CT, Soupene E, Selvaraj S, Jia K, Sheehan VA, Cromer MK, Porteus MH. Enhancement of erythropoietic output by Cas9-mediated insertion of a natural variant in haematopoietic stem and progenitor cells. Nat Biomed Eng 2024; 8:1540-1552. [PMID: 38886504 PMCID: PMC11668683 DOI: 10.1038/s41551-024-01222-6] [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: 08/17/2023] [Accepted: 05/02/2024] [Indexed: 06/20/2024]
Abstract
Some gene polymorphisms can lead to monogenic diseases, whereas other polymorphisms may confer beneficial traits. A well-characterized example is congenital erythrocytosis-the non-pathogenic hyper-production of red blood cells-that is caused by a truncated erythropoietin receptor. Here we show that Cas9-mediated genome editing in CD34+ human haematopoietic stem and progenitor cells (HSPCs) can recreate the truncated form of the erythropoietin receptor, leading to substantial increases in erythropoietic output. We also show that combining the expression of the cDNA of a truncated erythropoietin receptor with a previously reported genome-editing strategy to fully replace the HBA1 gene with an HBB transgene in HSPCs (to restore normal haemoglobin production in cells with a β-thalassaemia phenotype) gives the edited HSPCs and the healthy red blood cell phenotype a proliferative advantage. Combining knowledge of human genetics with precise genome editing to insert natural human variants into therapeutic cells may facilitate safer and more effective genome-editing therapies for patients with genetic diseases.
Collapse
Affiliation(s)
- Sofia E Luna
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Joab Camarena
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Jessica P Hampton
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Kiran R Majeti
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Carsten T Charlesworth
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Eric Soupene
- Department of Pediatrics, University of California, San Francisco, Oakland, CA, USA
| | - Sridhar Selvaraj
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Kun Jia
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Vivien A Sheehan
- Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - M Kyle Cromer
- Department of Surgery, University of California San Francisco, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, USA.
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
7
|
Ala C, Joshi RP, Gupta P, Goswami SG, Ramalingam S, Kondapalli Venkata Gowri CS, Sankaranarayanan M. A critical review of therapeutic interventions in sickle cell disease: Progress and challenges. Arch Pharm (Weinheim) 2024; 357:e2400381. [PMID: 39031925 DOI: 10.1002/ardp.202400381] [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: 05/16/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/22/2024]
Abstract
Sickle cell disease (SCD) is an autosomal recessive genetic disorder that occurs due to the point mutation in the β-globin gene, which results in the formation of sickle hemoglobin (HbS) in the red blood cells (RBCs). When HbS is exposed to an oxygen-depleted environment, it polymerizes, resulting in hemolysis, vaso-occlusion pain, and impaired blood flow. Still, there is no affordable cure for this inherited disease. Approved medications held promise but were met with challenges due to limited patient tolerance and undesired side effects, thereby inhibiting their ability to enhance the quality of life across various individuals with SCD. Progress has been made in understanding the pathophysiology of SCD during the past few decades, leading to the discovery of novel targets and therapies. However, there is a compelling need for research to discover medications with improved efficacy and reduced side effects. Also, more clinical investigations on various drug combinations with different mechanisms of action are needed. This review comprehensively presents therapeutic approaches for SCD, including those currently available or under investigation. It covers fundamental aspects of the disease, such as epidemiology and pathophysiology, and provides detailed discussions on various disease-modifying agents. Additionally, expert insights are offered on the future development of pharmacotherapy for SCD.
Collapse
Affiliation(s)
- Chandu Ala
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Medicinal Chemistry Research Laboratory, Pilani Campus, Pilani, Rajasthan, India
| | - Renuka Parshuram Joshi
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Medicinal Chemistry Research Laboratory, Pilani Campus, Pilani, Rajasthan, India
| | - Pragya Gupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | | | | | | | - Murugesan Sankaranarayanan
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Medicinal Chemistry Research Laboratory, Pilani Campus, Pilani, Rajasthan, India
| |
Collapse
|
8
|
Ju W, Din G, Huang J, Zheng M, Wang X, Liu L, Wang L, Xuan S, Xiao W, Chen A. Efficacy and safety of thalidomide in patients with β-thalassemia intermedia and major. Medicine (Baltimore) 2024; 103:e40328. [PMID: 39470504 PMCID: PMC11521010 DOI: 10.1097/md.0000000000040328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
To investigate the short-term and long-term efficacy and safety of thalidomide in the treatment of intermediate and severe β-thalassemia. We analyzed patients with intermediate and severe β-thalassemia treated at our hospital from February 2019 to February 2024. Patients who received treatment for more than 3 months were included. The efficacy of thalidomide was assessed by comparing changes in hemoglobin (Hb), ferritin, bilirubin, and Hb electrophoresis before and after treatment. Adverse drug reactions during treatment were also recorded. A total of 42 β-thalassemia patients were included, with thalidomide dosages ranging from 75 to 150 mg/d. The response rates at 1, 3, and 6 months of treatment were 73.8% (31/42), 75.0% (24/32), and 94.7% (18/19), respectively. The increase in Hb levels was primarily attributed to fetal hemoglobin (HbF). After 1 month of treatment, the HbF percentage increased from a baseline of 34.04 ± 27.58% to 56.25 ± 28.40% (P < .001). At 3 and 6 months, HbF further increased to 67.21 ± 27.12% (P < .001) and 73.93 ± 22.96% (P < .001), respectively. The average duration of thalidomide treatment was 25.3 ± 9.2 months (range: 4-60 months), with 6 patients treated for over 60 months and 18 patients for over 48 months. Two homozygous patients who failed thalidomide treatment achieved Hb levels above 100 g/L and discontinued transfusion therapy after 3 months of hydroxyurea treatment. The most common adverse reaction was somnolence, which was mild and tolerable. Thalidomide demonstrates significant and sustained therapeutic effects in β-thalassemia patients. The adverse reactions are mild and tolerable, allowing patients to continue treatment.
Collapse
Affiliation(s)
- Wendong Ju
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Gaomin Din
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Jun Huang
- Department of Electrocardiology, Zhongshan Boai Hospital, Zhongshan, China
| | - Minmin Zheng
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Xiaoyou Wang
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Lingling Liu
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Li Wang
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Suling Xuan
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Weiqun Xiao
- Department of Hematology Rheumatology, Zhongshan Boai Hospital, Zhongshan, China
| | - Ang Chen
- Department of Education Section, Zhongshan Boai Hospital, Zhongshan, China
| |
Collapse
|
9
|
Chu SN, Soupene E, Wienert B, Yin H, Sharma D, McCreary T, Jia K, Homma S, Hampton JP, Gardner JM, Conklin BR, MacKenzie TC, Porteus MH, Cromer MK. Dual α-globin and truncated EPO receptor knockin restores hemoglobin production in α-thalassemia-derived red blood cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.01.555926. [PMID: 38766216 PMCID: PMC11100611 DOI: 10.1101/2023.09.01.555926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Alpha-thalassemia is an autosomal recessive disease with increasing worldwide prevalence. The molecular basis is due to mutation or deletion of one or more duplicated α-globin genes, and disease severity is directly related to the number of allelic copies compromised. The most severe form, α-thalassemia major (αTM), results from loss of all four copies of α-globin and has historically resulted in fatality in utero. However, in utero transfusions now enable survival to birth. Postnatally, patients face challenges similar to β-thalassemia, including severe anemia and erythrotoxicity due to imbalance of β-globin and α-globin chains. While curative, hematopoietic stem cell transplantation (HSCT) is limited by donor availability and potential transplant-related complications. Despite progress in genome editing treatments for β-thalassemia, there is no analogous curative option for patients suffering from α-thalassemia. To address this, we designed a novel Cas9/AAV6-mediated genome editing strategy that integrates a functional α-globin gene into the β-globin locus in αTM patient-derived hematopoietic stem and progenitor cells (HSPCs). Incorporation of a truncated erythropoietin receptor transgene into the α-globin integration cassette dramatically increased erythropoietic output from edited HSPCs and led to the most robust production of α-globin, and consequently normal hemoglobin. By directing edited HSPCs toward increased production of clinically relevant RBCs instead of other divergent cell types, this approach has the potential to mitigate the limitations of traditional HSCT for the hemoglobinopathies, including low genome editing and low engraftment rates. These findings support development of a definitive ex vivo autologous genome editing strategy that may be curative for α-thalassemia.
Collapse
Affiliation(s)
- Simon N. Chu
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Eric Soupene
- Department of Pediatrics, University of California, San Francisco, Oakland, CA
| | | | - Han Yin
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Devesh Sharma
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA
| | - Travis McCreary
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA
| | - Kun Jia
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA
| | - Shota Homma
- Department of Genetics, Stanford University, Stanford, CA
| | | | - James M. Gardner
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Bruce R. Conklin
- Gladstone Institutes, San Francisco, CA
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| | - Tippi C. MacKenzie
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA
| | | | - M. Kyle Cromer
- Department of Surgery, University of California, San Francisco, San Francisco, CA
- Eli & Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA
- Department of Bioengineering & Therapeutic Sciences, University of California, San Francisco,San Francisco, CA
| |
Collapse
|
10
|
Papoin J, Yan H, Leduc M, Gall ML, Narla A, Palis J, Steiner LA, Gallagher PG, Hillyer CD, Gautier EF, Mohandas N, Blanc L. Phenotypic and proteomic characterization of the human erythroid progenitor continuum reveal dynamic changes in cell cycle and in metabolic pathways. Am J Hematol 2024; 99:99-112. [PMID: 37929634 PMCID: PMC10877306 DOI: 10.1002/ajh.27145] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/03/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023]
Abstract
Human erythropoiesis is a complex process leading to the production of 2.5 million red blood cells per second. Following commitment of hematopoietic stem cells to the erythroid lineage, this process can be divided into three distinct stages: erythroid progenitor differentiation, terminal erythropoiesis, and reticulocyte maturation. We recently resolved the heterogeneity of erythroid progenitors into four different subpopulations termed EP1-EP4. Here, we characterized the growth factor(s) responsiveness of these four progenitor populations in terms of proliferation and differentiation. Using mass spectrometry-based proteomics on sorted erythroid progenitors, we quantified the absolute expression of ~5500 proteins from EP1 to EP4. Further functional analyses highlighted dynamic changes in cell cycle in these populations with an acceleration of the cell cycle during erythroid progenitor differentiation. The finding that E2F4 expression was increased from EP1 to EP4 is consistent with the noted changes in cell cycle. Finally, our proteomic data suggest that the protein machinery necessary for both oxidative phosphorylation and glycolysis is present in these progenitor cells. Together, our data provide comprehensive insights into growth factor-dependence of erythroid progenitor proliferation and the proteome of four distinct populations of human erythroid progenitors which will be a useful framework for the study of erythroid disorders.
Collapse
Affiliation(s)
- Julien Papoin
- Institute of Molecular Medicine, Feinstein Institutes for
Medical Research, Manhasset, NY 11030 USA
- Université Jules Verne
| | - Hongxia Yan
- Red Cell Physiology Laboratory, Lindsey F. Kimball
Research Institute, New York Blood Center, New York, NY 10065 USA
| | - Marjorie Leduc
- Proteom’IC facility, Université Paris
Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Morgane Le Gall
- Proteom’IC facility, Université Paris
Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Anupama Narla
- Division of Hematology-Oncology, Department of Pediatrics,
Stanford University School of Medicine, Palo Alto, CA 94305 USA
| | - James Palis
- Center for Child Health Research, University of Rochester,
Rochester, NY 14642 USA
| | - Laurie A. Steiner
- Center for Child Health Research, University of Rochester,
Rochester, NY 14642 USA
| | - Patrick G. Gallagher
- Department of Pediatrics, Yale University, New Haven, CT
06520 USA
- Nationwide Children’s Hospital, Ohio State
University, Columbus, OH 43205 USA
| | - Christopher D. Hillyer
- Red Cell Physiology Laboratory, Lindsey F. Kimball
Research Institute, New York Blood Center, New York, NY 10065 USA
| | - Emilie-Fleur Gautier
- Proteom’IC facility, Université Paris
Cité, CNRS, INSERM, Institut Cochin, F-75014 Paris, France
| | - Narla Mohandas
- Red Cell Physiology Laboratory, Lindsey F. Kimball
Research Institute, New York Blood Center, New York, NY 10065 USA
| | - Lionel Blanc
- Institute of Molecular Medicine, Feinstein Institutes for
Medical Research, Manhasset, NY 11030 USA
- Division of Pediatrics Hematology/Oncology, Cohen
Children’s Medical Center, New Hyde Park NY 11040 USA
- Department of Molecular Medicine and Pediatrics, Zucker
School of Medicine at Hofstra/Northwell, Hempstead NY 11549 USA
| |
Collapse
|
11
|
Chung C. Current therapies for classic myeloproliferative neoplasms: A focus on pathophysiology and supportive care. Am J Health Syst Pharm 2023; 80:1624-1636. [PMID: 37556726 DOI: 10.1093/ajhp/zxad181] [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: 08/08/2023] [Indexed: 08/11/2023] Open
Abstract
PURPOSE This article concisely evaluates current therapies that have received regulatory approval for the treatment of classic myeloproliferative neoplasms (MPNs). Pertinent pathophysiology and supportive care are discussed. Emerging therapies are also briefly described. SUMMARY MPNs are a heterogeneous group of diseases characterized by acquired abnormalities of hematopoietic stem cells (HSCs), resulting in the generation of transformed myeloid progenitor cells that overproduce mature and immature cells within the myeloid lineage. Mutations in JAK2 and other driver oncogenes are central to the genetic variability of these diseases. Cytoreductive therapies such as hydroxyurea, anagrelide, interferon, and therapeutic phlebotomy aim to lower the risk of thrombotic events without exposing patients to an increased risk of leukemic transformation. However, no comparisons can be made between these therapies, as reduction of thrombotic risk has not been used as an endpoint. On the other hand, Janus kinase (JAK) inhibitors such as ruxolitinib, fedratinib, pacritinib, and momelotinib (an investigational agent at the time of writing) directly target the constitutively activated JAK-signal transducer and activator of transcription (JAK-STAT) pathway of HSCs in the bone marrow. Mutations of genes in the JAK-STAT signaling pathway provide a unifying understanding of MPNs, spur therapeutic innovations, and represent opportunities for pharmacists to optimize mitigation strategies for both disease-related and treatment-related adverse effects. CONCLUSION Treatment options for MPNs span a wide range of disease mechanisms. The growth of targeted therapies holds promise for expanding the treatment arsenal for these rare, yet complex diseases and creates opportunities to optimize supportive care for affected patients.
Collapse
|
12
|
Qin K, Lan X, Huang P, Saari MS, Khandros E, Keller CA, Giardine B, Abdulmalik O, Shi J, Hardison RC, Blobel GA. Molecular basis of polycomb group protein-mediated fetal hemoglobin repression. Blood 2023; 141:2756-2770. [PMID: 36893455 PMCID: PMC10273169 DOI: 10.1182/blood.2022019578] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/15/2023] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
The switch from fetal hemoglobin (HbF) to adult hemoglobin (HbA) is a paradigm for developmental gene expression control with relevance to sickle cell disease and β-thalassemia. Polycomb repressive complex (PRC) proteins regulate this switch, and an inhibitor of PRC2 has entered a clinical trial for HbF activation. Yet, how PRC complexes function in this process, their target genes, and relevant subunit composition are unknown. Here, we identified the PRC1 subunit BMI1 as a novel HbF repressor. We uncovered the RNA binding proteins LIN28B, IGF2BP1, and IGF2BP3 genes as direct BMI1 targets, and demonstrate that they account for the entirety of BMI1's effect on HbF regulation. BMI1 functions as part of the canonical PRC1 (cPRC1) subcomplex as revealed by the physical and functional dissection of BMI1 protein partners. Lastly, we demonstrate that BMI1/cPRC1 acts in concert with PRC2 to repress HbF through the same target genes. Our study illuminates how PRC silences HbF, highlighting an epigenetic mechanism involved in hemoglobin switching.
Collapse
Affiliation(s)
- Kunhua Qin
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Xianjiang Lan
- Department of Systems Biology for Medicine, School of Basic Medical Sciences, Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Huang
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Megan S. Saari
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Eugene Khandros
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Cheryl A. Keller
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, State College, PA
| | - Belinda Giardine
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, State College, PA
| | - Osheiza Abdulmalik
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Junwei Shi
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ross C. Hardison
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, State College, PA
| | - Gerd A. Blobel
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
13
|
Ureña-Bailén G, Block M, Grandi T, Aivazidou F, Quednau J, Krenz D, Daniel-Moreno A, Lamsfus-Calle A, Epting T, Handgretinger R, Wild S, Mezger M. Automated Good Manufacturing Practice-Compatible CRISPR-Cas9 Editing of Hematopoietic Stem and Progenitor Cells for Clinical Treatment of β-Hemoglobinopathies. CRISPR J 2023; 6:5-16. [PMID: 36662546 PMCID: PMC9986018 DOI: 10.1089/crispr.2022.0086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cellular therapies hold enormous potential for the cure of severe hematological and oncological disorders. The forefront of innovative gene therapy approaches including therapeutic gene editing and hematopoietic stem cell transplantation needs to be processed by good manufacturing practice to ensure safe application in patients. In the present study, an effective transfection protocol for automated clinical-scale production of genetically modified hematopoietic stem and progenitor cells (HSPCs) using the CliniMACS Prodigy® system including the CliniMACS Electroporator (Miltenyi Biotec) was established. As a proof-of-concept, the enhancer of the BCL11A gene, clustered regularly interspaced short palindromic repeat (CRISPR) target in ongoing clinical trials for β-thalassemia and sickle-cell disease treatment, was disrupted by the CRISPR-Cas9 system simulating a large-scale clinical scenario, yielding 100 million HSPCs with high editing efficiency. In vitro erythroid differentiation and high-performance liquid chromatography analyses corroborated fetal hemoglobin resurgence in edited samples, supporting the feasibility of running the complete process of HSPC gene editing in an automated closed system.
Collapse
Affiliation(s)
- Guillermo Ureña-Bailén
- Department of General Pediatrics, Oncology and Hematology, University Children's Hospital, Tübingen, Germany
| | - Milena Block
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tommaso Grandi
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | | | - Jona Quednau
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Dariusz Krenz
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Alberto Daniel-Moreno
- Department of General Pediatrics, Oncology and Hematology, University Children's Hospital, Tübingen, Germany
| | - Andrés Lamsfus-Calle
- Department of General Pediatrics, Oncology and Hematology, University Children's Hospital, Tübingen, Germany
| | - Thomas Epting
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital, Freiburg, Germany
| | - Rupert Handgretinger
- Department of General Pediatrics, Oncology and Hematology, University Children's Hospital, Tübingen, Germany.,Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Stefan Wild
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Markus Mezger
- Department of General Pediatrics, Oncology and Hematology, University Children's Hospital, Tübingen, Germany
| |
Collapse
|
14
|
Li H, Lin R, Li H, Ou R, Wang K, Lin J, Li C. MicroRNA-92a-3p-mediated inhibition of BCL11A upregulates γ-globin expression and inhibits oxidative stress and apoptosis in erythroid precursor cells. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2022; 27:1152-1162. [PMID: 36178486 DOI: 10.1080/16078454.2022.2128258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE This study attempted to investigate miR-92a-3p expression in peripheral blood of patients with severe β-thalassemia, and the effect and action mechanism of miR-92a-3p on γ-globin expression and oxidative stress in erythroid precursor cells. METHODS CD34+ hematopoietic progenitor cells (HPCs) were isolated from peripheral blood of healthy volunteers and patients with severe β-thalassemia. The levels of miR-92a-3p, BCL11A, and γ-globin were measured in erythroid precursor cells. High-performance liquid chromatography (HPLC) was used to analyze hemoglobin F (HbF) content. HPCs were induced with erythroid differentiation and erythroid precursor cells were then obtained. The relevance between miR-92a-3p and BCL11A was studied using dual luciferase reporter gene assay, and the correlation between miR-92a-3p and HbF was assayed by Pearson correlation analysis. Reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and superoxide dismutase (SOD) in erythroid precursor cells were tested to evaluate oxidative stress. Cell apoptosis was examined by flow cytometry. RESULTS Remarkably higher expression of miR-92a-3p was observed in erythroid precursor cells. Increased expression of miR-92a-3p resulted in elevated levels of γ-globin, GSH, and SOD, reduced expression of ROS and MDA, and decreased cell apoptosis. BCL11A was identified as a target of miR-92a-3p and to be downregulated by miR-92a-3p. Moreover, BCL11A knockdown alone increased the expression of γ-globin, SOD and GSH, and repressed the levels of ROS and MDA and cell apoptosis, and the following inhibition of miR-92a-3p changed these patterns. CONCLUSIONS Our data indicated that miR-92a-3p might increase γ-globin level and reduce oxidative stress and apoptosis in erythroid precursor cells by downregulating BCL11A.
Collapse
Affiliation(s)
- Huili Li
- Department of Pediatrics, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People's Republic of China
| | - Ruoping Lin
- Department of Pediatrics, Huizhou First Maternal and Child Health Care Hospital, Huizhou, People's Republic of China
| | - Huan Li
- Department of Laboratory, Nanfang-Chunfu Children's Institute of Hematology & Oncology, Dongguan, People's Republic of China
| | - Rilan Ou
- Department of Laboratory, Nanfang-Chunfu Children's Institute of Hematology & Oncology, Dongguan, People's Republic of China
| | - Kaiping Wang
- Department of Pediatrics, Huizhou First Maternal and Child Health Care Hospital, Huizhou, People's Republic of China
| | - Junrong Lin
- Department of Pediatrics, Huizhou First Maternal and Child Health Care Hospital, Huizhou, People's Republic of China
| | - Chunfu Li
- Department of Pediatrics, The First School of Clinical Medicine, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
15
|
Pavan AR, Lopes JR, Dos Santos JL. The state of the art of fetal hemoglobin-inducing agents. Expert Opin Drug Discov 2022; 17:1279-1293. [PMID: 36302760 DOI: 10.1080/17460441.2022.2141708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Sickle cell anemia (SCA) is a hematological genetic disorder caused by a mutation in the gene of the β-globin. Pharmacological treatments will continue to be an important approach, including the strategy to induce fetal hemoglobin (HbF). AREAS COVERED Here, we analyzed the articles described in the literature regarding the drug discovery of HbF inducers. The main approaches for such strategy will be discussed, highlighting those most promising. EXPERT OPINION The comprehension of the mechanisms involved in the β-globin regulation is the main key to design new drugs to induce HbF. Among the strategies, gamma-globin regulation by epigenetic enzymes seems to be a promising approach to be pursued, although the comprehension of the selectivity role for those new drugs is crucial to reduce adverse effects. The low druggability of transcription factors and their vital role in embryonic human development are critical points that should be taken in account for drug design. The guanylate cyclase and the NO/cGMP signaling pathway seem to be promising not only for HbF induction, but also for the protective effects in the cardiovascular system. The association of drugs acting through different mechanisms to induce HbF seems to be promising for the discovery of new drugs.
Collapse
Affiliation(s)
- Aline Renata Pavan
- São Paulo State University (UNESP), Institute of Chemistry, Araraquara, Brazil
| | - Juliana Romano Lopes
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Drugs and Medicine Department, Araraquara, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), Institute of Chemistry, Araraquara, Brazil.,School of Pharmaceutical Sciences, São Paulo State University (UNESP), Drugs and Medicine Department, Araraquara, Brazil
| |
Collapse
|
16
|
Bou-Fakhredin R, De Franceschi L, Motta I, Cappellini MD, Taher AT. Pharmacological Induction of Fetal Hemoglobin in β-Thalassemia and Sickle Cell Disease: An Updated Perspective. Pharmaceuticals (Basel) 2022; 15:ph15060753. [PMID: 35745672 PMCID: PMC9227505 DOI: 10.3390/ph15060753] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 12/04/2022] Open
Abstract
A significant amount of attention has recently been devoted to the mechanisms involved in hemoglobin (Hb) switching, as it has previously been established that the induction of fetal hemoglobin (HbF) production in significant amounts can reduce the severity of the clinical course in diseases such as β-thalassemia and sickle cell disease (SCD). While the induction of HbF using lentiviral and genome-editing strategies has been made possible, they present limitations. Meanwhile, progress in the use of pharmacologic agents for HbF induction and the identification of novel HbF-inducing strategies has been made possible as a result of a better understanding of γ-globin regulation. In this review, we will provide an update on all current pharmacological inducer agents of HbF in β-thalassemia and SCD in addition to the ongoing research into other novel, and potentially therapeutic, HbF-inducing agents.
Collapse
Affiliation(s)
- Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Verona, 37128 Verona, Italy;
| | - Irene Motta
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
- UOC General Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy; (R.B.-F.); (I.M.)
- UOC General Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Correspondence: (M.D.C.); (A.T.T.)
| | - Ali T. Taher
- Department of Internal Medicine, Division of Hematology-Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon
- Correspondence: (M.D.C.); (A.T.T.)
| |
Collapse
|
17
|
Lopez NH, Li B, Palani C, Siddaramappa U, Takezaki M, Xu H, Zhi W, Pace BS. Salubrinal induces fetal hemoglobin expression via the stress-signaling pathway in human sickle erythroid progenitors and sickle cell disease mice. PLoS One 2022; 17:e0261799. [PMID: 35639781 PMCID: PMC9154101 DOI: 10.1371/journal.pone.0261799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/05/2022] [Indexed: 11/25/2022] Open
Abstract
Sickle cell disease (SCD) is an inherited blood disorder caused by a mutation in the HBB gene leading to hemoglobin S production and polymerization under hypoxia conditions leading to vaso-occlusion, chronic hemolysis, and progressive organ damage. This disease affects ~100,000 people in the United States and millions worldwide. An effective therapy for SCD is fetal hemoglobin (HbF) induction by pharmacologic agents such as hydroxyurea, the only Food and Drug Administration-approved drug for this purpose. Therefore, the goal of our study was to determine whether salubrinal (SAL), a selective protein phosphatase 1 inhibitor, induces HbF expression through the stress-signaling pathway by activation of p-eIF2α and ATF4 trans-activation in the γ-globin gene promoter. Sickle erythroid progenitors treated with 24μM SAL increased F-cells levels 1.4-fold (p = 0.021) and produced an 80% decrease in reactive oxygen species. Western blot analysis showed SAL enhanced HbF protein by 1.6-fold (p = 0.0441), along with dose-dependent increases of p-eIF2α and ATF4 levels. Subsequent treatment of SCD mice by a single intraperitoneal injection of SAL (5mg/kg) produced peak plasma concentrations at 6 hours. Chronic treatments of SCD mice with SAL mediated a 2.3-fold increase in F-cells (p = 0.0013) and decreased sickle erythrocytes supporting in vivo HbF induction.
Collapse
Affiliation(s)
- Nicole H. Lopez
- Department of Biochemistry and Cancer Biology, Augusta University, Augusta, GA, United States of America
| | - Biaoru Li
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| | - Chithra Palani
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| | - Umapathy Siddaramappa
- Department of Medicine, Division of Hematology/Oncology Augusta University, Augusta GA, United States of America
| | - Mayuko Takezaki
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| | - Hongyan Xu
- Department of Biostatistics and Epidemiology, Augusta University, Augusta, GA, United States of America
| | - Wenbo Zhi
- Center for Biotechnology & Genomic Medicine, Augusta University, Augusta, GA, United States of America
| | - Betty S. Pace
- Department of Biochemistry and Cancer Biology, Augusta University, Augusta, GA, United States of America
- Department of Pediatrics, Augusta University, Augusta, GA, United States of America
| |
Collapse
|
18
|
Dulmovits BM, Tang Y, Papoin J, He M, Li J, Yang H, Addorisio ME, Kennedy L, Khan M, Brindley E, Ashley RJ, Ackert-Bicknell C, Hale J, Kurita R, Nakamura Y, Diamond B, Barnes BJ, Hermine O, Gallagher PG, Steiner LA, Lipton JM, Taylor N, Mohandas N, Andersson U, Al-Abed Y, Tracey KJ, Blanc L. HMGB1-mediated restriction of EPO signaling contributes to anemia of inflammation. Blood 2022; 139:3181-3193. [PMID: 35040907 PMCID: PMC9136881 DOI: 10.1182/blood.2021012048] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 12/22/2021] [Indexed: 11/20/2022] Open
Abstract
Anemia of inflammation, also known as anemia of chronic disease, is refractory to erythropoietin (EPO) treatment, but the mechanisms underlying the EPO refractory state are unclear. Here, we demonstrate that high mobility group box-1 protein (HMGB1), a damage-associated molecular pattern molecule recently implicated in anemia development during sepsis, leads to reduced expansion and increased death of EPO-sensitive erythroid precursors in human models of erythropoiesis. HMGB1 significantly attenuates EPO-mediated phosphorylation of the Janus kinase 2/STAT5 and mTOR signaling pathways. Genetic ablation of receptor for advanced glycation end products, the only known HMGB1 receptor expressed by erythroid precursors, does not rescue the deleterious effects of HMGB1 on EPO signaling, either in human or murine precursors. Furthermore, surface plasmon resonance studies highlight the ability of HMGB1 to interfere with the binding between EPO and the EPOR. Administration of a monoclonal anti-HMGB1 antibody after sepsis onset in mice partially restores EPO signaling in vivo. Thus, HMGB1-mediated restriction of EPO signaling contributes to the chronic phase of anemia of inflammation.
Collapse
Affiliation(s)
- Brian M Dulmovits
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | | | | | - Mingzhu He
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Jianhua Li
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Huan Yang
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Meghan E Addorisio
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | | | | | - Elena Brindley
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | - Ryan J Ashley
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | | | - John Hale
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY
| | - Ryo Kurita
- Central Blood Institute, Japanese Red Cross Society, Minato-ku, Tokyo, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Betty Diamond
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | - Betsy J Barnes
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | - Olivier Hermine
- INSERM Unité Mixte de Recherche (UMR) 1163, IMAGINE Institute, Paris, France
| | | | - Laurie A Steiner
- Department of Pediatrics, University of Rochester, Rochester, NY
| | - Jeffrey M Lipton
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
- Pediatric Hematology/Oncology, Cohen Children's Medical Center, New Hyde Park, NY
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD; and
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Yousef Al-Abed
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Kevin J Tracey
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Lionel Blanc
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
- INSERM Unité Mixte de Recherche (UMR) 1163, IMAGINE Institute, Paris, France
| |
Collapse
|
19
|
Abstract
INTRODUCTION Sickle cell disease and β thalassemia are the principal β hemoglobinopathies. The complex pathophysiology of sickle cell disease is initiated by sickle hemoglobin polymerization. In β thalassemia, insufficient β-globin synthesis results in excessive free α globin, ineffective erythropoiesis and severe anemia. Fetal hemoglobin (HbF) prevents sickle hemoglobin polymerization; in β thalassemia HbF compensates for the deficit of normal hemoglobin. When HbF constitutes about a third of total cell hemoglobin, the complications of sickle cell disease are nearly totally prevented. Similarly, sufficient HbF in β thalassemia diminishes or prevents ineffective erythropoiesis and hemolysis. AREAS COVERED This article examines the pathophysiology of β hemoglobinopathies, the physiology of HbF, intracellular distribution and the regulation of HbF expression. Inducing high levels of HbF by targeting its regulatory pathways pharmacologically or with cell-based therapeutics provides major clinical benefit and perhaps a "cure." EXPERT OPINION Erythrocytes must contain about 10 pg of HbF to "cure" sickle cell disease. If HbF is the only hemoglobin present, much higher levels are needed to "cure" β thalassemia. These levels of HbF can be obtained by different iterations of gene therapy. Small molecule drugs that can achieve even modest pancellular HbF concentrations are a major unmet need.
Collapse
Affiliation(s)
- Martin H Steinberg
- Professor of Medicine, Pediatrics, Pathology and Laboratory Medicine, Boston University School of Medicine.,Department of Medicine, Division of Hematology/Oncology, Center of Excellence for Sickle Cell Disease, Boston University School of Medicine, 72 East Concord St., Boston, MA, 02118, USA.,Department of Medicine, Boston University School of Medicine, 72 E. Concord St. Boston, MA 02118. ., Tel
| |
Collapse
|
20
|
Lu Y, Wei Z, Yang G, Lai Y, Liu R. Investigating the Efficacy and Safety of Thalidomide for Treating Patients With ß-Thalassemia: A Meta-Analysis. Front Pharmacol 2022; 12:814302. [PMID: 35087410 PMCID: PMC8786914 DOI: 10.3389/fphar.2021.814302] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/17/2021] [Indexed: 12/20/2022] Open
Abstract
At present, the main therapies for ß-thalassemia patients include regular blood transfusion and iron chelation, associating with a number of limitations. Thalidomide, a fetal hemoglobin (HbF) inducer that promotes γ-globin gene expression, has been reported to be effective for ß-thalassemia. Thus, this meta-analysis was conducted to assess the efficacy and safety of thalidomide for treating patients with ß-thalassemia. We searched the related studies from eight databases published from inception until December 1, 2021. The R 4.0.5 language programming was used to perform meta-analysis. After screening of retrieved articles, 12 articles were included that enrolled a total of 451 patients. The Cochrane Collaboration risk assessment tool was used to evaluate the quality and the bias risk of the randomized controlled trials (RCTs), and non randomized trials were assessed using Newcastle-Ottawa Scale (NOS). After treatment with thalidomide, the pooled overall response rate (ORR) was 85% (95% confidence interval (CI): 80–90%), and the pooled complete response rate (CRR) was 54% (95% confidence interval: 31–76%). Compared with the placebo group, the thalidomide group had higher odds of overall response rate (odds ratio = 20.4; 95% CI: 6.75–61.64) and complete response rate (odds ratio = 20.4; 95% CI: 6.75–61.64). A statistically significant increase in hemoglobin level and HbF level after treatment, while there was no statistically significant difference in adult hemoglobin (HbA) level, spleen size, and serum ferritin. According to the results of ORR and CRR, transfusion-dependent thalassemia (TDT) patients showed remarkable efficacy of thalidomide, 83 and 52% respectively. So we analyzed 30 transfusion-dependent thalassemia patients from three studies and found that the most frequent ß-globin gene mutations were CD41-42 (-TCTT), while response to thalidomide did not show any statistically significant relationship with XmnI polymorphism or CD41-42 (-TCTT) mutation. About 30% of patients experienced mild adverse effects of thalidomide. Collectively, thalidomide is a relatively safe and effective therapy to reduce the blood transfusion requirements and to increase Hb level in patients with ß-thalassemia.
Collapse
Affiliation(s)
- Yanfei Lu
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenbin Wei
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gaohui Yang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yongrong Lai
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Rongrong Liu
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
21
|
Chen JM, Zhu WJ, Liu J, Wang GZ, Chen XQ, Tan Y, Xu WW, Qu LW, Li JY, Yang HJ, Huang L, Cai N, Wang WD, Huang K, Xu JQ, Li GH, He S, Luo TY, Huang Y, Liu SH, Wu WQ, Lu QY, Zhou MG, Chen SY, Li RL, Hu ML, Huang Y, Wei JH, Li JM, Chen SJ, Zhou GB. Safety and efficacy of thalidomide in patients with transfusion-dependent β-thalassemia: a randomized clinical trial. Signal Transduct Target Ther 2021; 6:405. [PMID: 34795208 PMCID: PMC8602273 DOI: 10.1038/s41392-021-00811-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 01/19/2023] Open
Abstract
Thalidomide induces γ-globin expression in erythroid progenitor cells, but its efficacy on patients with transfusion-dependent β-thalassemia (TDT) remains unclear. In this phase 2, multi-center, randomized, double-blind clinical trial, we aimed to determine the safety and efficacy of thalidomide in TDT patients. A hundred patients of 14 years or older were randomly assigned to receive placebo or thalidomide for 12 weeks, followed by an extension phase of at least 36 weeks. The primary endpoint was the change of hemoglobin (Hb) level in the patients. The secondary endpoints included the red blood cell (RBC) units transfused and adverse effects. In the placebo-controlled period, Hb concentrations in patients treated with thalidomide achieved a median elevation of 14.0 (range, 2.5 to 37.5) g/L, whereas Hb in patients treated with placebo did not significantly change. Within the 12 weeks, the mean RBC transfusion volume for patients treated with thalidomide and placebo was 5.4 ± 5.0 U and 10.3 ± 6.4 U, respectively (P < 0.001). Adverse events of drowsiness, dizziness, fatigue, pyrexia, sore throat, and rash were more common with thalidomide than placebo. In the extension phase, treatment with thalidomide for 24 weeks resulted in a sustainable increase in Hb concentrations which reached 104.9 ± 19.0 g/L, without blood transfusion. Significant increase in Hb concentration and reduction in RBC transfusions were associated with non β0/β0 and HBS1L-MYB (rs9399137 C/T, C/C; rs4895441 A/G, G/G) genotypes. These results demonstrated that thalidomide is effective in patients with TDT.
Collapse
Affiliation(s)
- Jiang-Ming Chen
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China.
| | - Wei-Jian Zhu
- Department of Hematology, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 541000, Guangdong, China
| | - Jie Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Reproductive Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Gui-Zhen Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiao-Qin Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Medical Oncology Department, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Yun Tan
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei-Wei Xu
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Li-Wei Qu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences & University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Jin-Yan Li
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Huan-Ju Yang
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Lan Huang
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Ning Cai
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Wei-Da Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Medical Oncology Department, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, China
| | - Ken Huang
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise City, 533000, Guangxi, China
| | - Jian-Quan Xu
- Department of Hematology, Yulin Guinan Hospital, Yulin, 537005, Guangxi, China
| | - Guo-Hui Li
- Department of neurology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Sheng He
- Guangxi Key Laboratory of Basic Research on Birth Defects Prevention and Treatment, Guangxi Zhuang Autonomous Region Women and Children Health Care Hospital, Nanning, 530000, Guangxi, China
| | - Tian-Ying Luo
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Yi Huang
- Department of Hematology, Guigang People's Hospital, Guigang, 537100, Guangxi, China
| | - Song-Hua Liu
- Department of Hematology, Hospital of Traditional Chinese Medicine of Wuzhou City, Wuzhou, 543002, Guangxi, China
| | - Wen-Qiang Wu
- Department of Hematology, Wuzhou Red Cross Hospital, Wuzhou, 543002, Guangxi, China
| | - Qi-Yang Lu
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Mei-Guang Zhou
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Shu-Ying Chen
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Rong-Lan Li
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Mei-Ling Hu
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Ying Huang
- Department of Hematology, Wuzhou Gongren Hospital, Wuzhou, 543001, Guangxi, China
| | - Jin-Hua Wei
- Department of Hematology, Hechi People's Hospital, Hechi City, 547000, Guangxi, China
| | - Jun-Min Li
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Sai-Juan Chen
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Guang-Biao Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
22
|
de Melo TRF, Dulmovits BM, Fernandes GFDS, de Souza CM, Lanaro C, He M, Al Abed Y, Chung MC, Blanc L, Costa FF, Dos Santos JL. Synthesis and pharmacological evaluation of pomalidomide derivatives useful for sickle cell disease treatment. Bioorg Chem 2021; 114:105077. [PMID: 34130111 PMCID: PMC8387409 DOI: 10.1016/j.bioorg.2021.105077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Fetal hemoglobin (HbF) induction constitutes a valuable and validated approach to treat the symptoms of sickle cell disease (SCD). Here, we synthesized pomalidomide-nitric oxide (NO) donor derivatives (3a-f) and evaluated their suitability as novel HbF inducers. All compounds demonstrated different capacities of releasing NO, ranging 0.3-30.3%. Compound 3d was the most effective HbF inducer for CD34+ cells, exhibiting an effect similar to that of hydroxyurea. We investigated the mode of action of compound 3d for HbF induction by studying the in vitro alterations in the levels of transcription factors (BCL11A, IKAROS, and LRF), inhibition of histone deacetylase enzymes (HDAC-1 and HDAC-2), and measurement of cGMP levels. Additionally, compound 3d exhibited a potent anti-inflammatory effect similar to that of pomalidomide by reducing the TNF-α levels in human mononuclear cells treated with lipopolysaccharides up to 58.6%. Chemical hydrolysis studies revealed that compound 3d was stable at pH 7.4 up to 24 h. These results suggest that compound 3d is a novel HbF inducer prototype with the potential to treat SCD symptoms.
Collapse
Affiliation(s)
| | - Brian M Dulmovits
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | | | - Cristiane M de Souza
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Carolina Lanaro
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Minghzu He
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Yousef Al Abed
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Man Chin Chung
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil
| | - Lionel Blanc
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Fernando Ferreira Costa
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil.
| |
Collapse
|
23
|
Lattanzi A, Camarena J, Lahiri P, Segal H, Srifa W, Vakulskas CA, Frock RL, Kenrick J, Lee C, Talbott N, Skowronski J, Cromer MK, Charlesworth CT, Bak RO, Mantri S, Bao G, DiGiusto D, Tisdale J, Wright JF, Bhatia N, Roncarolo MG, Dever DP, Porteus MH. Development of β-globin gene correction in human hematopoietic stem cells as a potential durable treatment for sickle cell disease. Sci Transl Med 2021; 13:13/598/eabf2444. [PMID: 34135108 DOI: 10.1126/scitranslmed.abf2444] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 05/25/2021] [Indexed: 12/11/2022]
Abstract
Sickle cell disease (SCD) is the most common serious monogenic disease with 300,000 births annually worldwide. SCD is an autosomal recessive disease resulting from a single point mutation in codon six of the β-globin gene (HBB). Ex vivo β-globin gene correction in autologous patient-derived hematopoietic stem and progenitor cells (HSPCs) may potentially provide a curative treatment for SCD. We previously developed a CRISPR-Cas9 gene targeting strategy that uses high-fidelity Cas9 precomplexed with chemically modified guide RNAs to induce recombinant adeno-associated virus serotype 6 (rAAV6)-mediated HBB gene correction of the SCD-causing mutation in HSPCs. Here, we demonstrate the preclinical feasibility, efficacy, and toxicology of HBB gene correction in plerixafor-mobilized CD34+ cells from healthy and SCD patient donors (gcHBB-SCD). We achieved up to 60% HBB allelic correction in clinical-scale gcHBB-SCD manufacturing. After transplant into immunodeficient NSG mice, 20% gene correction was achieved with multilineage engraftment. The long-term safety, tumorigenicity, and toxicology study demonstrated no evidence of abnormal hematopoiesis, genotoxicity, or tumorigenicity from the engrafted gcHBB-SCD drug product. Together, these preclinical data support the safety, efficacy, and reproducibility of this gene correction strategy for initiation of a phase 1/2 clinical trial in patients with SCD.
Collapse
Affiliation(s)
- Annalisa Lattanzi
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.,Center for Definitive and Curative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Joab Camarena
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Premanjali Lahiri
- Laboratory for Cell and Gene Medicine, Stanford University, Stanford, CA 94304, USA
| | - Helen Segal
- Laboratory for Cell and Gene Medicine, Stanford University, Stanford, CA 94304, USA
| | - Waracharee Srifa
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | | | - Richard L Frock
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Josefin Kenrick
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Ciaran Lee
- APC Microbiome Ireland, University College Cork, T12 YN60 Cork, Ireland
| | - Narae Talbott
- Laboratory for Cell and Gene Medicine, Stanford University, Stanford, CA 94304, USA
| | - Jason Skowronski
- Laboratory for Cell and Gene Medicine, Stanford University, Stanford, CA 94304, USA
| | - M Kyle Cromer
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | | | - Rasmus O Bak
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark.,Aarhus Institute of Advanced Studies (AIAS), Aarhus University, DK-8000 Aarhus, Denmark
| | - Sruthi Mantri
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX 77006, USA
| | - David DiGiusto
- Laboratory for Cell and Gene Medicine, Stanford University, Stanford, CA 94304, USA
| | - John Tisdale
- Molecular and Clinical Hematology Branch, NHLBI, Bethesda, MD 20814, USA
| | - J Fraser Wright
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.,Center for Definitive and Curative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Neehar Bhatia
- Laboratory for Cell and Gene Medicine, Stanford University, Stanford, CA 94304, USA.,Deceased
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.,Center for Definitive and Curative Medicine, Stanford University, Stanford, CA 94305, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Daniel P Dever
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA. .,Center for Definitive and Curative Medicine, Stanford University, Stanford, CA 94305, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
24
|
Ineffective Erythropoiesis in β-Thalassaemia: Key Steps and Therapeutic Options by Drugs. Int J Mol Sci 2021; 22:ijms22137229. [PMID: 34281283 PMCID: PMC8268821 DOI: 10.3390/ijms22137229] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 01/19/2023] Open
Abstract
β-thalassaemia is a rare genetic condition caused by mutations in the β-globin gene that result in severe iron-loading anaemia, maintained by a detrimental state of ineffective erythropoiesis (IE). The role of multiple mechanisms involved in the pathophysiology of the disease has been recently unravelled. The unbalanced production of α-globin is a major source of oxidative stress and membrane damage in red blood cells (RBC). In addition, IE is tightly linked to iron metabolism dysregulation, and the relevance of new players of this pathway, i.e., hepcidin, erythroferrone, matriptase-2, among others, has emerged. Advances have been made in understanding the balance between proliferation and maturation of erythroid precursors and the role of specific factors in this process, such as members of the TGF-β superfamily, and their downstream effectors, or the transcription factor GATA1. The increasing understanding of IE allowed for the development of a broad set of potential therapeutic options beyond the current standard of care. Many candidates of disease-modifying drugs are currently under clinical investigation, targeting the regulation of iron metabolism, the production of foetal haemoglobin, the maturation process, or the energetic balance and membrane stability of RBC. Overall, they provide tools and evidence for multiple and synergistic approaches that are effectively moving clinical research in β-thalassaemia from bench to bedside.
Collapse
|
25
|
Pavan AR, Dos Santos JL. Advances in Sickle Cell Disease Treatments. Curr Med Chem 2021; 28:2008-2032. [PMID: 32520675 DOI: 10.2174/0929867327666200610175400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/23/2020] [Accepted: 05/07/2020] [Indexed: 11/22/2022]
Abstract
Sickle Cell Disease (SCD) is an inherited disorder of red blood cells that is caused by a single mutation in the β -globin gene. The disease, which afflicts millions of patients worldwide mainly in low income countries, is characterized by high morbidity, mortality and low life expectancy. The new pharmacological and non-pharmacological strategies for SCD is urgent in order to promote treatments able to reduce patient's suffering and improve their quality of life. Since the FDA approval of HU in 1998, there have been few advances in discovering new drugs; however, in the last three years voxelotor, crizanlizumab, and glutamine have been approved as new therapeutic alternatives. In addition, new promising compounds have been described to treat the main SCD symptoms. Herein, focusing on drug discovery, we discuss new strategies to treat SCD that have been carried out in the last ten years to discover new, safe, and effective treatments. Moreover, non-pharmacological approaches, including red blood cell exchange, gene therapy and hematopoietic stem cell transplantation will be presented.
Collapse
Affiliation(s)
- Aline Renata Pavan
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Brazil
| | - Jean Leandro Dos Santos
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, Sao Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
26
|
HRI depletion cooperates with pharmacologic inducers to elevate fetal hemoglobin and reduce sickle cell formation. Blood Adv 2021; 4:4560-4572. [PMID: 32956454 DOI: 10.1182/bloodadvances.2020002475] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Increasing fetal hemoglobin (HbF) provides clinical benefit in patients with sickle cell disease (SCD). We recently identified heme-regulated inhibitor (HRI, EIF2AK1), as a novel HbF regulator. Because HRI is an erythroid-specific protein kinase, it presents a potential target for pharmacologic intervention. We found that maximal HbF induction required >80% to 85% HRI depletion. Because it remains unclear whether this degree of HRI inhibition can be achieved pharmacologically, we explored whether HRI knockdown can be combined with pharmacologic HbF inducers to achieve greater HbF production and minimize potential adverse effects associated with treatments. Strongly cooperative HbF induction was observed when HRI depletion was combined with exposure to pomalidomide or the EHMT1/2 inhibitor UNC0638, but not to hydroxyurea. Mechanistically, reduction in the levels of the HbF repressor BCL11A reflected the cooperativity of HRI loss and pomalidomide treatment, whereas UNC0638 did not modulate BCL11A levels. In conjunction with HRI loss, pomalidomide maintained its HbF-inducing activity at 10-fold lower concentrations, in which condition there were minimal observed detrimental effects on erythroid cell maturation and viability, as well as fewer alterations in the erythroid transcriptome. When tested in cells from patients with SCD, combining HRI depletion with pomalidomide or UNC0638 achieved up to 50% to 60% HbF and 45% to 50% HbF, respectively, as measured by high-performance liquid chromatography, and markedly counteracted cell sickling. In summary, this study provides a foundation for the exploration of combining future small-molecule HRI inhibitors with additional pharmacologic HbF inducers to maximize HbF production and preserve erythroid cell functionality for the treatment of SCD and other hemoglobinopathies.
Collapse
|
27
|
Isidori A, Loscocco F, Visani G, Chiarucci M, Musto P, Kubasch AS, Platzbecker U, Vinchi F. Iron Toxicity and Chelation Therapy in Hematopoietic Stem Cell Transplant. Transplant Cell Ther 2021; 27:371-379. [PMID: 33969823 DOI: 10.1016/j.jtct.2020.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 01/19/2023]
Abstract
Many patients with hematologic malignancies receive RBC transfusion support, which often causes systemic and tissue iron toxicity. Because of their compromised bone marrow function, hematopoietic stem cell transplant (HSCT) recipients are especially vulnerable to excess iron levels. Iron toxicity may compromise transplant engraftment and eventually promote relapse by mediating oxidative and genotoxic stress in hematopoietic stem cells (HSCs) and further impairing the already dysfunctional bone marrow microenvironment in HSCT recipients. Iron toxicity is thought to be primarily mediated by its ability to induce reactive oxygen species and trigger inflammation. Elevated iron levels in the bone marrow can decrease the number of HSCs and progenitor cells, as well as their clonogenic potential, alter mesenchymal stem cell differentiation, and inhibit the expression of chemokines and adhesion molecules involved in hematopoiesis. In vivo, in vitro, and clinical studies support the concept that iron chelation therapy may limit iron toxicity in the bone marrow and promote hematologic improvement and engraftment in HSCT recipients. This review will provide an overview of the current knowledge of the detrimental impact of iron toxicity in the setting of HSCT in patients with hematologic malignancies and the use of iron restriction approaches to improve transplant outcome.
Collapse
Affiliation(s)
- Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy.
| | - Federica Loscocco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Martina Chiarucci
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Pellegrino Musto
- Unit of Hematology and Stem Cell Transplantation, Department of Emergency and Organ Transplantation, "Aldo Moro" University School of Medicine, AOU Consorziale Policlinico, Bari, Italy
| | - Anne-Sophie Kubasch
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Francesca Vinchi
- Iron Research Program, Lindsley Kimball Research Institute, New York Blood Center, New York, New York; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
28
|
Cromer MK, Camarena J, Martin RM, Lesch BJ, Vakulskas CA, Bode NM, Kurgan G, Collingwood MA, Rettig GR, Behlke MA, Lemgart VT, Zhang Y, Goyal A, Zhao F, Ponce E, Srifa W, Bak RO, Uchida N, Majeti R, Sheehan VA, Tisdale JF, Dever DP, Porteus MH. Gene replacement of α-globin with β-globin restores hemoglobin balance in β-thalassemia-derived hematopoietic stem and progenitor cells. Nat Med 2021; 27:677-687. [PMID: 33737751 PMCID: PMC8265212 DOI: 10.1038/s41591-021-01284-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
β-Thalassemia pathology is due not only to loss of β-globin (HBB), but also to erythrotoxic accumulation and aggregation of the β-globin-binding partner, α-globin (HBA1/2). Here we describe a Cas9/AAV6-mediated genome editing strategy that can replace the entire HBA1 gene with a full-length HBB transgene in β-thalassemia-derived hematopoietic stem and progenitor cells (HSPCs), which is sufficient to normalize β-globin:α-globin messenger RNA and protein ratios and restore functional adult hemoglobin tetramers in patient-derived red blood cells. Edited HSPCs were capable of long-term and bilineage hematopoietic reconstitution in mice, establishing proof of concept for replacement of HBA1 with HBB as a novel therapeutic strategy for curing β-thalassemia.
Collapse
Affiliation(s)
- M Kyle Cromer
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Joab Camarena
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Renata M Martin
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Benjamin J Lesch
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | | - Nicole M Bode
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| | - Gavin Kurgan
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| | | | | | - Mark A Behlke
- Integrated DNA Technologies, Inc., Coralville, IA, USA
| | - Viktor T Lemgart
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Yankai Zhang
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Ankush Goyal
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Feifei Zhao
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Ezequiel Ponce
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Waracharee Srifa
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Rasmus O Bak
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Naoya Uchida
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Vivien A Sheehan
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - John F Tisdale
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel P Dever
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
- Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
29
|
Insight of fetal to adult hemoglobin switch: Genetic modulators and therapeutic targets. Blood Rev 2021; 49:100823. [PMID: 33726930 DOI: 10.1016/j.blre.2021.100823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/08/2021] [Accepted: 03/03/2021] [Indexed: 01/31/2023]
Abstract
The clinical heterogeneity of β-hemoglobinopathies is so variable that it prompted the researchers to identify the genetic modulators of these diseases. Though the primary modulator is the type of β-globin mutation which affects the degree of β-globin chain synthesis, the co-inheritance of α-thalassemia and the fetal hemoglobin (HbF) levels also act as potent secondary genetic modifiers. As elevated HbF levels ameliorate the severity of hemoglobinopathies, in this review, the genetic modulators lying within and outside the β-globin gene cluster with their plausible role in governing the HbF levels have been summarised, which in future may act as potential therapeutic targets.
Collapse
|
30
|
Bottardi S, Milot E. An early start of Coup-TFII promotes γ-globin gene expression in adult erythroid cells. Haematologica 2021; 106:335-336. [PMID: 33522785 PMCID: PMC7849336 DOI: 10.3324/haematol.2020.266791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 11/27/2022] Open
Affiliation(s)
- Stefania Bottardi
- Maisonneuve Rosemont Hospital Research Center, CIUSSS Est de l'Île de Montréal, Montréal
| | - Eric Milot
- Maisonneuve Rosemont Hospital Research Center, CIUSSS Est de l'Île de Montréal, Montréal; Department of Medicine, University of Montreal, Montréal, Québec.
| |
Collapse
|
31
|
Sharma R, Dever DP, Lee CM, Azizi A, Pan Y, Camarena J, Köhnke T, Bao G, Porteus MH, Majeti R. The TRACE-Seq method tracks recombination alleles and identifies clonal reconstitution dynamics of gene targeted human hematopoietic stem cells. Nat Commun 2021; 12:472. [PMID: 33473139 PMCID: PMC7817666 DOI: 10.1038/s41467-020-20792-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Targeted DNA correction of disease-causing mutations in hematopoietic stem and progenitor cells (HSPCs) may enable the treatment of genetic diseases of the blood and immune system. It is now possible to correct mutations at high frequencies in HSPCs by combining CRISPR/Cas9 with homologous DNA donors. Because of the precision of gene correction, these approaches preclude clonal tracking of gene-targeted HSPCs. Here, we describe Tracking Recombination Alleles in Clonal Engraftment using sequencing (TRACE-Seq), a methodology that utilizes barcoded AAV6 donor template libraries, carrying in-frame silent mutations or semi-randomized nucleotides outside the coding region, to track the in vivo lineage contribution of gene-targeted HSPC clones. By targeting the HBB gene with an AAV6 donor template library consisting of ~20,000 possible unique exon 1 in-frame silent mutations, we track the hematopoietic reconstitution of HBB targeted myeloid-skewed, lymphoid-skewed, and balanced multi-lineage repopulating human HSPC clones in mice. We anticipate this methodology could potentially be used for HSPC clonal tracking of Cas9 RNP and AAV6-mediated gene targeting outcomes in translational and basic research settings.
Collapse
Affiliation(s)
- Rajiv Sharma
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Daniel P Dever
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Ciaran M Lee
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Armon Azizi
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Yidan Pan
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Joab Camarena
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Thomas Köhnke
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA.
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
32
|
When basic science reaches into rational therapeutic design: from historical to novel leads for the treatment of β-globinopathies. Curr Opin Hematol 2021; 27:141-148. [PMID: 32167946 DOI: 10.1097/moh.0000000000000577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW β-hemoglobinopathies, such as β-Thalassemias (β-Thal) and sickle cell disease (SCD) are among the most common inherited genetic disorders in humans worldwide. These disorders are characterized by a quantitative (β-Thal) or qualitative (SCD) defects in adult hemoglobin production, leading to anemia, ineffective erythropoiesis and severe secondary complications. Reactivation of the fetal globin genes (γ-globin), making-up fetal hemoglobin (HbF), which are normally silenced in adults, represents a major strategy to ameliorate anemia and disease severity. RECENT FINDINGS Following the identification of the first 'switching factors' for the reactivation of fetal globin gene expression more than 10 years ago, a multitude of novel leads have recently been uncovered. SUMMARY Recent findings provided invaluable functional insights into the genetic and molecular networks controlling globin genes expression, revealing that complex repression systems evolved in erythroid cells to maintain HbF silencing in adults. This review summarizes these unique and exciting discoveries of the regulatory factors controlling the globin switch. New insights and novel leads for therapeutic strategies based on the pharmacological induction of HbF are discussed. This represents a major breakthrough for rational drug design in the treatment of β-Thal and SCD.
Collapse
|
33
|
Ashley RJ, Yan H, Wang N, Hale J, Dulmovits BM, Papoin J, Olive ME, Udeshi ND, Carr SA, Vlachos A, Lipton JM, Da Costa L, Hillyer C, Kinet S, Taylor N, Mohandas N, Narla A, Blanc L. Steroid resistance in Diamond Blackfan anemia associates with p57Kip2 dysregulation in erythroid progenitors. J Clin Invest 2020; 130:2097-2110. [PMID: 31961825 DOI: 10.1172/jci132284] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
Despite the effective clinical use of steroids for the treatment of Diamond Blackfan anemia (DBA), the mechanisms through which glucocorticoids regulate human erythropoiesis remain poorly understood. We report that the sensitivity of erythroid differentiation to dexamethasone is dependent on the developmental origin of human CD34+ progenitor cells, specifically increasing the expansion of CD34+ progenitors from peripheral blood (PB) but not cord blood (CB). Dexamethasone treatment of erythroid-differentiated PB, but not CB, CD34+ progenitors resulted in the expansion of a newly defined CD34+CD36+CD71hiCD105med immature colony-forming unit-erythroid (CFU-E) population. Furthermore, proteomics analyses revealed the induction of distinct proteins in dexamethasone-treated PB and CB erythroid progenitors. Dexamethasone treatment of PB progenitors resulted in the specific upregulation of p57Kip2, a Cip/Kip cyclin-dependent kinase inhibitor, and we identified this induction as critical; shRNA-mediated downregulation of p57Kip2, but not the related p27Kip1, significantly attenuated the impact of dexamethasone on erythroid differentiation and inhibited the expansion of the immature CFU-E subset. Notably, in the context of DBA, we found that steroid resistance was associated with dysregulated p57Kip2 expression. Altogether, these data identify a unique glucocorticoid-responsive human erythroid progenitor and provide new insights into glucocorticoid-based therapeutic strategies for the treatment of patients with DBA.
Collapse
Affiliation(s)
- Ryan J Ashley
- Department of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Hempstead, New York, USA.,Center for Autoimmunity, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Hongxia Yan
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York, USA.,Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Montpellier, France
| | - Nan Wang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - John Hale
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York, USA
| | - Brian M Dulmovits
- Department of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Hempstead, New York, USA.,Center for Autoimmunity, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Julien Papoin
- Center for Autoimmunity, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Meagan E Olive
- Proteomics Platform, Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Namrata D Udeshi
- Proteomics Platform, Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Steven A Carr
- Proteomics Platform, Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts, USA
| | - Adrianna Vlachos
- Department of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Hempstead, New York, USA.,Center for Autoimmunity, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Pediatric Hematology/Oncology, Cohen Children's Medical Center, New Hyde Park, New York, USA
| | - Jeffrey M Lipton
- Department of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Hempstead, New York, USA.,Center for Autoimmunity, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Pediatric Hematology/Oncology, Cohen Children's Medical Center, New Hyde Park, New York, USA
| | | | - Christopher Hillyer
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York, USA
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Montpellier, France
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, Montpellier, France.,Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York, USA
| | - Anupama Narla
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | - Lionel Blanc
- Department of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine, Hofstra/Northwell, Hempstead, New York, USA.,Center for Autoimmunity, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Pediatric Hematology/Oncology, Cohen Children's Medical Center, New Hyde Park, New York, USA
| |
Collapse
|
34
|
Robledo RF, Ciciotte SL, Graber JH, Zhao Y, Lambert AJ, Gwynn B, Maki NJ, Brindley EC, Hartman E, Blanc L, Peters LL. Differential effects of RASA3 mutations on hematopoiesis are profoundly influenced by genetic background and molecular variant. PLoS Genet 2020; 16:e1008857. [PMID: 33370780 PMCID: PMC7793307 DOI: 10.1371/journal.pgen.1008857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 01/08/2021] [Accepted: 11/24/2020] [Indexed: 01/31/2023] Open
Abstract
Studies of the severely pancytopenic scat mouse model first demonstrated the crucial role of RASA3, a dual RAS and RAP GTPase activating protein (GAP), in hematopoiesis. RASA3 is required for survival in utero; germline deletion is lethal at E12.5–13.5 due to severe hemorrhage. Here, conditional deletion in hematopoietic stem and progenitor cells (HSPCs) using Vav-iCre recapitulates the null phenotype demonstrating that RASA3 is required at the stem and progenitor level to maintain blood vessel development and integrity and effective blood production. In adults, bone marrow blood cell production and spleen stress erythropoiesis are suppressed significantly upon induction of RASA3 deficiency, leading to pancytopenia and death within two weeks. Notably, RASA3 missense mutations in two mouse models, scat (G125V) and hlb381 (H794L), show dramatically different hematopoietic consequences specific to both genetic background and molecular variant. The mutation effect is mediated at least in part by differential effects on RAS and RAP activation. In addition, we show that the role of RASA3 is conserved during human terminal erythropoiesis, highlighting a potential function for the RASA3-RAS axis in disordered erythropoiesis in humans. Finally, global transcriptomic studies in scat suggest potential targets to ameliorate disease progression. Hematopoiesis is the process by which blood cells are formed. An individual must have a normal complement of red blood cells to prevent anemia, platelets to control bleeding, and white blood cells to maintain immune functions. All blood cells are derived from hematopoietic stem cells that differentiate into progenitor cells that then develop into mature circulating cells. We studied several mouse strains carrying different mutations in the gene encoding RASA3 and human CD34+ cells, which can be induced to produce blood cells in culture. We show that RASA3 is required at the earliest stages of blood formation, the stem and progenitor cells, and that the complement of genes other than RASA3, or the genetic background, profoundly alters the overall effect on blood formation. Further, the molecular nature of the mutation in RASA3 also has a profound and independent effect on overall blood formation. One mutant mouse strain, designated scat, suffers cyclic anemia characterized by severe anemic crisis episodes interspersed with remissions where the anemia significantly improves. Comparison of scat crisis and remission hematopoietic stem and progenitor cells reveals striking differences in gene expression. Analyses of these expression differences provide clues to processes that potentially drive improvement of anemia in scat and provide new avenues to pursue in future studies to identify novel therapeutics for anemia.
Collapse
Affiliation(s)
| | | | - Joel H. Graber
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine, United States of America
| | - Yue Zhao
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Amy J. Lambert
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Babette Gwynn
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Nathaniel J. Maki
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine, United States of America
| | - Elena C. Brindley
- Feinstein Institutes for Medical Research, Manhasset, New York, United States of America
| | - Emily Hartman
- Feinstein Institutes for Medical Research, Manhasset, New York, United States of America
| | - Lionel Blanc
- Feinstein Institutes for Medical Research, Manhasset, New York, United States of America
- * E-mail: (LB); (LLP)
| | - Luanne L. Peters
- The Jackson Laboratory, Bar Harbor, Maine, United States of America
- * E-mail: (LB); (LLP)
| |
Collapse
|
35
|
Venkatesan V, Srinivasan S, Babu P, Thangavel S. Manipulation of Developmental Gamma-Globin Gene Expression: an Approach for Healing Hemoglobinopathies. Mol Cell Biol 2020; 41:e00253-20. [PMID: 33077498 PMCID: PMC7849396 DOI: 10.1128/mcb.00253-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
β-Hemoglobinopathies are the most common monogenic disorders, and a century of research has provided us with a better understanding of the attributes of these diseases. Allogenic stem cell transplantation was the only potentially curative option available for these diseases until the discovery of gene therapy. The findings on the protective nature of fetal hemoglobin in sickle cell disease (SCD) and thalassemia patients carrying hereditary persistence of fetal hemoglobin (HPFH) mutations has given us the best evidence that the cure for β-hemoglobinopathies remains hidden in the hemoglobin locus. The detailed understanding of the developmental gene regulation of gamma-globin (γ-globin) and the emergence of gene manipulation strategies offer us the opportunity for developing a γ-globin gene-modified autologous stem cell transplantation therapy. In this review, we summarize different therapeutic strategies that reactivate fetal hemoglobin for the gene therapy of β-hemoglobinopathies.
Collapse
Affiliation(s)
- Vigneshwaran Venkatesan
- Centre for Stem Cell Research (CSCR), InStem Bengaluru, Christian Medical College, Vellore, Tamil Nadu, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Saranya Srinivasan
- Centre for Stem Cell Research (CSCR), InStem Bengaluru, Christian Medical College, Vellore, Tamil Nadu, India
| | - Prathibha Babu
- Centre for Stem Cell Research (CSCR), InStem Bengaluru, Christian Medical College, Vellore, Tamil Nadu, India
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Saravanabhavan Thangavel
- Centre for Stem Cell Research (CSCR), InStem Bengaluru, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
36
|
J. Verheul TC, Trinh VT, Vázquez O, Philipsen S. Targeted Protein Degradation as a Promising Tool for Epigenetic Upregulation of Fetal Hemoglobin. ChemMedChem 2020; 15:2436-2443. [PMID: 33002296 PMCID: PMC7756256 DOI: 10.1002/cmdc.202000574] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/28/2020] [Indexed: 12/17/2022]
Abstract
The level of fetal hemoglobin (HbF) is an important disease modifier for β-thalassemia and sickle cell disease patients. Indeed, genetic tinkering with the HbF repression machinery has demonstrated great potential for disease mitigation. Such genetic treatments are costly and the high incidence of β-hemoglobinopathies in low-income countries, therefore, calls for the development of affordable, off-the-shelf, oral treatments. The use of PROTAC (PRoteolysis TArgeting Chimeras) technology to influence the epigenetic mechanisms involved in HbF suppression may provide a solution. In this minireview, we briefly explain the HbF repression network highlighting the epigenetic factors that could be targeted for degradation by PROTACs. We hope that this review will inspire clinicians, molecular and chemical biologists to collaborate and contribute to this fascinating field, which should ultimately deliver drugs that reactivate HbF expression with high specificity and low toxicity.
Collapse
Affiliation(s)
- Thijs C. J. Verheul
- Department of Cell BiologyErasmus University Medical Center RotterdamWytemaweg 803000 CARotterdamThe Netherlands
| | - Van Tuan Trinh
- Department of ChemistryUniversity of MarburgHans-Meerwein-Straβe 435043MarburgGermany
| | - Olalla Vázquez
- SYNMIKRO Research CenterUniversity of Marburg35043MarburgGermany
- Department of ChemistryUniversity of MarburgHans-Meerwein-Straβe 435043MarburgGermany
| | - Sjaak Philipsen
- Department of Cell BiologyErasmus University Medical Center RotterdamWytemaweg 803000 CARotterdamThe Netherlands
| |
Collapse
|
37
|
Abstract
Fetal hemoglobin (HbF) can blunt the pathophysiology, temper the clinical course, and offer prospects for curative therapy of sickle cell disease. This review focuses on (1) HbF quantitative trait loci and the geography of β-globin gene haplotypes, especially those found in the Middle East; (2) how HbF might differentially impact the pathophysiology and many subphenotypes of sickle cell disease; (3) clinical implications of person-to-person variation in the distribution of HbF among HbF-containing erythrocytes; and (4) reactivation of HbF gene expression using both pharmacologic and cell-based therapeutic approaches. A confluence of detailed understanding of the molecular basis of HbF gene expression, coupled with the ability to precisely target by genomic editing most areas of the genome, is producing important preliminary therapeutic results that could provide new options for cell-based therapeutics with curative intent.
Collapse
Affiliation(s)
- Martin H Steinberg
- Division of Hematology/Oncology, Department of Medicine, Center of Excellence for Sickle Cell Disease, Center for Regenerative Medicine, Genome Science Institute, Boston University School of Medicine and Boston Medical Center, Boston, MA
| |
Collapse
|
38
|
Papayannopoulou T. Control of fetal globin expression in man: new opportunities to challenge past discoveries. Exp Hematol 2020; 92:43-50. [PMID: 32976950 DOI: 10.1016/j.exphem.2020.09.195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 01/01/2023]
Abstract
Decades-old findings supporting origin of F cells in adult life from adult-type progenitors and the in vitro and in vivo enhancement of fetal globin under stress conditions have been juxtaposed against recent mechanistic underpinnings. An updated molecular interrogation did not debunk prior conclusions on the origin of F cells. Although fetal globin reactivation by widely diverse approaches in vitro and in response to anemic stresses in vivo is a work in progress, accumulating evidence converges toward an integrated stress response pathway. The newly uncovered developmental regulators of globin gene switching not only have provided answers to the long-awaited quest of transregulation of switching, they are also reaching a clinical threshold. Although the effect of fetal globin silencers has been robustly validated in adult cells, the response of cells at earlier developmental stages has been unclear and inadequately studied.
Collapse
|
39
|
Verma R. Molecular Pathways Engaged by Immunomodulatory Agents in Monoclonal Gammopathy-Associated Pure Red Cell Aplasia Rescue. Front Oncol 2020; 10:1490. [PMID: 33014791 PMCID: PMC7493653 DOI: 10.3389/fonc.2020.01490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/13/2020] [Indexed: 11/26/2022] Open
Affiliation(s)
- Rakesh Verma
- Yale Cancer Center, Yale University, New Haven, CT, United States
| |
Collapse
|
40
|
Amjad F, Fatima T, Fayyaz T, Khan MA, Qadeer MI. Novel genetic therapeutic approaches for modulating the severity of β-thalassemia (Review). Biomed Rep 2020; 13:48. [PMID: 32953110 PMCID: PMC7484974 DOI: 10.3892/br.2020.1355] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
Thalassemia is a genetic haematological disorder that arises due to defects in the α and β-globin genes. Worldwide, 0.3-0.4 million children are born with haemoglobinopathies per year. Thalassemic patients, as well as their families, face various serious clinical, socio-economic, and psychosocial challenges throughout their life. Different therapies are available in clinical practice to minimize the suffering of thalassemic patients to some extent and potentially cure the disease. Predominantly, patients undergo transfusion therapy to maintain their haemoglobin levels. Due to multiple transfusions, the iron levels in their bodies are elevated. Iron overload results in damage to body organs, resulting in heart failure, liver function failure or endocrine failure, all of which are commonly observed. Certain drugs have been developed to enhance the expression of the γ-gene, which ultimately results in augmentation of fetal haemoglobin (HbF) levels and total haemoglobin levels in the body. However, its effectiveness is dependent on the genetic makeup of the individual patient. At present, allogeneic haematopoietic Stem Cell Transplantation (HSCT) is the only practically available option with a high curative rate. However, the outcome of HSCT is strongly influenced by factors such as age at transplantation, irregular iron chelation history before transplantation, histocompatibility, and source of stem cells. Gene therapy using the lentiglobin vector is the most recent method for cure without any mortality, graft rejection and clonal dominance issues. However, delayed platelet engraftment is being reported in some patients. Genome editing is a novel approach which may be used to treat patients with thalassemia; it makes use of targeted nucleases to correct the mutations in specific DNA sequences and modify the sequence to the normal wild-type sequence. To edit the genome at the required sites, CRISPR/Cas9 is an efficient and accurate tool that is used in various genetic engineering programs. Genome editing mediated by CRISPR/Cas9 has the ability to restore the normal β-globin function with minimal side effects. Using CRISPR/Cas9, expression of BCL11A can be downregulated along with increased production of HbF. However, these genome editing tools are still under in-vitro trials. CRISPR/Cas9 has can be used for precise transcriptional regulation, genome modification and epigenetic editing. Additional research is required in this regard, as CRISPR/Cas9 may potentially exhibit off-target activity and there are legal and ethical considerations regarding its use.
Collapse
Affiliation(s)
- Fareeha Amjad
- Department of Microbiology and Molecular Genetics, University of The Punjab, Lahore, Punjab 54590, Pakistan
| | - Tamseel Fatima
- Department of Microbiology and Molecular Genetics, University of The Punjab, Lahore, Punjab 54590, Pakistan
| | - Tuba Fayyaz
- Department of Microbiology and Molecular Genetics, University of The Punjab, Lahore, Punjab 54590, Pakistan
| | - Muhammad Aslam Khan
- Sundas Molecular Analysis Centre (SUNMAC), Sundas Foundation, Lahore, Punjab 54000, Pakistan
| | - Muhammad Imran Qadeer
- Department of Microbiology and Molecular Genetics, University of The Punjab, Lahore, Punjab 54590, Pakistan.,Sundas Molecular Analysis Centre (SUNMAC), Sundas Foundation, Lahore, Punjab 54000, Pakistan
| |
Collapse
|
41
|
El Hoss S, Cochet S, Godard A, Yan H, Dussiot M, Frati G, Boutonnat-Faucher B, Laurance S, Renaud O, Joseph L, Miccio A, Brousse V, Mohandas N, El Nemer W. Fetal hemoglobin rescues ineffective erythropoiesis in sickle cell disease. Haematologica 2020; 106:2707-2719. [PMID: 32855279 PMCID: PMC8485663 DOI: 10.3324/haematol.2020.265462] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Indexed: 11/09/2022] Open
Abstract
While ineffective erythropoiesis has long been recognized as a key contributor to anemia in thalassemia, its role in anemia of sickle cell disease (SCD) has not been critically explored. Using in vitro and in vivo derived human erythroblasts we assessed the extent of ineffective erythropoiesis in SCD. Modeling the bone marrow hypoxic environment, we found that hypoxia induces death of sickle erythroblasts starting at the polychromatic stage, positively selecting cells with high levels of fetal hemoglobin (HbF). Cell death was associated with cytoplasmic sequestration of heat shock protein 70 and was rescued by induction of HbF synthesis. Importantly, we document that in bone marrow of SCD patients similar cell loss occurs during the final stages of terminal differentiation. Our study provides evidence for ineffective erythropoiesis in SCD and highlights an anti-apoptotic role for HbF during the terminal stages of erythroid differentiation. These findings imply that the beneficial effect on anemia of increased HbF levels is not only due to the increased life span of red cells but also a consequence of decreased ineffective erythropoiesis.
Collapse
Affiliation(s)
- Sara El Hoss
- Université de Paris, INSERM UMR_S 1134, Paris, France
| | | | - Auria Godard
- Inserm - INTS - University of Paris, Paris, France
| | - Hongxia Yan
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY, USA
| | - Michaël Dussiot
- Imagine Institute, Université de Paris, Hopital Necker Enfants Malades, Paris, France
| | - Giacomo Frati
- Université de Paris, Imagine Institute, Paris, France
| | | | | | - Olivier Renaud
- Institut Curie, Paris Sciences et Lettres Research University, Paris, France
| | - Laure Joseph
- Service de biotherapie, Hôpital Universitaire Necker Enfants Malades, Paris, France
| | | | | | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY, USA
| | | |
Collapse
|
42
|
The E3 ligase adaptor molecule SPOP regulates fetal hemoglobin levels in adult erythroid cells. Blood Adv 2020; 3:1586-1597. [PMID: 31126914 DOI: 10.1182/bloodadvances.2019032318] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/23/2019] [Indexed: 12/21/2022] Open
Abstract
Reactivation of fetal hemoglobin (HbF) production benefits patients with sickle cell disease and β-thalassemia. To identify new HbF regulators that might be amenable to pharmacologic control, we screened a protein domain-focused CRISPR-Cas9 library targeting chromatin regulators, including BTB domain-containing proteins. Speckle-type POZ protein (SPOP), a substrate adaptor of the CUL3 ubiquitin ligase complex, emerged as a novel HbF repressor. Depletion of SPOP or overexpression of a dominant negative version significantly raised fetal globin messenger RNA and protein levels with minimal detrimental effects on normal erythroid maturation, as determined by transcriptome and proteome analyses. SPOP controls HbF expression independently of the major transcriptional HbF repressors BCL11A and LRF. Finally, pharmacologic HbF inducers cooperate with SPOP depletion during HbF upregulation. Our study implicates SPOP and the CUL3 ubiquitin ligase system in controlling HbF production in human erythroid cells and may offer new therapeutic strategies for the treatment of β-hemoglobinopathies.
Collapse
|
43
|
Lamsfus-Calle A, Daniel-Moreno A, Antony JS, Epting T, Heumos L, Baskaran P, Admard J, Casadei N, Latifi N, Siegmund DM, Kormann MSD, Handgretinger R, Mezger M. Comparative targeting analysis of KLF1, BCL11A, and HBG1/2 in CD34 + HSPCs by CRISPR/Cas9 for the induction of fetal hemoglobin. Sci Rep 2020; 10:10133. [PMID: 32576837 PMCID: PMC7311455 DOI: 10.1038/s41598-020-66309-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 05/19/2020] [Indexed: 12/22/2022] Open
Abstract
β-hemoglobinopathies are caused by abnormal or absent production of hemoglobin in the blood due to mutations in the β-globin gene (HBB). Imbalanced expression of adult hemoglobin (HbA) induces strong anemia in patients suffering from the disease. However, individuals with natural-occurring mutations in the HBB cluster or related genes, compensate this disparity through γ-globin expression and subsequent fetal hemoglobin (HbF) production. Several preclinical and clinical studies have been performed in order to induce HbF by knocking-down genes involved in HbF repression (KLF1 and BCL11A) or disrupting the binding sites of several transcription factors in the γ-globin gene (HBG1/2). In this study, we thoroughly compared the different CRISPR/Cas9 gene-disruption strategies by gene editing analysis and assessed their safety profile by RNA-seq and GUIDE-seq. All approaches reached therapeutic levels of HbF after gene editing and showed similar gene expression to the control sample, while no significant off-targets were detected by GUIDE-seq. Likewise, all three gene editing platforms were established in the GMP-grade CliniMACS Prodigy, achieving similar outcome to preclinical devices. Based on this gene editing comparative analysis, we concluded that BCL11A is the most clinically relevant approach while HBG1/2 could represent a promising alternative for the treatment of β-hemoglobinopathies.
Collapse
Affiliation(s)
- Andrés Lamsfus-Calle
- University Children's Hospital. Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Alberto Daniel-Moreno
- University Children's Hospital. Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Justin S Antony
- University Children's Hospital. Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Thomas Epting
- Institute for Clinical Chemistry and Laboratory Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas Heumos
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Praveen Baskaran
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Ngadhnjim Latifi
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Darina M Siegmund
- University Hospital Freiburg. Department of Hematology, Oncology, and Stem-Cell Transplantation, Medical Center, University of Freiburg, Freiburg, Germany
| | - Michael S D Kormann
- University Children's Hospital. Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- University Children's Hospital. Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany
| | - Markus Mezger
- University Children's Hospital. Department of Pediatrics I, Hematology and Oncology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
44
|
UNC0638 induces high levels of fetal hemoglobin expression in β-thalassemia/HbE erythroid progenitor cells. Ann Hematol 2020; 99:2027-2036. [PMID: 32567028 DOI: 10.1007/s00277-020-04136-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/09/2020] [Indexed: 12/23/2022]
Abstract
Increased expression of fetal hemoglobin (HbF) improves the clinical severity of β-thalassemia patients. EHMT1/2 histone methyltransferases are epigenetic modifying enzymes that are responsible for catalyzing addition of the repressive histone mark H3K9me2 at silenced genes, including the γ-globin genes. UNC0638, a chemical inhibitor of EHMT1/2, has been shown to induce HbF expression in human erythroid progenitor cell cultures. Here, we report the HbF-inducing activity of UNC0638 in erythroid progenitor cells from β-thalassemia/HbE patients. UNC0638 treatment led to significant increases in γ-globin mRNA, HbF expression, and HbF-containing cells in the absence of significant cytotoxicity. Moreover, UNC0638 showed additive effects on HbF induction in combination with the immunomodulatory drug pomalidomide and the DNMT1 inhibitor decitabine. These studies provide a scientific proof of concept that a small molecule targeting EHMT1/2 epigenetic enzymes, used alone or in combination with pomalidomide or decitabine, is a potential therapeutic approach for HbF induction. Further development of structural analogs of UNC0638 with similar biological effects but improved pharmacokinetic properties may lead to promising therapies and possible clinical application for the treatment of β-thalassemia.
Collapse
|
45
|
Khandros E, Huang P, Peslak SA, Sharma M, Abdulmalik O, Giardine BM, Zhang Z, Keller CA, Hardison RC, Blobel GA. Understanding heterogeneity of fetal hemoglobin induction through comparative analysis of F and A erythroblasts. Blood 2020; 135:1957-1968. [PMID: 32268371 PMCID: PMC7256358 DOI: 10.1182/blood.2020005058] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/05/2020] [Indexed: 01/27/2023] Open
Abstract
Reversing the developmental switch from fetal hemoglobin (HbF, α2γ2) to adult hemoglobin (HbA, α2β2) is an important therapeutic approach in sickle cell disease (SCD) and β-thalassemia. In healthy individuals, SCD patients, and patients treated with pharmacologic HbF inducers, HbF is present only in a subset of red blood cells known as F cells. Despite more than 50 years of observations, the cause for this heterocellular HbF expression pattern, even among genetically identical cells, remains unknown. Adult F cells might represent a reversion of a given cell to a fetal-like epigenetic and transcriptional state. Alternatively, isolated transcriptional or posttranscriptional events at the γ-globin genes might underlie heterocellularity. Here, we set out to understand the heterogeneity of HbF activation by developing techniques to purify and profile differentiation stage-matched late erythroblast F cells and non-F cells (A cells) from the human HUDEP2 erythroid cell line and primary human erythroid cultures. Transcriptional and proteomic profiling of these cells demonstrated very few differences between F and A cells at the RNA level either under baseline conditions or after treatment with HbF inducers hydroxyurea or pomalidomide. Surprisingly, we did not find differences in expression of any known HbF regulators, including BCL11A or LRF, that would account for HbF activation. Our analysis shows that F erythroblasts are not significantly different from non-HbF-expressing cells and that the primary differences likely occur at the transcriptional level at the β-globin locus.
Collapse
Affiliation(s)
- Eugene Khandros
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Peng Huang
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Scott A Peslak
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Medicine, Division of Hematology/Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Malini Sharma
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Osheiza Abdulmalik
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Belinda M Giardine
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA; and
| | - Zhe Zhang
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Cheryl A Keller
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA; and
| | - Ross C Hardison
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA; and
| | - Gerd A Blobel
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
46
|
Khamphikham P, Nualkaew T, Pongpaksupasin P, Kaewsakulthong W, Songdej D, Paiboonsukwong K, Engel JD, Hongeng S, Fucharoen S, Sripichai O, Jearawiriyapaisarn N. High-level induction of fetal haemoglobin by pomalidomide in β-thalassaemia/HbE erythroid progenitor cells. Br J Haematol 2020; 189:e240-e245. [PMID: 32358840 DOI: 10.1111/bjh.16670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Pinyaphat Khamphikham
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.,Department of Forensic Science, Faculty of Allied Health Sciences, Thammasat University, Pathum Thani, Thailand
| | - Tiwaporn Nualkaew
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Phitchapa Pongpaksupasin
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.,Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Woratree Kaewsakulthong
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.,Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Duantida Songdej
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kittiphong Paiboonsukwong
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - James D Engel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Orapan Sripichai
- Department of Medical Sciences, National Institute of Health, Ministry of Public Health, Nonthaburi, Thailand
| | - Natee Jearawiriyapaisarn
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW The current review focuses on recent insights into the development of small molecule therapeutics to treat the β-globinopathies. RECENT FINDINGS Recent studies of fetal γ-globin gene regulation reveal multiple insights into how γ-globin gene reactivation may lead to novel treatment for β-globinopathies. SUMMARY We summarize current information regarding the binding of transcription factors that appear to be impeded or augmented by different hereditary persistence of fetal hemoglobin (HPFH) mutations. As transcription factors have historically proven to be difficult to target for therapeutic purposes, we next address the contributions of protein complexes associated with these HPFH mutation-affected transcription factors with the aim of defining proteins that might provide additional targets for chemical molecules to inactivate the corepressors. Among the enzymes associated with the transcription factor complexes, a group of corepressors with currently available inhibitors were initially thought to be good candidates for potential therapeutic purposes. We discuss possibilities for pharmacological inhibition of these corepressor enzymes that might significantly reactivate fetal γ-globin gene expression. Finally, we summarize the current clinical trial data regarding the inhibition of select corepressor proteins for the treatment of sickle cell disease and β-thalassemia.
Collapse
Affiliation(s)
- Lei Yu
- Departments of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109
| | - Greggory Myers
- Departments of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109
| | - James Douglas Engel
- Departments of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, Michigan 48109
| |
Collapse
|
48
|
Fong C, Mendoza Y, Barreto G. Genetic variants in the G gamma-globin promoter modulate fetal hemoglobin expression in the Colombian population. Genet Mol Biol 2020; 43:e20190076. [PMID: 32325484 PMCID: PMC7210980 DOI: 10.1590/1678-4685-gmb-2019-0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 11/28/2019] [Indexed: 11/23/2022] Open
Abstract
Fetal hemoglobin (HbF) is a determining factor for the development of sickle cell anemia. High HbF levels lower the intensity of symptoms of this disease. HbF levels can vary in patients with sickle cell anemia and individuals without the disease. The purpose of this study was to identify the genetic variants in the G gamma-globin gene promoter that can modulate HbF expression in patients with sickle cell anemia and healthy individuals from Colombia. In total, 413 bp of the G gamma-globin gene promoter were sequenced in 60 patients with sickle cell anemia and 113 healthy individuals. The allelic and genotype frequencies of the identified variants were compared between individuals with low and high HbF for both patients and healthy individuals. In total, we identified 15 variants in both groups, only three of which were shared between patients and healthy individuals. In healthy individuals, sites -16 and -309 (rs112479156) exhibited differences in allele frequencies. The mutant allele of -16 lowered the production of HbF, whereas the mutant allele of -309 increased its production. These results reveal the presence of different mechanisms of HbF regulation between patients with sickle cell and healthy individuals.
Collapse
Affiliation(s)
- Cristian Fong
- GIOD Group, Faculty of Dentistry, Universidad Cooperativa de Colombia, Pasto, Nariño, Colombia
| | - Yesica Mendoza
- Human Molecular Genetics Group, Biology Department, Universidad del Valle, Cali, Valle del Cauca, Colombia
| | - Guillermo Barreto
- Human Molecular Genetics Group, Biology Department, Universidad del Valle, Cali, Valle del Cauca, Colombia
| |
Collapse
|
49
|
Narla A, Mohandas N. Staying hydrated is important also for erythroblasts. Haematologica 2020; 105:528-529. [PMID: 32115411 DOI: 10.3324/haematol.2019.233999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Anupama Narla
- Department of Pediatrics, Stanford University, School of Medicine, Stanford, CA
| | - Narla Mohandas
- Laboratory of Red Cell Physiology, New York Blood Center, New York, NY, USA
| |
Collapse
|
50
|
Heme-regulated eIF2α kinase in erythropoiesis and hemoglobinopathies. Blood 2020; 134:1697-1707. [PMID: 31554636 DOI: 10.1182/blood.2019001915] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
As essential components of hemoglobin, iron and heme play central roles in terminal erythropoiesis. The impairment of this process in iron/heme deficiency results in microcytic hypochromic anemia, the most prevalent anemia globally. Heme-regulated eIF2α kinase, also known as heme-regulated inhibitor (HRI), is a key heme-binding protein that senses intracellular heme concentrations to balance globin protein synthesis with the amount of heme available for hemoglobin production. HRI is activated during heme deficiency to phosphorylate eIF2α (eIF2αP), which simultaneously inhibits the translation of globin messenger RNAs (mRNAs) and selectively enhances the translation of activating transcription factor 4 (ATF4) mRNA to induce stress response genes. This coordinated translational regulation is a universal hallmark across the eIF2α kinase family under various stress conditions and is termed the integrated stress response (ISR). Inhibition of general protein synthesis by HRI-eIF2αP in erythroblasts is necessary to prevent proteotoxicity and maintain protein homeostasis in the cytoplasm and mitochondria. Additionally, the HRI-eIF2αP-ATF4 pathway represses mechanistic target of rapamycin complex 1 (mTORC1) signaling, specifically in the erythroid lineage as a feedback mechanism of erythropoietin-stimulated erythropoiesis during iron/heme deficiency. Furthermore, ATF4 target genes are most highly activated during iron deficiency to maintain mitochondrial function and redox homeostasis, as well as to enable erythroid differentiation. Thus, heme and translation regulate erythropoiesis through 2 key signaling pathways, ISR and mTORC1, which are coordinated by HRI to circumvent ineffective erythropoiesis (IE). HRI-ISR is also activated to reduce the severity of β-thalassemia intermedia in the Hbbth1/th1 murine model. Recently, HRI has been implicated in the regulation of human fetal hemoglobin production. Therefore, HRI-ISR has emerged as a potential therapeutic target for hemoglobinopathies.
Collapse
|