1
|
Jeong TK, Frater RCM, Yoon J, Groth A, Song JJ. CODANIN-1 sequesters ASF1 by using a histone H3 mimic helix to regulate the histone supply. Nat Commun 2025; 16:2181. [PMID: 40038274 DOI: 10.1038/s41467-025-56976-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/06/2025] [Indexed: 03/06/2025] Open
Abstract
ASF1 is a major histone chaperone that regulates the supply of histone H3-H4 and facilitates nucleosome assembly to maintain chromatin structure during DNA replication and transcription. CODANIN-1 negatively regulates the function of ASF1. However, the molecular mechanism by which CODANIN-1 inhibits the ASF1-mediated histone supply remains elusive. Here, we present the cryo-EM structure of a human CODANIN-1_ASF1A complex at 3.75 Å resolution. The structure reveals that CODANIN-1 forms a dimer where each monomer holds two ASF1 molecules, utilizing two B-domains and two histone H3 mimic helices (HMHs). The interaction of CODANIN-1 with ASF1 via the HMH and B-domains inhibits the formation of an ASF1/H3-H4 complex and sequesters ASF1 in the cytoplasm. Our study provides a structural and molecular basis for the function of CODANIN-1 as negative regulator that highjacks ASF1 interaction sites with histones and downstream chaperones to inhibit nucleosome assembly.
Collapse
Affiliation(s)
- Tae-Kyeong Jeong
- Department of Biological Sciences, KI for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - R Ciaran MacKenzie Frater
- The Novo Nordisk Foundation Center for Protein Research (CPR), University of Copenhagen, Copenhagen, Denmark
| | - Jongha Yoon
- Department of Biological Sciences, KI for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Anja Groth
- The Novo Nordisk Foundation Center for Protein Research (CPR), University of Copenhagen, Copenhagen, Denmark.
- Biotech Research & Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
| | - Ji-Joon Song
- Department of Biological Sciences, KI for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
| |
Collapse
|
2
|
Xu Y, Jia P, Li Y, Zhang H, Zhang J, Li W, Zhen Y, Li Y, Cao J, Zheng T, Wang Y, Liu Y, An X, Zhang S. A novel role of AURKA kinase in erythroblast enucleation. Haematologica 2024; 109:3721-3734. [PMID: 38961734 PMCID: PMC11532702 DOI: 10.3324/haematol.2023.284873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 06/07/2024] [Indexed: 07/05/2024] Open
Abstract
Generation of mammalian red blood cells requires the expulsion of polarized nuclei late in terminal erythroid differentiation. However, the mechanisms by which spherical erythroblasts determine the direction of nuclear polarization and maintain asymmetry during nuclear expulsion are poorly understood. Given the analogy of erythroblast enucleation to asymmetric cell division and the key role of Aurora kinases in mitosis, we sought to investigate the function of Aurora kinases in erythroblast enucleation. We found that AURKA (Aurora kinase A) is abundantly expressed in orthochromatic erythroblasts. Intriguingly, high-resolution confocal microscopy analyses revealed that AURKA co-localized with the centrosome on the side of the nucleus opposite its membrane contact point during polarization and subsequently translocated to the anterior end of the protrusive nucleus upon nuclear exit. Mechanistically, AURKA regulated centrosome maturation and localization via interaction with γ-tubulin to provide polarization orientation for the nucleus. Furthermore, we identified ECT2 (epithelial cell transforming 2), a guanine nucleotide exchange factor, as a new interacting protein and ubiquitination substrate of AURKA. After forming the nuclear protrusion, AURKA translocated to the anterior end of the protrusive nucleus to directly degrade ECT2, which is partly dependent on kinase activity of AURKA. Moreover, knockdown of ECT2 rescued impaired enucleation caused by AURKA inhibition. Our findings have uncovered a previously unrecognized role of Aurora kinases in the establishment of nuclear polarization and eventual nuclear extrusion and provide new mechanistic insights into erythroblast enucleation.
Collapse
Affiliation(s)
- Yuanlin Xu
- School of Life Sciences, Zhengzhou University, Zhengzhou, China; Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Yating Li
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Huan Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Jingxin Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Wanxin Li
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Yazhe Zhen
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Yan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Jiaming Cao
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Tingting Zheng
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Yihan Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou
| | - Yanyan Liu
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY, USA.
| | - Shijie Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou.
| |
Collapse
|
3
|
Newton LM, Fowler VM, Humbert PO. Erythroblast enucleation at a glance. J Cell Sci 2024; 137:jcs261673. [PMID: 39397781 PMCID: PMC11529606 DOI: 10.1242/jcs.261673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Erythroid enucleation, the penultimate step in mammalian erythroid terminal differentiation, is a unique cellular process by which red blood cells (erythrocytes) remove their nucleus and accompanying nuclear material. This complex, multi-stage event begins with chromatin compaction and cell cycle arrest and ends with generation of two daughter cells: a pyrenocyte, which contains the expelled nucleus, and an anucleate reticulocyte, which matures into an erythrocyte. Although enucleation has been compared to asymmetric cell division (ACD), many mechanistic hallmarks of ACD appear to be absent. Instead, enucleation appears to rely on mechanisms borrowed from cell migration, endosomal trafficking and apoptosis, as well as unique cellular interactions within the microenvironment. In this Cell Science at a Glance article and the accompanying poster, we summarise current insights into the morphological features and genetic drivers regulating the key intracellular events that culminate in erythroid enucleation and engulfment of pyrenocytes by macrophages within the bone marrow microenvironment.
Collapse
Affiliation(s)
- Lucas M. Newton
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, VIC 3073, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3073, Australia
| | - Velia M. Fowler
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Patrick O. Humbert
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, VIC 3073, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3073, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
4
|
Hu X, Wang J, Yang K, Fan H, Wu J, Ren J, Han G, Li J, Xue Z, Liu X, Lv X. The GWAS SNP rs80207740 modulates erythrocyte traits via allele-specific binding of IKZF1 and targeting XPO7 gene. FASEB J 2024; 38:e23666. [PMID: 38780091 DOI: 10.1096/fj.202302017r] [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: 10/04/2023] [Revised: 03/31/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Genome-wide association studies have identified many single nucleotide polymorphisms (SNPs) associated with erythrocyte traits. However, the functional variants and their working mechanisms remain largely unknown. Here, we reported that the SNP of rs80207740, which was associated with red blood cell (RBC) volume and hemoglobin content across populations, conferred enhancer activity to XPO7 gene via allele-differentially binding to Ikaros family zinc finger 1 (IKZF1). We showed that the region around rs80207740 was an erythroid-specific enhancer using reporter assays, and that the G-allele further enhanced activity. 3D genome evidence showed that the enhancer interacted with the XPO7 promoter, and eQTL analysis suggested that the G-allele upregulated expression of XPO7. We further showed that the rs80207740-G allele facilitated the binding of transcription factor IKZF1 in EMSA and ChIP analyses. Knockdown of IKZF1 and GATA1 resulted in decreased expression of Xpo7 in both human and mouse erythroid cells. Finally, we constructed Xpo7 knockout mouse by CRISPR/Cas9 and observed anemic phenotype with reduced volume and hemoglobin content of RBC, consistent to the effect of rs80207740 on erythrocyte traits. Overall, our study demonstrated that rs80207740 modulated erythroid indices by regulating IKZF1 binding and Xpo7 expression.
Collapse
Affiliation(s)
- Xinjun Hu
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Jiaxin Wang
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Ke Yang
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Hong Fan
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Jie Wu
- Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jiuqiang Ren
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Gaijing Han
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Jing Li
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Zheng Xue
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Xuehui Liu
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Xiang Lv
- State Key Laboratory of Complex, Severe, and Rare Diseases, Haihe Laboratory of Cell Ecosystem, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
5
|
Ju H, Sohn Y, Nam Y, Rim YA. Progresses in overcoming the limitations of in vitro erythropoiesis using human induced pluripotent stem cells. Stem Cell Res Ther 2024; 15:142. [PMID: 38750578 PMCID: PMC11094930 DOI: 10.1186/s13287-024-03754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/04/2024] [Indexed: 05/19/2024] Open
Abstract
Researchers have attempted to generate transfusable oxygen carriers to mitigate RBC supply shortages. In vitro generation of RBCs using stem cells such as hematopoietic stem and progenitor cells (HSPCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs) has shown promise. Specifically, the limited supplies of HSPCs and ethical issues with ESCs make iPSCs the most promising candidate for in vitro RBC generation. However, researchers have encountered some major challenges when using iPSCs to produce transfusable RBC products, such as enucleation and RBC maturation. In addition, it has proven difficult to manufacture these products on a large scale. In this review, we provide a brief overview of erythropoiesis and examine endeavors to recapitulate erythropoiesis in vitro using various cell sources. Furthermore, we explore the current obstacles and potential solutions aimed at enabling the large-scale production of transfusable RBCs in vitro.
Collapse
Affiliation(s)
- Hyeonwoo Ju
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
- YiPSCELL Inc., L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Yeri Alice Rim
- YiPSCELL Inc., L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, 06591, Republic of Korea.
- CiSTEM laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
6
|
Xiong D, Zhang L, Sun ZJ. Targeting the epigenome to reinvigorate T cells for cancer immunotherapy. Mil Med Res 2023; 10:59. [PMID: 38044445 PMCID: PMC10694991 DOI: 10.1186/s40779-023-00496-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/14/2023] [Indexed: 12/05/2023] Open
Abstract
Cancer immunotherapy using immune-checkpoint inhibitors (ICIs) has revolutionized the field of cancer treatment; however, ICI efficacy is constrained by progressive dysfunction of CD8+ tumor-infiltrating lymphocytes (TILs), which is termed T cell exhaustion. This process is driven by diverse extrinsic factors across heterogeneous tumor immune microenvironment (TIME). Simultaneously, tumorigenesis entails robust reshaping of the epigenetic landscape, potentially instigating T cell exhaustion. In this review, we summarize the epigenetic mechanisms governing tumor microenvironmental cues leading to T cell exhaustion, and discuss therapeutic potential of targeting epigenetic regulators for immunotherapies. Finally, we outline conceptual and technical advances in developing potential treatment paradigms involving immunostimulatory agents and epigenetic therapies.
Collapse
Affiliation(s)
- Dian Xiong
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, China
| | - Lu Zhang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430079, China.
- Department of Oral Maxillofacial-Head Neck Oncology, School and and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
7
|
Ginzburg Y, An X, Rivella S, Goldfarb A. Normal and dysregulated crosstalk between iron metabolism and erythropoiesis. eLife 2023; 12:e90189. [PMID: 37578340 PMCID: PMC10425177 DOI: 10.7554/elife.90189] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023] Open
Abstract
Erythroblasts possess unique characteristics as they undergo differentiation from hematopoietic stem cells. During terminal erythropoiesis, these cells incorporate large amounts of iron in order to generate hemoglobin and ultimately undergo enucleation to become mature red blood cells, ultimately delivering oxygen in the circulation. Thus, erythropoiesis is a finely tuned, multifaceted process requiring numerous properly timed physiological events to maintain efficient production of 2 million red blood cells per second in steady state. Iron is required for normal functioning in all human cells, the erythropoietic compartment consuming the majority in light of the high iron requirements for hemoglobin synthesis. Recent evidence regarding the crosstalk between erythropoiesis and iron metabolism sheds light on the regulation of iron availability by erythroblasts and the consequences of insufficient as well as excess iron on erythroid lineage proliferation and differentiation. In addition, significant progress has been made in our understanding of dysregulated iron metabolism in various congenital and acquired malignant and non-malignant diseases. Finally, we report several actual as well as theoretical opportunities for translating the recently acquired robust mechanistic understanding of iron metabolism regulation to improve management of patients with disordered erythropoiesis, such as anemia of chronic inflammation, β-thalassemia, polycythemia vera, and myelodysplastic syndromes.
Collapse
Affiliation(s)
- Yelena Ginzburg
- Division of Hematology and Medical Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Xiuli An
- LFKRI, New York Blood CenterNew YorkUnited States
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Cell and Molecular Biology affinity group (CAMB), University of PennsylvaniaPhiladelphiaUnited States
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics at the Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Penn Center for Musculoskeletal Disorders at the Children’s Hospital of PhiladelphiaPhiladelphiaUnited States
- Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at University of PennsylvaniaPhiladelphiaUnited States
- RNA Institute at University of PennsylvaniaPhiladelphiaUnited States
| | - Adam Goldfarb
- Department of Pathology, University of VirginiaCharlottesvilleUnited States
| |
Collapse
|
8
|
Penagos-Puig A, Claudio-Galeana S, Stephenson-Gussinye A, Jácome-López K, Aguilar-Lomas A, Chen X, Pérez-Molina R, Furlan-Magaril M. RNA polymerase II pausing regulates chromatin organization in erythrocytes. Nat Struct Mol Biol 2023; 30:1092-1104. [PMID: 37500929 DOI: 10.1038/s41594-023-01037-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/16/2023] [Indexed: 07/29/2023]
Abstract
Chicken erythrocytes are nucleated cells often considered to be transcriptionally inactive, although the epigenetic changes and chromatin remodeling that would mediate transcriptional repression and the extent of gene silencing during avian terminal erythroid differentiation are not fully understood. Here, we characterize the changes in gene expression, chromatin accessibility, genome organization and chromatin nuclear disposition during the terminal stages of erythropoiesis in chicken and uncover complex chromatin reorganization at different genomic scales. We observe a robust decrease in transcription in erythrocytes, but a set of genes maintains their expression, including genes involved in RNA polymerase II (Pol II) promoter-proximal pausing. Erythrocytes exhibit a reoriented nuclear architecture, with accessible chromatin positioned towards the nuclear periphery together with the paused RNA Pol II. In erythrocytes, chromatin domains are partially lost genome-wide, except at minidomains retained around paused promoters. Our results suggest that promoter-proximal pausing of RNA Pol II contributes to the transcriptional regulation of the erythroid genome and highlight the role of RNA polymerase in the maintenance of local chromatin organization.
Collapse
Affiliation(s)
- Andrés Penagos-Puig
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sherlyn Claudio-Galeana
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aura Stephenson-Gussinye
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Karina Jácome-López
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Amaury Aguilar-Lomas
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Xingqi Chen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rosario Pérez-Molina
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mayra Furlan-Magaril
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
9
|
Wu J, Moriwaki K, Asuka T, Nakai R, Kanda S, Taniguchi M, Sugiyama T, Yoshimura SI, Kunii M, Nagasawa T, Hosen N, Miyoshi E, Harada A. EHBP1L1, an apicobasal polarity regulator, is critical for nuclear polarization during enucleation of erythroblasts. Blood Adv 2023; 7:3382-3394. [PMID: 37042948 PMCID: PMC10345855 DOI: 10.1182/bloodadvances.2022008930] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/13/2023] [Accepted: 03/31/2023] [Indexed: 04/13/2023] Open
Abstract
Cell polarity, the asymmetric distribution of proteins and organelles, is permanently or transiently established in various cell types and plays an important role in many physiological events. epidermal growth factor receptor substrate 15 homology domain-binding protein 1-like 1 (EHBP1L1) is an adapter protein that is localized on recycling endosomes and regulates apical-directed transport in polarized epithelial cells. However, the role of EHBP1L1 in nonepithelial cells, remains unknown. Here, Ehbp1l1-/- mice showed impaired erythroblast enucleation. Further analyses showed that nuclear polarization before enucleation was impaired in Ehbp1l1-/- erythroblasts. It was also revealed that EHBP1L1 interactors Rab10, Bin1, and dynamin were involved in erythroblast enucleation. In addition, Ehbp1l1-/- erythrocytes exhibited stomatocytic morphology and dehydration. These defects in erythroid cells culminated in early postnatal anemic lethality in Ehbp1l1-/- mice. Moreover, we found the mislocalization of nuclei and mitochondria in the skeletal muscle cells of Ehbp1l1-/- mice, as observed in patients with centronuclear myopathy with genetic mutations in Bin1 or dynamin 2. Taken together, our findings indicate that the Rab8/10-EHBP1L1-Bin1-dynamin axis plays an important role in multiple cell polarity systems in epithelial and nonepithelial cells.
Collapse
Affiliation(s)
- Ji Wu
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kenta Moriwaki
- Department of Biochemistry, Toho University School of Medicine, Tokyo, Japan
| | - Tatsuya Asuka
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ritsuko Nakai
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Satoshi Kanda
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Manabu Taniguchi
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tatsuki Sugiyama
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Shin-ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masataka Kunii
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Naoki Hosen
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
10
|
Lee SJ, Jung C, Oh JE, Kim S, Lee S, Lee JY, Yoon YS. Generation of Red Blood Cells from Human Pluripotent Stem Cells-An Update. Cells 2023; 12:1554. [PMID: 37296674 PMCID: PMC10253210 DOI: 10.3390/cells12111554] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Red blood cell (RBC) transfusion is a lifesaving medical procedure that can treat patients with anemia and hemoglobin disorders. However, the shortage of blood supply and risks of transfusion-transmitted infection and immune incompatibility present a challenge for transfusion. The in vitro generation of RBCs or erythrocytes holds great promise for transfusion medicine and novel cell-based therapies. While hematopoietic stem cells and progenitors derived from peripheral blood, cord blood, and bone marrow can give rise to erythrocytes, the use of human pluripotent stem cells (hPSCs) has also provided an important opportunity to obtain erythrocytes. These hPSCs include both human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). As hESCs carry ethical and political controversies, hiPSCs can be a more universal source for RBC generation. In this review, we first discuss the key concepts and mechanisms of erythropoiesis. Thereafter, we summarize different methodologies to differentiate hPSCs into erythrocytes with an emphasis on the key features of human definitive erythroid lineage cells. Finally, we address the current limitations and future directions of clinical applications using hiPSC-derived erythrocytes.
Collapse
Affiliation(s)
- Shin-Jeong Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Cholomi Jung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jee Eun Oh
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sangsung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sangho Lee
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Ji Yoon Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
| | - Young-sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| |
Collapse
|
11
|
Heterochromatin rewiring and domain disruption-mediated chromatin compaction during erythropoiesis. Nat Struct Mol Biol 2023; 30:463-474. [PMID: 36914797 DOI: 10.1038/s41594-023-00939-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 02/08/2023] [Indexed: 03/16/2023]
Abstract
Mammalian erythropoiesis involves progressive chromatin compaction and subsequent enucleation in terminal differentiation, but the mechanisms underlying the three-dimensional chromatin reorganization remain obscure. Here, we systematically analyze the higher-order chromatin in purified populations of primary human erythroblasts. Our results reveal that heterochromatin regions undergo substantial compression, with H3K9me3 markers relocalizing to the nuclear periphery and forming a significant number of long-range interactions, and that ~58% of the topologically associating domain (TAD) boundaries are disrupted, while certain TADs enriched for markers of the active transcription state and erythroid master regulators, GATA1 and KLF1, are selectively maintained during terminal erythropoiesis. Finally, we demonstrate that GATA1 is involved in safeguarding selected essential chromatin domains during terminal erythropoiesis. Our study therefore delineates the molecular characteristics of a development-driven chromatin compaction process, which reveals transcription competence as a key indicator of the selected domain maintenance to ensure appropriate gene expression during the extreme compaction of chromatin.
Collapse
|
12
|
Vong P, Messaoudi K, Jankovsky N, Gomilla C, Demont Y, Caulier A, Jedraszak G, Demagny J, Djordjevic S, Boyer T, Marolleau JP, Rochette J, Ouled‐Haddou H, Garçon L. HDAC6 regulates human erythroid differentiation through modulation of JAK2 signalling. J Cell Mol Med 2022; 27:174-188. [PMID: 36578217 PMCID: PMC9843532 DOI: 10.1111/jcmm.17559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 08/25/2022] [Accepted: 09/05/2022] [Indexed: 12/30/2022] Open
Abstract
Among histone deacetylases, HDAC6 is unusual in its cytoplasmic localization. Its inhibition leads to hyperacetylation of non-histone proteins, inhibiting cell cycle, proliferation and apoptosis. Ricolinostat (ACY-1215) is a selective inhibitor of the histone deacetylase HDAC6 with proven efficacy in the treatment of malignant diseases, but anaemia is one of the most frequent side effects. We investigated here the underlying mechanisms of this erythroid toxicity. We first confirmed that HDAC6 was strongly expressed at both RNA and protein levels in CD34+ -cells-derived erythroid progenitors. ACY-1215 exposure on CD34+ -cells driven in vitro towards the erythroid lineage led to a decreased cell count, an increased apoptotic rate and a delayed erythroid differentiation with accumulation of weakly hemoglobinized immature erythroblasts. This was accompanied by drastic changes in the transcriptomic profile of primary cells as shown by RNAseq. In erythroid cells, ACY-1215 and shRNA-mediated HDAC6 knockdown inhibited the EPO-dependent JAK2 phosphorylation. Using acetylome, we identified 14-3-3ζ, known to interact directly with the JAK2 negative regulator LNK, as a potential HDAC6 target in erythroid cells. We confirmed that 14-3-3ζ was hyperacetylated after ACY-1215 exposure, which decreased the 14-3-3ζ/LNK interaction while increased LNK ability to interact with JAK2. Thus, in addition to its previously described role in the enucleation of mouse fetal liver erythroblasts, we identified here a new mechanism of HDAC6-dependent control of erythropoiesis through 14-3-3ζ acetylation level, LNK availability and finally JAK2 activation in response to EPO, which is crucial downstream of EPO-R activation for human erythroid cell survival, proliferation and differentiation.
Collapse
Affiliation(s)
- Pascal Vong
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | | | | | - Cathy Gomilla
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance
| | - Yohann Demont
- Service d'Hématologie BiologiqueCentre Hospitalier UniversitaireAmiensFrance
| | - Alexis Caulier
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance,Service des Maladies du SangCentre Hospitalier UniversitaireAmiensFrance
| | - Guillaume Jedraszak
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance,Laboratoire de Génétique ConstitutionnelleCentre Hospitalier UniversitaireAmiensFrance
| | - Julien Demagny
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance,Service d'Hématologie BiologiqueCentre Hospitalier UniversitaireAmiensFrance
| | | | - Thomas Boyer
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance,Service d'Hématologie BiologiqueCentre Hospitalier UniversitaireAmiensFrance
| | - Jean Pierre Marolleau
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance,Service des Maladies du SangCentre Hospitalier UniversitaireAmiensFrance
| | | | | | - Loïc Garçon
- HEMATIM UR4666Université Picardie Jules VerneAmiensFrance,Service d'Hématologie BiologiqueCentre Hospitalier UniversitaireAmiensFrance
| |
Collapse
|
13
|
Vong P, Ouled-Haddou H, Garçon L. Histone Deacetylases Function in the Control of Early Hematopoiesis and Erythropoiesis. Int J Mol Sci 2022; 23:9790. [PMID: 36077192 PMCID: PMC9456231 DOI: 10.3390/ijms23179790] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Numerous studies have highlighted the role of post-translational modifications in the regulation of cell proliferation, differentiation and death. Among these modifications, acetylation modifies the physicochemical properties of proteins and modulates their activity, stability, localization and affinity for partner proteins. Through the deacetylation of a wide variety of functional and structural, nuclear and cytoplasmic proteins, histone deacetylases (HDACs) modulate important cellular processes, including hematopoiesis, during which different HDACs, by controlling gene expression or by regulating non-histone protein functions, act sequentially to provide a fine regulation of the differentiation process both in early hematopoietic stem cells and in more mature progenitors. Considering that HDAC inhibitors represent promising targets in cancer treatment, it is necessary to decipher the role of HDACs during hematopoiesis which could be impacted by these therapies. This review will highlight the main mechanisms by which HDACs control the hematopoietic stem cell fate, particularly in the erythroid lineage.
Collapse
Affiliation(s)
- Pascal Vong
- Université Picardie Jules Verne, HEMATIM UR4666, 80000 Amiens, France
| | | | - Loïc Garçon
- Université Picardie Jules Verne, HEMATIM UR4666, 80000 Amiens, France
- Service d’Hématologie Biologique, Centre Hospitalier Universitaire, CEDEX 1, 80054 Amiens, France
- Laboratoire de Génétique Constitutionnelle, Centre Hospitalier Universitaire, CEDEX 1, 80054 Amiens, France
| |
Collapse
|
14
|
Tátrai P, Gergely F. Centrosome function is critical during terminal erythroid differentiation. EMBO J 2022; 41:e108739. [PMID: 35678476 PMCID: PMC9289712 DOI: 10.15252/embj.2021108739] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 05/03/2022] [Accepted: 05/25/2022] [Indexed: 11/26/2022] Open
Abstract
Red blood cells are produced by terminal erythroid differentiation, which involves the dramatic morphological transformation of erythroblasts into enucleated reticulocytes. Microtubules are important for enucleation, but it is not known if the centrosome, a key microtubule-organizing center, is required as well. Mice lacking the conserved centrosome component, CDK5RAP2, are likely to have defective erythroid differentiation because they develop macrocytic anemia. Here, we show that fetal liver-derived, CDK5RAP2-deficient erythroid progenitors generate fewer and larger reticulocytes, hence recapitulating features of macrocytic anemia. In erythroblasts, but not in embryonic fibroblasts, loss of CDK5RAP2 or pharmacological depletion of centrosomes leads to highly aberrant spindle morphologies. Consistent with such cells exiting mitosis without chromosome segregation, tetraploidy is frequent in late-stage erythroblasts, thereby giving rise to fewer but larger reticulocytes than normal. Our results define a critical role for CDK5RAP2 and centrosomes in spindle formation specifically during blood production. We propose that disruption of centrosome and spindle function could contribute to the emergence of macrocytic anemias, for instance, due to nutritional deficiency or exposure to chemotherapy.
Collapse
Affiliation(s)
- Péter Tátrai
- Cancer Research UK Cambridge InstituteLi Ka Shing CentreUniversity of CambridgeCambridgeUK
- Present address:
Solvo BiotechnologyBudapestHungary
| | - Fanni Gergely
- Cancer Research UK Cambridge InstituteLi Ka Shing CentreUniversity of CambridgeCambridgeUK
- Department of BiochemistryUniversity of OxfordOxfordUK
| |
Collapse
|
15
|
The path from stem cells to red blood cells. Int J Hematol 2022; 116:160-162. [PMID: 35841459 DOI: 10.1007/s12185-022-03413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
As oxygen is essential for energy production in mitochondria, a sufficient amount of oxygen should be continuously delivered to the tissues to maintain life. Therefore, the number of red blood cells which carry the oxygen is considerable, at up to 25 trillion in the body, and 2 million new red blood cells are generated per second.
Collapse
|
16
|
Soboleva S, Miharada K. Induction of enucleation in primary and immortalized erythroid cells. Int J Hematol 2022; 116:192-198. [PMID: 35610497 DOI: 10.1007/s12185-022-03386-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Enucleation is a crucial event during the erythropoiesis, implicating drastic morphologic and transcriptomic/proteomic changes. While many genes deletion lead to failed or impaired enucleation have been identified, directly triggering the erythroid maturation, particularly enucleation, is still challenging. Inducing enucleation at the desired timing is necessary to develop efficient methods to generate mature, fully functional red blood cells in vitro for future transfusion therapies. However, there are considerable differences between primary erythroid cells and cultured cell sources, particularly pluripotent stem cell-derived erythroid cells and immortalized erythroid cell lines. For instance, the difference in the proliferative status between those cell types could be a critical factor, as cell cycle exit is closely connected to the terminal maturation of primary. In this review, we will discuss previous findings on the enucleation machinery and current challengings to trigger the enucleation of infinite erythroid cell sources.
Collapse
Affiliation(s)
- Svetlana Soboleva
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kenichi Miharada
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden. .,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
17
|
Ren K, Li E, Ji P. Proteome remodeling and organelle clearance in mammalian terminal erythropoiesis. Curr Opin Hematol 2022; 29:137-143. [PMID: 35441599 DOI: 10.1097/moh.0000000000000707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The differentiation from colony forming unit-erythroid (CFU-E) cells to mature enucleated red blood cells is named terminal erythropoiesis in mammals. Apart from enucleation, several unique features during these developmental stages include proteome remodeling and organelle clearance that are important to achieve hemoglobin enrichment. Here, we review the recent advances in the understanding of novel regulatory mechanisms in these processes, focusing on the master regulators that link these major events during terminal erythropoiesis. RECENT FINDINGS Comprehensive proteomic studies revealed a mismatch of protein abundance to their corresponding transcript abundance, which indicates that the proteome remodeling is regulated in a complex way from transcriptional control to posttranslational modifications. Key regulators in organelle clearance were also found to play critical roles in proteome remodeling. SUMMARY These studies demonstrate that the complexity of terminal erythropoiesis is beyond the conventional transcriptomic centric perspective. Posttranslational modifications such as ubiquitination are critical in terminal erythroid proteome remodeling that is also closely coupled with organelle clearance.
Collapse
Affiliation(s)
- Kehan Ren
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | |
Collapse
|
18
|
Wells M, Steiner L. Epigenetic and Transcriptional Control of Erythropoiesis. Front Genet 2022; 13:805265. [PMID: 35330735 PMCID: PMC8940284 DOI: 10.3389/fgene.2022.805265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/16/2022] [Indexed: 12/21/2022] Open
Abstract
Erythropoiesis is a process of enormous magnitude, with the average person generating two to three million red cells every second. Erythroid progenitors start as large cells with large nuclei, and over the course of three to four cell divisions they undergo a dramatic decrease in cell size accompanied by profound nuclear condensation, which culminates in enucleation. As maturing erythroblasts are undergoing these dramatic phenotypic changes, they accumulate hemoglobin and express high levels of other erythroid-specific genes, while silencing much of the non-erythroid transcriptome. These phenotypic and gene expression changes are associated with distinct changes in the chromatin landscape, and require close coordination between transcription factors and epigenetic regulators, as well as precise regulation of RNA polymerase II activity. Disruption of these processes are associated with inherited anemias and myelodysplastic syndromes. Here, we review the epigenetic mechanisms that govern terminal erythroid maturation, and their role in human disease.
Collapse
Affiliation(s)
- Maeve Wells
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Laurie Steiner
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| |
Collapse
|
19
|
Wang S, Zhao H, Zhang H, Gao C, Guo X, Chen L, Lobo C, Yazdanbakhsh K, Zhang S, An X. Analyses of erythropoiesis from embryonic stem cell‐CD34
+
and cord blood‐CD34
+
cells reveal mechanisms for defective expansion and enucleation of embryomic stem cell‐erythroid cells. J Cell Mol Med 2022; 26:2404-2416. [PMID: 35249258 PMCID: PMC8995447 DOI: 10.1111/jcmm.17263] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 11/28/2022] Open
Abstract
Red blood cells (RBCs) generated ex vivo have the potential to be used for transfusion. Human embryonic stem cells (ES) and induced pluripotent stem cells (iPS) possess unlimited self‐renewal capacity and are the preferred cell sources to be used for ex vivo RBC generation. However, their applications are hindered by the facts that the expansion of ES/iPS‐derived erythroid cells is limited and the enucleation of ES/iPS‐derived erythroblasts is low compared to that derived from cord blood (CB) or peripheral blood (PB). To address this, we sought to investigate the underlying mechanisms by comparing the in vitro erythropoiesis profiles of CB CD34+ and ES CD34+ cells. We found that the limited expansion of ES CD34+ cell‐derived erythroid cells was associated with defective cell cycle of erythroid progenitors. In exploring the cellular and molecular mechanisms for the impaired enucleation of ES CD34+ cell‐derived orthochromatic erythroblasts (ES‐ortho), we found the chromatin of ES‐ortho was less condensed than that of CB CD34+ cell‐derived orthochromatic erythroblasts (CB‐ortho). At the molecular level, both RNA‐seq and ATAC‐seq analyses revealed that pathways involved in chromatin modification were down‐regulated in ES‐ortho. Additionally, the expression levels of molecules known to play important role in chromatin condensation or/and enucleation were significantly lower in ES‐ortho compared to that in CB‐ortho. Together, our findings have uncovered mechanisms for the limited expansion and impaired enucleation of ES CD34+ cell‐derived erythroid cells and may help to improve ex vivo RBC production from stem cells.
Collapse
Affiliation(s)
- Shihui Wang
- School of Life Sciences Zhengzhou University Zhengzhou China
- Laboratory of Membrane Biology New York Blood Center New York New York USA
| | - Huizhi Zhao
- School of Life Sciences Zhengzhou University Zhengzhou China
| | - Huan Zhang
- Laboratory of Membrane Biology New York Blood Center New York New York USA
| | - Chengjie Gao
- Laboratory of Membrane Biology New York Blood Center New York New York USA
| | - Xinhua Guo
- Laboratory of Membrane Biology New York Blood Center New York New York USA
| | - Lixiang Chen
- School of Life Sciences Zhengzhou University Zhengzhou China
| | - Cheryl Lobo
- Laboratory of Blood Borne Parasites New York Blood Center New York New York USA
| | - Karina Yazdanbakhsh
- Laboratory of Complement Biology New York Blood Center New York New York USA
| | - Shijie Zhang
- School of Life Sciences Zhengzhou University Zhengzhou China
| | - Xiuli An
- Laboratory of Membrane Biology New York Blood Center New York New York USA
| |
Collapse
|
20
|
Murphy ZC, Murphy K, Myers J, Getman M, Couch T, Schulz VP, Lezon-Geyda K, Palumbo C, Yan H, Mohandas N, Gallagher PG, Steiner LA. Regulation of RNA polymerase II activity is essential for terminal erythroid maturation. Blood 2021; 138:1740-1756. [PMID: 34075391 PMCID: PMC8569412 DOI: 10.1182/blood.2020009903] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/30/2021] [Indexed: 11/20/2022] Open
Abstract
The terminal maturation of human erythroblasts requires significant changes in gene expression in the context of dramatic nuclear condensation. Defects in this process are associated with inherited anemias and myelodysplastic syndromes. The progressively dense appearance of the condensing nucleus in maturing erythroblasts led to the assumption that heterochromatin accumulation underlies this process, but despite extensive study, the precise mechanisms underlying this essential biologic process remain elusive. To delineate the epigenetic changes associated with the terminal maturation of human erythroblasts, we performed mass spectrometry of histone posttranslational modifications combined with chromatin immunoprecipitation coupled with high-throughput sequencing, Assay for Transposase Accessible Chromatin, and RNA sequencing. Our studies revealed that the terminal maturation of human erythroblasts is associated with a dramatic decline in histone marks associated with active transcription elongation, without accumulation of heterochromatin. Chromatin structure and gene expression were instead correlated with dynamic changes in occupancy of elongation competent RNA polymerase II, suggesting that terminal erythroid maturation is controlled largely at the level of transcription. We further demonstrate that RNA polymerase II "pausing" is highly correlated with transcriptional repression, with elongation competent RNA polymerase II becoming a scare resource in late-stage erythroblasts, allocated to erythroid-specific genes. Functional studies confirmed an essential role for maturation stage-specific regulation of RNA polymerase II activity during erythroid maturation and demonstrate a critical role for HEXIM1 in the regulation of gene expression and RNA polymerase II activity in maturing erythroblasts. Taken together, our findings reveal important insights into the mechanisms that regulate terminal erythroid maturation and provide a novel paradigm for understanding normal and perturbed erythropoiesis.
Collapse
Affiliation(s)
| | | | - Jacquelyn Myers
- Department of Pediatrics and
- Genomics Resource Center, University of Rochester, Rochester, NY
| | | | | | | | | | - Cal Palumbo
- Genomics Resource Center, University of Rochester, Rochester, NY
| | | | | | | | | |
Collapse
|
21
|
Mehrpouri M, Pourbagheri-Sigaroodi A, Bashash D. The contributory roles of histone deacetylases (HDACs) in hematopoiesis regulation and possibilities for pharmacologic interventions in hematologic malignancies. Int Immunopharmacol 2021; 100:108114. [PMID: 34492531 DOI: 10.1016/j.intimp.2021.108114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 12/17/2022]
Abstract
Although the definitive role of epigenetic modulations in a wide range of hematologic malignancies, spanning from leukemia to lymphoma and multiple myeloma, has been evidenced, few articles reviewed the task. Given the high accessibility of histone deacetylase (HDACs) to necessary transcription factors involved in hematopoiesis, this review aims to outline physiologic impacts of these enzymes in normal hematopoiesis, and also to outline the original data obtained from international research laboratories on their regulatory role in the differentiation and maturation of different hematopoietic lineages. Questions on how aberrant expression of HDACs contributes to the formation of hematologic malignancies are also responded, because these classes of enzymes have a respectable share in the development, progression, and recurrence of leukemia, lymphoma, and multiple myeloma. The last section provides a special focus on the therapeutic perspectiveof HDACs inhibitors, either as single agents or in a combined-modal strategy, in these neoplasms. In conclusion, optimizing the dose and the design of more patient-tailored inhibitors, while maintaining low toxicity against normal cells, will help improve clinical outcomes of HDAC inhibitors in hematologic malignancies.
Collapse
Affiliation(s)
- Mahdieh Mehrpouri
- Department of Laboratory Sciences, School of Allied Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Garcia AA, Koperniku A, Ferreira JCB, Mochly-Rosen D. Treatment strategies for glucose-6-phosphate dehydrogenase deficiency: past and future perspectives. Trends Pharmacol Sci 2021; 42:829-844. [PMID: 34389161 DOI: 10.1016/j.tips.2021.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/19/2021] [Accepted: 07/13/2021] [Indexed: 01/20/2023]
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) maintains redox balance in a variety of cell types and is essential for erythrocyte resistance to oxidative stress. G6PD deficiency, caused by mutations in the G6PD gene, is present in ~400 million people worldwide, and can cause acute hemolytic anemia. Currently, there are no therapeutics for G6PD deficiency. We discuss the role of G6PD in hemolytic and nonhemolytic disorders, treatment strategies attempted over the years, and potential reasons for their failure. We also discuss potential pharmacological pathways, including glutathione (GSH) metabolism, compensatory NADPH production routes, transcriptional upregulation of the G6PD gene, highlighting potential drug targets. The needs and opportunities described here may motivate the development of a therapeutic for hematological and other chronic diseases associated with G6PD deficiency.
Collapse
Affiliation(s)
- Adriana A Garcia
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ana Koperniku
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Julio C B Ferreira
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, USA; Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daria Mochly-Rosen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
Jeffery NN, Davidson C, Peslak SA, Kingsley PD, Nakamura Y, Palis J, Bulger M. Histone H2A.X phosphorylation and Caspase-Initiated Chromatin Condensation in late-stage erythropoiesis. Epigenetics Chromatin 2021; 14:37. [PMID: 34330317 PMCID: PMC8325214 DOI: 10.1186/s13072-021-00408-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Background Condensation of chromatin prior to enucleation is an essential component of terminal erythroid maturation, and defects in this process are associated with inefficient erythropoiesis and anemia. However, the mechanisms involved in this phenomenon are not well understood. Here, we describe a potential role for the histone variant H2A.X in erythropoiesis. Results We find in multiple model systems that this histone is essential for normal maturation, and that the loss of H2A.X in erythroid cells results in dysregulation in expression of erythroid-specific genes as well as a nuclear condensation defect. In addition, we demonstrate that erythroid maturation is characterized by phosphorylation at both S139 and Y142 on the C-terminal tail of H2A.X during late-stage erythropoiesis. Knockout of the kinase BAZ1B/WSTF results in loss of Y142 phosphorylation and a defect in nuclear condensation, but does not replicate extensive transcriptional changes to erythroid-specific genes observed in the absence of H2A.X. Conclusions We relate these findings to Caspase-Initiated Chromatin Condensation (CICC) in terminal erythroid maturation, where aspects of the apoptotic pathway are invoked while apoptosis is specifically suppressed. Supplementary Information The online version contains supplementary material available at 10.1186/s13072-021-00408-5.
Collapse
Affiliation(s)
- Nazish N Jeffery
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Christina Davidson
- Wilmot Cancer Institute, Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Scott A Peslak
- Department of Medicine, Division of Hematology/Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.,Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Paul D Kingsley
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - James Palis
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester, Rochester, NY, USA
| | - Michael Bulger
- Center for Pediatric Biomedical Research, Department of Pediatrics, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
24
|
Ryzhkova A, Battulin N. Genome Reorganization during Erythroid Differentiation. Genes (Basel) 2021; 12:genes12071012. [PMID: 34208866 PMCID: PMC8306769 DOI: 10.3390/genes12071012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 01/02/2023] Open
Abstract
Hematopoiesis is a convenient model to study how chromatin dynamics plays a decisive role in regulation of cell fate. During erythropoiesis a population of stem and progenitor cells becomes increasingly lineage restricted, giving rise to terminally differentiated progeny. The concerted action of transcription factors and epigenetic modifiers leads to a silencing of the multipotent transcriptome and activation of the transcriptional program that controls terminal differentiation. This article reviews some aspects of the biology of red blood cells production with the focus on the extensive chromatin reorganization during differentiation.
Collapse
Affiliation(s)
- Anastasia Ryzhkova
- Institute of Cytology and Genetics SB RAS, Laboratory of Developmental Genetics, 630090 Novosibirsk, Russia;
| | - Nariman Battulin
- Institute of Cytology and Genetics SB RAS, Laboratory of Developmental Genetics, 630090 Novosibirsk, Russia;
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
- Correspondence:
| |
Collapse
|
25
|
Soboleva S, Kurita R, Ek F, Åkerstrand H, Silvério-Alves R, Olsson R, Nakamura Y, Miharada K. Identification of potential chemical compounds enhancing generation of enucleated cells from immortalized human erythroid cell lines. Commun Biol 2021; 4:677. [PMID: 34083702 PMCID: PMC8175573 DOI: 10.1038/s42003-021-02202-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 05/11/2021] [Indexed: 11/09/2022] Open
Abstract
Immortalized erythroid cell lines are expected to be a promising source of ex vivo manufactured red blood cells (RBCs), however the induction of enucleation in these cell lines is inefficient at present. We utilized an imaging-based high-throughput system to identify chemical compounds that trigger enucleation of human erythroid cell lines. Among >3,300 compounds, we identified multiple histone deacetylase inhibitors (HDACi) inducing enucleated cells from the cell line, although an increase in membrane fragility of enucleated cells was observed. Gene expression profiling revealed that HDACi treatment increased the expression of cytoskeletal genes, while an erythroid-specific cell membrane protein, SPTA1, was significantly down-regulated. Restoration of SPTA1 expression using CRISPR-activation partially rescued the fragility of cells and thereby improved the enucleation efficiency. Our observations provide a potential solution for the generation of mature cells from erythroid cell lines, contributing to the future realization of the use of immortalized cell lines for transfusion therapies. In an imaging-based screen of >3,300 compounds compounds, Soboleva et al identify HDAC inhibitors as mediators of erythroid cell enucleation. They further show that the erythroid-specific cell membrane protein, SPTA1, is downregulated in HDAC inhibited cells and that restoration of SPTA1 expression using CRISPR-activation partially rescues the fragility of cells, improving enucleation efficiency.
Collapse
Affiliation(s)
- Svetlana Soboleva
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Ryo Kurita
- Department of Research and Development, Central Blood Institute, Japanese Red Cross Society, Tokyo, Japan
| | - Fredrik Ek
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Hugo Åkerstrand
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Rita Silvério-Alves
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Roger Olsson
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Kenichi Miharada
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden. .,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
26
|
Impairment of human terminal erythroid differentiation by histone deacetylase 5 deficiency. Blood 2021; 138:1615-1627. [PMID: 34036344 DOI: 10.1182/blood.2020007401] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 04/22/2021] [Indexed: 11/20/2022] Open
Abstract
Histone deacetylases (HDACs) are a group of enzymes catalyzing the removal of acetyl groups from histone and non-histone proteins. HDACs have been shown to play diverse functions in a wide range of biological processes. However, their roles in mammalian erythropoiesis remain to be fully defined. We show here that of the eleven classic HDAC family members, six of them (HDAC 1,2,3 and HDAC 5,6,7) are expressed in human erythroid cells with HDAC5 most significantly up regulated during terminal erythroid differentiation. Knockdown of HDAC5 by either shRNA or siRNA in human CD34+ cells followed by erythroid cell culture led to increased apoptosis, decreased chromatin condensation, and impaired enucleation of erythroblasts. Biochemical analyses revealed that HDAC5 deficiency resulted in activation of p53 in association with increased acetylation of p53. Furthermore, while acetylation of histone 4 (H4) is decreased during normal terminal erythroid differentiation, HDAC5 deficiency led to increased acetylation of H4 (K12) in late stage erythroblasts. This increased acetylation was accompanied by decreased chromatin condensation, implying a role for H4 (K12) deacetylation in chromatin condensation. ATAC-seq and RNA-seq analyses revealed that HDAC5 knockdown leads to increased chromatin accessibility genome wide and global changes in gene expression. Moreover, pharmacological inhibition of HDAC5 by the inhibitor LMK235 also led to increased H4 acetylation, impaired chromatin condensation and enucleation. Taken together, our findings have uncovered previously unrecognized roles and molecular mechanisms of action for HDAC5 in human erythropoiesis. These results may provide insights into understanding the anemia associated with HDAC inhibitor treatment.
Collapse
|
27
|
Wenmaekers S, Viergever BJ, Kumar G, Kranenburg O, Black PC, Daugaard M, Meijer RP. A Potential Role for HUWE1 in Modulating Cisplatin Sensitivity. Cells 2021; 10:cells10051262. [PMID: 34065298 PMCID: PMC8160634 DOI: 10.3390/cells10051262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022] Open
Abstract
Cisplatin is a widely used antineoplastic agent, whose efficacy is limited by primary and acquired therapeutic resistance. Recently, a bladder cancer genome-wide CRISPR/Cas9 knock-out screen correlated cisplatin sensitivity to multiple genetic biomarkers. Among the screen’s top hits was the HECT domain-containing ubiquitin E3 ligase (HUWE1). In this review, HUWE1 is postulated as a therapeutic response modulator, affecting the collision between platinum-DNA adducts and the replication fork, the primary cytotoxic action of platins. HUWE1 can alter the cytotoxic response to platins by targeting essential components of the DNA damage response including BRCA1, p53, and Mcl-1. Deficiency of HUWE1 could lead to enhanced DNA damage repair and a dysfunctional apoptotic apparatus, thereby inducing resistance to platins. Future research on the relationship between HUWE1 and platins could generate new mechanistic insights into therapy resistance. Ultimately, HUWE1 might serve as a clinical biomarker to tailor cancer treatment strategies, thereby improving cancer care and patient outcomes.
Collapse
Affiliation(s)
- Stijn Wenmaekers
- Laboratory Translational Oncology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands; (S.W.); (B.J.V.); (O.K.)
- Department of Oncological Urology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Bastiaan J. Viergever
- Laboratory Translational Oncology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands; (S.W.); (B.J.V.); (O.K.)
- Department of Oncological Urology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Gunjan Kumar
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; (G.K.); (P.C.B.)
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Onno Kranenburg
- Laboratory Translational Oncology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands; (S.W.); (B.J.V.); (O.K.)
| | - Peter C. Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; (G.K.); (P.C.B.)
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | - Mads Daugaard
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada; (G.K.); (P.C.B.)
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Correspondence: (M.D.); (R.P.M.)
| | - Richard P. Meijer
- Laboratory Translational Oncology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands; (S.W.); (B.J.V.); (O.K.)
- Department of Oncological Urology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
- Correspondence: (M.D.); (R.P.M.)
| |
Collapse
|
28
|
A new role of glutathione peroxidase 4 during human erythroblast enucleation. Blood Adv 2021; 4:5666-5680. [PMID: 33211827 DOI: 10.1182/bloodadvances.2020003100] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
The selenoprotein glutathione peroxidase 4 (GPX4), the only member of the glutathione peroxidase family able to directly reduce cell membrane-oxidized fatty acids and cholesterol, was recently identified as the central regulator of ferroptosis. GPX4 knockdown in mouse hematopoietic cells leads to hemolytic anemia and to increased spleen erythroid progenitor death. The role of GPX4 during human erythropoiesis is unknown. Using in vitro erythroid differentiation, we show here that GPX4-irreversible inhibition by 1S,3R-RSL3 (RSL3) and its short hairpin RNA-mediated knockdown strongly impaired enucleation in a ferroptosis-independent manner not restored by tocopherol or iron chelators. During enucleation, GPX4 localized with lipid rafts at the cleavage furrows between reticulocytes and pyrenocytes. Its inhibition impacted enucleation after nuclear condensation and polarization and was associated with a defect in lipid raft clustering (cholera toxin staining) and myosin-regulatory light-chain phosphorylation. Because selenoprotein translation and cholesterol synthesis share a common precursor, we investigated whether the enucleation defect could represent a compensatory mechanism favoring GPX4 synthesis at the expense of cholesterol, known to be abundant in lipid rafts. Lipidomics and filipin staining failed to show any quantitative difference in cholesterol content after RSL3 exposure. However, addition of cholesterol increased cholera toxin staining and myosin-regulatory light-chain phosphorylation, and improved enucleation despite GPX4 knockdown. In summary, we identified GPX4 as a new actor of human erythroid enucleation, independent of its function in ferroptosis control. We described its involvement in lipid raft organization required for contractile ring assembly and cytokinesis, leading in fine to nucleus extrusion.
Collapse
|
29
|
Feola M, Zamperone A, Moskop D, Chen H, Casu C, Lama D, Di Martino J, Djedaini M, Papa L, Martinez MR, Choesang T, Bravo-Cordero JJ, MacKay M, Zumbo P, Brinkman N, Abrams CS, Rivella S, Hattangadi S, Mason CE, Hoffman R, Ji P, Follenzi A, Ginzburg YZ. Pleckstrin-2 is essential for erythropoiesis in β-thalassemic mice, reducing apoptosis and enhancing enucleation. Commun Biol 2021; 4:517. [PMID: 33941818 PMCID: PMC8093212 DOI: 10.1038/s42003-021-02046-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/23/2021] [Indexed: 02/03/2023] Open
Abstract
Erythropoiesis involves complex interrelated molecular signals influencing cell survival, differentiation, and enucleation. Diseases associated with ineffective erythropoiesis, such as β-thalassemias, exhibit erythroid expansion and defective enucleation. Clear mechanistic determinants of what make erythropoiesis effective are lacking. We previously demonstrated that exogenous transferrin ameliorates ineffective erythropoiesis in β-thalassemic mice. In the current work, we utilize transferrin treatment to elucidate a molecular signature of ineffective erythropoiesis in β-thalassemia. We hypothesize that compensatory mechanisms are required in β-thalassemic erythropoiesis to prevent apoptosis and enhance enucleation. We identify pleckstrin-2-a STAT5-dependent lipid binding protein downstream of erythropoietin-as an important regulatory node. We demonstrate that partial loss of pleckstrin-2 leads to worsening ineffective erythropoiesis and pleckstrin-2 knockout leads to embryonic lethality in β-thalassemic mice. In addition, the membrane-associated active form of pleckstrin-2 occurs at an earlier stage during β-thalassemic erythropoiesis. Furthermore, membrane-associated activated pleckstrin-2 decreases cofilin mitochondrial localization in β-thalassemic erythroblasts and pleckstrin-2 knockdown in vitro induces cofilin-mediated apoptosis in β-thalassemic erythroblasts. Lastly, pleckstrin-2 enhances enucleation by interacting with and activating RacGTPases in β-thalassemic erythroblasts. This data elucidates the important compensatory role of pleckstrin-2 in β-thalassemia and provides support for the development of targeted therapeutics in diseases of ineffective erythropoiesis.
Collapse
Affiliation(s)
- Maria Feola
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- University of Piemonte Orientale, Amedeo Avogadro, Novara, Italy
| | - Andrea Zamperone
- Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Daniel Moskop
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Huiyong Chen
- Erythropoiesis Laboratory, New York Blood Center, New York, NY, USA
- Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, China
| | - Carla Casu
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dechen Lama
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie Di Martino
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mansour Djedaini
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luena Papa
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marc Ruiz Martinez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tenzin Choesang
- Erythropoiesis Laboratory, New York Blood Center, New York, NY, USA
| | | | | | - Paul Zumbo
- Weill Cornell Medical College, New York, NY, USA
| | | | - Charles S Abrams
- Perelman Center for Advanced Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peng Ji
- Northwestern University, Chicago, IL, USA
| | - Antonia Follenzi
- University of Piemonte Orientale, Amedeo Avogadro, Novara, Italy
| | - Yelena Z Ginzburg
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
30
|
Mei Y, Liu Y, Ji P. Understanding terminal erythropoiesis: An update on chromatin condensation, enucleation, and reticulocyte maturation. Blood Rev 2021; 46:100740. [PMID: 32798012 DOI: 10.1016/j.blre.2020.100740] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/02/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
A characteristic feature of terminal erythropoiesis in mammals is extrusion of the highly condensed nucleus out of the cytoplasm. Other vertebrates, including fish, reptiles, amphibians, and birds, undergo nuclear condensation but do not enucleate. Enucleation provides mammals evolutionary advantages by gaining extra space for hemoglobin and being more flexible to migrate through capillaries. Nascent reticulocytes further mature into red blood cells through membrane and proteome remodeling and organelle clearance. Over the past decade, novel molecular mechanisms and signaling pathways have been uncovered that play important roles in chromatin condensation, enucleation, and reticulocyte maturation. These advances not only increase understanding of the physiology of erythropoiesis, but also facilitate efforts in generating in vitro red blood cells for various translational application. In the present review, recent studies in epigenetic modification and release of histones during chromatin condensation are highlighted. New insights in enucleation, including protein sorting, vesicle trafficking, transcriptional regulation, noncoding RNA, cytoskeleton remodeling, erythroblastic islands, and cytokinesis, are summarized. Moreover, organelle clearance and proteolysis mediated by ubiquitin-proteasome degradation during reticulocytes maturation is also examined. Perspectives for future directions in this rapidly evolving research area are also provided.
Collapse
Affiliation(s)
- Yang Mei
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| | - Yijie Liu
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| | - Peng Ji
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
31
|
Menon V, Ghaffari S. Erythroid enucleation: a gateway into a "bloody" world. Exp Hematol 2021; 95:13-22. [PMID: 33440185 PMCID: PMC8147720 DOI: 10.1016/j.exphem.2021.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/18/2022]
Abstract
Erythropoiesis is an intricate process starting in hematopoietic stem cells and leading to the daily production of 200 billion red blood cells (RBCs). Enucleation is a greatly complex and rate-limiting step during terminal maturation of mammalian RBC production involving expulsion of the nucleus from the orthochromatic erythroblasts, resulting in the formation of reticulocytes. The dynamic enucleation process involves many factors ranging from cytoskeletal proteins to transcription factors to microRNAs. Lack of optimum terminal erythroid maturation and enucleation has been an impediment to optimum RBC production ex vivo. Major efforts in the past two decades have exposed some of the mechanisms that govern the enucleation process. This review focuses in detail on mechanisms implicated in enucleation and discusses the future perspectives of this fascinating process.
Collapse
Affiliation(s)
- Vijay Menon
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Saghi Ghaffari
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY; Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
32
|
Sundaravel S, Steidl U, Wickrema A. Epigenetic modifiers in normal and aberrent erythropoeisis. Semin Hematol 2021; 58:15-26. [PMID: 33509439 PMCID: PMC7883935 DOI: 10.1053/j.seminhematol.2020.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022]
Abstract
Erythroid differentiation program is comprised of lineage commitment, erythroid progenitor proliferation, and termination differentiation. Each stage of the differentiation program is heavily influenced by epigenetic modifiers that alter the epigenome in a dynamic fashion influenced by cytokines/humeral factors and are amicable to target by drugs. The epigenetic modifiers can be classified as DNA modifiers (DNMT, TET), mRNA modifiers (RNA methylases and demethylases) and histone protein modifiers (methyltransferases, acetyltransferases, demethylases, and deacetylases). Here we describe mechanisms by which these epigenetic modifiers influence and guide erythroid-lineage differentiation during normal and malignant erythropoiesis and also benign diseases that arise from their altered structure or function.
Collapse
Affiliation(s)
- Sriram Sundaravel
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, Albert Einstein College of Medicine-Montefiore Medical center, Bronx, NY
| | | |
Collapse
|
33
|
An Overview of Different Strategies to Recreate the Physiological Environment in Experimental Erythropoiesis. Int J Mol Sci 2020; 21:ijms21155263. [PMID: 32722249 PMCID: PMC7432157 DOI: 10.3390/ijms21155263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/19/2022] Open
Abstract
Human erythropoiesis is a complex process leading to the production of mature, enucleated erythrocytes (RBCs). It occurs mainly at bone marrow (BM), where hematopoietic stem cells (HSCs) are engaged in the early erythroid differentiation to commit into erythroid progenitor cells (burst-forming unit erythroid (BFU-E) and colony-forming unit erythroid (CFU-E)). Then, during the terminal differentiation, several erythropoietin-induced signaling pathways trigger the differentiation of CFU-E on successive stages from pro-erythroblast to reticulocytes. The latter are released into the circulation, finalizing their maturation into functional RBCs. This process is finely regulated by the physiological environment including the erythroblast-macrophage interaction in the erythroblastic island (EBI). Several human diseases have been associated with ineffective erythropoiesis, either by a defective or an excessive production of RBCs, as well as an increase or a hemoglobinization defect. Fully understanding the production of mature red blood cells is crucial for the comprehension of erythroid pathologies as well as to the field of transfusion. Many experimental approaches have been carried out to achieve a complete differentiation in vitro to produce functional biconcave mature RBCs. However, the various protocols usually fail to achieve enough quantities of completely mature RBCs. In this review, we focus on the evolution of erythropoiesis studies over the years, taking special interest in efforts that were made to include the microenvironment and erythroblastic islands paradigm. These more physiological approaches will contribute to a deeper comprehension of erythropoiesis, improve the treatment of dyserythropoietic disorders, and break through the barriers in massive RBCs production for transfusion.
Collapse
|
34
|
Romano O, Petiti L, Felix T, Meneghini V, Portafax M, Antoniani C, Amendola M, Bicciato S, Peano C, Miccio A. GATA Factor-Mediated Gene Regulation in Human Erythropoiesis. iScience 2020; 23:101018. [PMID: 32283524 PMCID: PMC7155206 DOI: 10.1016/j.isci.2020.101018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/14/2020] [Accepted: 03/24/2020] [Indexed: 01/31/2023] Open
Abstract
Erythroid commitment and differentiation are regulated by the coordinated action of a host of transcription factors, including GATA2 and GATA1. Here, we explored GATA-mediated transcriptional regulation through the integrative analysis of gene expression, chromatin modifications, and GATA factors' binding in human multipotent hematopoietic stem/progenitor cells, early erythroid progenitors, and late precursors. A progressive loss of H3K27 acetylation and a diminished usage of active enhancers and super-enhancers were observed during erythroid commitment and differentiation. GATA factors mediate transcriptional changes through a stage-specific interplay with regulatory elements: GATA1 binds different sets of regulatory elements in erythroid progenitors and precursors and controls the transcription of distinct genes during commitment and differentiation. Importantly, our results highlight a pivotal role of promoters in determining the transcriptional program activated upon erythroid differentiation. Finally, we demonstrated that GATA1 binding to a stage-specific super-enhancer sustains the expression of the KIT receptor in human erythroid progenitors. GATA2/1 binding to regulatory regions and transcriptional changes during erythropoiesis GATA1 sustains KIT expression in human erythroid progenitors
Collapse
Affiliation(s)
- Oriana Romano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Petiti
- Institute of Biomedical Technologies, CNR, Milan, Italy
| | - Tristan Felix
- Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, INSERM UMR, 1163 Paris, France
| | - Vasco Meneghini
- Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, INSERM UMR, 1163 Paris, France
| | - Michel Portafax
- Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, INSERM UMR, 1163 Paris, France
| | - Chiara Antoniani
- Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, INSERM UMR, 1163 Paris, France
| | | | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Clelia Peano
- Institute of Biomedical Technologies, CNR, Milan, Italy; Institute of Genetic and Biomedical Research, UOS Milan, National Research Council, Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, IRCCS, Rozzano, Milan, Italy.
| | - Annarita Miccio
- Laboratory of Chromatin and Gene Regulation during Development, Imagine Institute, INSERM UMR, 1163 Paris, France; Paris Descartes, Sorbonne Paris Cité University, Imagine Institute, Paris, France.
| |
Collapse
|
35
|
Wang P, Wang Z, Liu J. Role of HDACs in normal and malignant hematopoiesis. Mol Cancer 2020; 19:5. [PMID: 31910827 PMCID: PMC6945581 DOI: 10.1186/s12943-019-1127-7] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/26/2019] [Indexed: 01/09/2023] Open
Abstract
Normal hematopoiesis requires the accurate orchestration of lineage-specific patterns of gene expression at each stage of development, and epigenetic regulators play a vital role. Disordered epigenetic regulation has emerged as a key mechanism contributing to hematological malignancies. Histone deacetylases (HDACs) are a series of key transcriptional cofactors that regulate gene expression by deacetylation of lysine residues on histone and nonhistone proteins. In normal hematopoiesis, HDACs are widely involved in the development of various lineages. Their functions involve stemness maintenance, lineage commitment determination, cell differentiation and proliferation, etc. Deregulation of HDACs by abnormal expression or activity and oncogenic HDAC-containing transcriptional complexes are involved in hematological malignancies. Currently, HDAC family members are attractive targets for drug design, and a variety of HDAC-based combination strategies have been developed for the treatment of hematological malignancies. Drug resistance and limited therapeutic efficacy are key issues that hinder the clinical applications of HDAC inhibitors (HDACis). In this review, we summarize the current knowledge of how HDACs and HDAC-containing complexes function in normal hematopoiesis and highlight the etiology of HDACs in hematological malignancies. Moreover, the implication and drug resistance of HDACis are also discussed. This review presents an overview of the physiology and pathology of HDACs in the blood system.
Collapse
Affiliation(s)
- Pan Wang
- The Xiangya Hospital, Central South University, Changsha, 410005, Hunan, China.,Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Zi Wang
- The Xiangya Hospital, Central South University, Changsha, 410005, Hunan, China. .,Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| | - Jing Liu
- Molecular Biology Research Center and Hunan Province Key Laboratory of Basic and Applied Hematology, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
36
|
Fibach E. Erythropoiesis In Vitro-A Research and Therapeutic Tool in Thalassemia. J Clin Med 2019; 8:jcm8122124. [PMID: 31810354 PMCID: PMC6947291 DOI: 10.3390/jcm8122124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Thalassemia (thal) is a hereditary chronic hemolytic anemia due to a partial or complete deficiency in the production of globin chains, in most cases, α or β, which compose, together with the iron-containing porphyrins (hemes), the hemoglobin molecules in red blood cells (RBC). The major clinical symptom of β-thal is severe chronic anemia—a decrease in RBC number and their hemoglobin content. In spite of the improvement in therapy, thal still severely affects the quality of life of the patients and their families and imposes a substantial financial burden on the community. These considerations position β-thal, among other hemoglobinopathies, as a major health and social problem that deserves increased efforts in research and its clinical application. These efforts are based on clinical studies, experiments in animal models and the use of erythroid cells grown in culture. The latter include immortal cell lines and cultures initiated by erythroid progenitor and stem cells derived from the blood and RBC producing (erythropoietic) sites of normal and thal donors, embryonic stem cells, and recently, "induced pluripotent stem cells" generated by manipulation of differentiated somatic cells. The present review summarizes the use of erythroid cultures, their technological aspects and their contribution to the research and its clinical application in thal. The former includes deciphering of the normal and pathological biology of the erythroid cell development, and the latter—their role in developing innovative therapeutics—drugs and methods of gene therapy, as well as providing an alternative source of RBC that may complement or substitute blood transfusions.
Collapse
Affiliation(s)
- Eitan Fibach
- The Hematology Department, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| |
Collapse
|
37
|
Yeo JH, Lam YW, Fraser ST. Cellular dynamics of mammalian red blood cell production in the erythroblastic island niche. Biophys Rev 2019; 11:873-894. [PMID: 31418139 PMCID: PMC6874942 DOI: 10.1007/s12551-019-00579-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
Red blood cells, or erythrocytes, make up approximately a quarter of all cells in the human body with over 2 billion new erythrocytes made each day in a healthy adult human. This massive cellular production system is coupled with a set of cell biological processes unique to mammals, in particular, the elimination of all organelles, and the expulsion and destruction of the condensed erythroid nucleus. Erythrocytes from birds, reptiles, amphibians and fish possess nuclei, mitochondria and other organelles: erythrocytes from mammals lack all of these intracellular components. This review will focus on the dynamic changes that take place in developing erythroid cells that are interacting with specialized macrophages in multicellular clusters termed erythroblastic islands. Proerythroblasts enter the erythroblastic niche as large cells with active nuclei, mitochondria producing heme and energy, and attach to the central macrophage via a range of adhesion molecules. Proerythroblasts then mature into erythroblasts and, following enucleation, in reticulocytes. When reticulocytes exit the erythroblastic island, they are smaller cells, without nuclei and with few mitochondria, possess some polyribosomes and have a profoundly different surface molecule phenotype. Here, we will review, step-by-step, the biophysical mechanisms that regulate the remarkable process of erythropoiesis with a particular focus on the events taking place in the erythroblastic island niche. This is presented from the biological perspective to offer insight into the elements of red blood cell development in the erythroblastic island niche which could be further explored with biophysical modelling systems.
Collapse
Affiliation(s)
- Jia Hao Yeo
- Discipline of Anatomy and Histology, School of Medical Sciences, University of Sydney, Sydney, Australia.
- School of Chemistry, University of Sydney, Sydney, Australia.
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Sydney, Australia.
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Stuart T Fraser
- Discipline of Anatomy and Histology, School of Medical Sciences, University of Sydney, Sydney, Australia.
- Discipline of Physiology, School of Medical Sciences, University of Sydney, Sydney, Australia.
- Bosch Institute, School of Medical Sciences, University of Sydney, Sydney, Australia.
- University of Sydney Nano Institute, Sydney, Australia.
| |
Collapse
|
38
|
Cantú I, van de Werken HJG, Gillemans N, Stadhouders R, Heshusius S, Maas A, Esteghamat F, Ozgur Z, van IJcken WFJ, Grosveld F, von Lindern M, Philipsen S, van Dijk TB. The mouse KLF1 Nan variant impairs nuclear condensation and erythroid maturation. PLoS One 2019; 14:e0208659. [PMID: 30921348 PMCID: PMC6438607 DOI: 10.1371/journal.pone.0208659] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 03/11/2019] [Indexed: 12/13/2022] Open
Abstract
Krüppel-like factor 1 (KLF1) is an essential transcription factor for erythroid development, as demonstrated by Klf1 knockout mice which die around E14 due to severe anemia. In humans, >140 KLF1 variants, causing different erythroid phenotypes, have been described. The KLF1 Nan variant, a single amino acid substitution (p.E339D) in the DNA binding domain, causes hemolytic anemia and is dominant over wildtype KLF1. Here we describe the effects of the KLF1 Nan variant during fetal development. We show that Nan embryos have defects in erythroid maturation. RNA-sequencing of the KLF1 Nan fetal liver cells revealed that Exportin 7 (Xpo7) was among the 782 deregulated genes. This nuclear exportin is implicated in terminal erythroid differentiation; in particular it is involved in nuclear condensation. Indeed, KLF1 Nan fetal liver cells had larger nuclei and reduced chromatin condensation. Knockdown of XPO7 in wildtype erythroid cells caused a similar phenotype. We propose that reduced expression of XPO7 is partially responsible for the erythroid defects observed in KLF1 Nan erythroid cells.
Collapse
Affiliation(s)
- Ileana Cantú
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Nynke Gillemans
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Steven Heshusius
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
- Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - Alex Maas
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Zeliha Ozgur
- Center for Biomics, Erasmus MC, Rotterdam, The Netherlands
| | | | - Frank Grosveld
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | | | - Sjaak Philipsen
- Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
- * E-mail:
| | | |
Collapse
|
39
|
Khabarova AA, Ryzhkova AS, Battulin NR. Reorganisation of chromatin during erythroid differentiation. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A totipotent zygote has unlimited potential for differentiation into all cell types found in an adult organism. During ontogenesis proliferating and maturing cells gradually lose their differentiation potential, limiting the spectrum of possible developmental transitions to a specific cell type. Following the initiation of the developmental program cells acquire specific morphological and functional properties. Deciphering the mechanisms that coordinate shifts in gene expression revealed a critical role of three-dimensional chromatin structure in the regulation of gene activity during lineage commitment. Several levels of DNA packaging have been recently identified using chromosome conformation capture based techniques such a Hi-C. It is now clear that chromatin regions with high transcriptional activity assemble into Mb-scale compartments in the nuclear space, distinct from transcriptionally silent regions. More locally chromatin is organized into topological domains, serving as functionally insulated units with cell type – specific regulatory loop interactions. However, molecular mechanisms establishing and maintaining such 3D organization are yet to be investigated. Recent focus on studying chromatin reorganization accompanying cell cycle progression and cellular differentiation partially explained some aspects of 3D genome folding. Throughout erythropoiesis cells undergo a dramatic reorganization of the chromatin landscape leading to global nuclear condensation and transcriptional silencing, followed by nuclear extrusion at the final stage of mammalian erythropoiesis. Drastic changes of genome architecture and function accompanying erythroid differentiation seem to be an informative model for studying the ways of how genome organization and dynamic gene activity are connected. Here we summarize current views on the role of global rearrangement of 3D chromatin structure in erythroid differentiation.
Collapse
Affiliation(s)
| | | | - N. R. Battulin
- Institute of Cytology and Genetics, SB RAS; Novosibirsk State University
| |
Collapse
|
40
|
Maea expressed by macrophages, but not erythroblasts, maintains postnatal murine bone marrow erythroblastic islands. Blood 2019; 133:1222-1232. [PMID: 30674470 DOI: 10.1182/blood-2018-11-888180] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/18/2019] [Indexed: 12/11/2022] Open
Abstract
The erythroblastic island (EI), formed by a central macrophage and developing erythroblasts (EBs), was first described decades ago and was recently shown to play an in vivo role in homeostatic and pathological erythropoiesis. The exact molecular mechanisms, however, mediating the interactions between macrophages and EBs remain unclear. Macrophage-EB attacher (Maea) has previously been suggested to mediate homophilic adhesion bounds bridging macrophages and EBs. Maea-deficient mice die perinatally with anemia and defective erythrocyte enucleation, suggesting a critical role in fetal erythropoiesis. Here, we generated conditional knockout mouse models of Maea to assess its cellular and postnatal contributions. Deletion of Maea in macrophages using Csf1r-Cre or CD169-Cre caused severe reductions of bone marrow (BM) macrophages, EBs, and in vivo island formation, whereas its deletion in the erythroid lineage using Epor-Cre had no such phenotype, suggesting a dominant role of Maea in the macrophage for BM erythropoiesis. Interestingly, Maea deletion in spleen macrophages did not alter their numbers or functions. Postnatal Maea deletion using Mx1-Cre or function inhibition using a novel monoclonal antibody also impaired BM erythropoiesis. These results indicate that Maea contributes to adult BM erythropoiesis by regulating the maintenance of macrophages and their interaction with EBs via an as-yet-unidentified EB receptor.
Collapse
|
41
|
Aoto M, Iwashita A, Mita K, Ohkubo N, Tsujimoto Y, Mitsuda N. Transferrin receptor 1 is required for enucleation of mouse erythroblasts during terminal differentiation. FEBS Open Bio 2019; 9:291-303. [PMID: 30761254 PMCID: PMC6356176 DOI: 10.1002/2211-5463.12573] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/04/2018] [Accepted: 12/04/2018] [Indexed: 12/25/2022] Open
Abstract
Enucleation is the process whereby the nucleus is extruded from the erythroblast during late stage mammalian erythropoiesis. However, the specific signaling pathways involved in this process remain unclear. To better understand the mechanisms underlying erythroblast enucleation, we investigated erythroblast enucleation using both the spleens of adult mice with phenylhydrazine‐induced anemia and mouse fetal livers. Our results indicated that both iron‐bound transferrin (holo‐Tf) and the small‐molecule iron transporter hinokitiol with iron ions (hinokitiol plus iron) promote hemoglobin synthesis and the enucleation of mouse spleen‐derived erythroblasts. Although an antitransferrin receptor 1 (TfR1) monoclonal antibody inhibited both enucleation and hemoglobin synthesis promoted by holo‐Tf, it inhibited only enucleation, but not hemoglobin synthesis, promoted by hinokitiol plus iron. Furthermore, siRNA against mouse TfR1 were found to suppress the enucleation of mouse fetal liver‐derived erythroblasts, and the endocytosis inhibitor MitMAB inhibited enucleation, hemoglobin synthesis, and the internalization of TfR1 promoted by both types of stimuli. Collectively, our results suggest that TfR1, iron ions, and endocytosis play important roles in mouse erythroblast enucleation.
Collapse
Affiliation(s)
- Mamoru Aoto
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Akiho Iwashita
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Kanako Mita
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Nobutaka Ohkubo
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| | - Yoshihide Tsujimoto
- Department of Molecular and Cellular Biology Research Center Osaka International Cancer Institute Japan
| | - Noriaki Mitsuda
- Department of Circulatory Physiology Graduate School of Medicine Ehime University Japan
| |
Collapse
|
42
|
Ovchynnikova E, Aglialoro F, von Lindern M, van den Akker E. The Shape Shifting Story of Reticulocyte Maturation. Front Physiol 2018; 9:829. [PMID: 30050448 PMCID: PMC6050374 DOI: 10.3389/fphys.2018.00829] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022] Open
Abstract
The final steps of erythropoiesis involve unique cellular processes including enucleation and reorganization of membrane proteins and the cytoskeleton to produce biconcave erythrocytes. Surprisingly this process is still poorly understood. In vitro erythropoiesis protocols currently produce reticulocytes rather than biconcave erythrocytes. In addition, immortalized lines and iPSC-derived erythroid cell suffer from low enucleation and suboptimal final maturation potential. In light of the increasing prospect to use in vitro produced erythrocytes as (personalized) transfusion products or as therapeutic delivery agents, the mechanisms driving this last step of erythropoiesis are in dire need of resolving. Here we review the elusive last steps of reticulocyte maturation with an emphasis on protein sorting during the defining steps of reticulocyte formation during enucleation and maturation.
Collapse
Affiliation(s)
- Elina Ovchynnikova
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Francesca Aglialoro
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Marieke von Lindern
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Emile van den Akker
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
43
|
Malik J, Lillis JA, Couch T, Getman M, Steiner LA. The Methyltransferase Setd8 Is Essential for Erythroblast Survival and Maturation. Cell Rep 2018; 21:2376-2383. [PMID: 29186677 DOI: 10.1016/j.celrep.2017.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/28/2017] [Accepted: 11/01/2017] [Indexed: 12/31/2022] Open
Abstract
Erythropoiesis is a highly regulated process that generates enucleate red blood cells from committed erythroid progenitors. Chromatin condensation culminating in enucleation is a defining feature of this process. Setd8 is the sole enzyme that can mono-methylate histone H4, lysine 20 and is highly expressed in erythroblasts compared to most other cell types. Erythroid Setd8 deletion results in embryonic lethality from severe anemia due to impaired erythroblast survival and proliferation. Setd8 protein levels are also uniquely regulated in erythroblasts, suggesting a cell-type-specific role for Setd8 during terminal maturation. Consistent with this hypothesis, Setd8 Δ/Δ erythroblasts have profound defects in transcriptional repression, chromatin condensation, and heterochromatin accumulation. Together, these results suggest that Setd8, used by most cells to promote mitotic chromatin condensation, is an essential aspect of the transcriptional repression and chromatin condensation that are hallmarks of terminal erythroid maturation.
Collapse
Affiliation(s)
- Jeffrey Malik
- Department of Pediatrics, University of Rochester, Rochester, NY 14620, USA
| | - Jacquelyn A Lillis
- Department of Pediatrics, University of Rochester, Rochester, NY 14620, USA; Genomics Research Center, University of Rochester, Rochester, NY 14620, USA
| | - Tyler Couch
- Department of Pediatrics, University of Rochester, Rochester, NY 14620, USA
| | - Michael Getman
- Department of Pediatrics, University of Rochester, Rochester, NY 14620, USA
| | - Laurie A Steiner
- Department of Pediatrics, University of Rochester, Rochester, NY 14620, USA.
| |
Collapse
|
44
|
Huang ZW, Fong CY, Gauthaman K, Sukumar P, Mahyuddin AP, Barrett AN, Bongso A, Choolani M. Biology of human primitive erythroblasts for application in noninvasive prenatal diagnosis. Prenat Diagn 2018; 38:673-684. [PMID: 29876942 DOI: 10.1002/pd.5295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 05/23/2018] [Accepted: 05/30/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Human primitive erythroblasts produced during early embryogenesis have been found in maternal circulation at early gestation and are considered good target cells for noninvasive prenatal diagnosis. We aimed to gain a better understanding of the biology of primitive erythroblasts and maximize their potential utility for noninvasive prenatal diagnosis. METHODS Cells were obtained from first trimester human placental tissues. Biological properties including surface antigen composition, differentiation, proliferation, enucleation, and degeneration were studied as gestation progressed. A microdroplet culture system was developed to observe the behavior of these cells in vitro. RESULTS Histology showed that primitive erythroblasts undergo maturation from polychromatic to orthochromatic erythroblasts and can differentiate spontaneously in vitro. Cell surface markers and nuclear gene expression suggest that the cells do not possess stemness properties, despite being primitive in nature. They have limited proliferative activity and highly deacetylated chromatin, but a microdroplet culture system can prolong their viability under normoxic conditions. No apoptosis was seen by 11 weeks' gestation, and there was no enucleation in vitro. CONCLUSION These properties confirm that viable cells with intact nuclei can be obtained at very early gestation for genetic analysis.
Collapse
Affiliation(s)
- Zhou-Wei Huang
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Chui-Yee Fong
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Kalamegam Gauthaman
- King Abdulaziz University, King Fahd Medical Research Centre, Jeddah, 21589, Saudi Arabia
| | - Ponnusamy Sukumar
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore.,Management Development Institute of Singapore, 501 Stirling Road, 148951, Singapore
| | - Aniza P Mahyuddin
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Angela N Barrett
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Ariff Bongso
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| | - Mahesh Choolani
- National University of Singapore, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, 119228, Singapore
| |
Collapse
|
45
|
Minetti G, Achilli C, Perotti C, Ciana A. Continuous Change in Membrane and Membrane-Skeleton Organization During Development From Proerythroblast to Senescent Red Blood Cell. Front Physiol 2018; 9:286. [PMID: 29632498 PMCID: PMC5879444 DOI: 10.3389/fphys.2018.00286] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
Within the context of erythropoiesis and the possibility of producing artificial red blood cells (RBCs) in vitro, a most critical step is the final differentiation of enucleated erythroblasts, or reticulocytes, to a fully mature biconcave discocyte, the RBC. Reviewed here is the current knowledge about this fundamental maturational process. By combining literature data with our own experimental evidence we propose that the early phase in the maturation of reticulocytes to RBCs is driven by a membrane raft-based mechanism for the sorting of disposable membrane proteins, mostly the no longer needed transferrin receptor (TfR), to the multivesicular endosome (MVE) as cargo of intraluminal vesicles that are subsequently exocytosed as exosomes, consistently with the seminal and original observation of Johnstone and collaborators of more than 30 years ago (Pan BT, Johnstone RM. Cell. 1983;33:967-978). According to a strikingly selective sorting process, the TfR becomes cargo destined to exocytosis while other molecules, including the most abundant RBC transmembrane protein, band 3, are completely retained in the cell membrane. It is also proposed that while this process could be operating in the early maturational steps in the bone marrow, additional mechanism(s) must be at play for the final removal of the excess reticulocyte membrane that is observed to occur in the circulation. This processing will most likely require the intervention of the spleen, whose function is also necessary for the continuous remodeling of the RBC membrane all along this cell's circulatory life.
Collapse
Affiliation(s)
- Giampaolo Minetti
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Cesare Achilli
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Cesare Perotti
- Servizio Immunoematologia e Medicina Trasfusionale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Annarita Ciana
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| |
Collapse
|
46
|
Huang Y, Hale J, Wang Y, Li W, Zhang S, Zhang J, Zhao H, Guo X, Liu J, Yan H, Yazdanbakhsh K, Huang G, Hillyer CD, Mohandas N, Chen L, Sun L, An X. SF3B1 deficiency impairs human erythropoiesis via activation of p53 pathway: implications for understanding of ineffective erythropoiesis in MDS. J Hematol Oncol 2018; 11:19. [PMID: 29433555 PMCID: PMC5810112 DOI: 10.1186/s13045-018-0558-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/23/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND SF3B1 is a core component of splicing machinery. Mutations in SF3B1 are frequently found in myelodysplastic syndromes (MDS), particularly in patients with refractory anemia with ringed sideroblasts (RARS), characterized by isolated anemia. SF3B1 mutations have been implicated in the pathophysiology of RARS; however, the physiological function of SF3B1 in erythropoiesis remains unknown. METHODS shRNA-mediated approach was used to knockdown SF3B1 in human CD34+ cells. The effects of SF3B1 knockdown on human erythroid cell differentiation, cell cycle, and apoptosis were assessed by flow cytometry. RNA-seq, qRT-PCR, and western blot analyses were used to define the mechanisms of phenotypes following knockdown of SF3B1. RESULTS We document that SF3B1 knockdown in human CD34+ cells leads to increased apoptosis and cell cycle arrest of early-stage erythroid cells and generation of abnormally nucleated late-stage erythroblasts. RNA-seq analysis of SF3B1-knockdown erythroid progenitor CFU-E cells revealed altered splicing of an E3 ligase Makorin Ring Finger Protein 1 (MKRN1) and subsequent activation of p53 pathway. Importantly, ectopic expression of MKRN1 rescued SF3B1-knockdown-induced alterations. Decreased expression of genes involved in mitosis/cytokinesis pathway including polo-like kinase 1 (PLK1) was noted in SF3B1-knockdown polychromatic and orthochromatic erythroblasts comparing to control cells. Pharmacologic inhibition of PLK1 also led to generation of abnormally nucleated erythroblasts. CONCLUSIONS These findings enabled us to identify novel roles for SF3B1 in human erythropoiesis and provided new insights into its role in regulating normal erythropoiesis. Furthermore, these findings have implications for improved understanding of ineffective erythropoiesis in MDS patients with SF3B1 mutations.
Collapse
Affiliation(s)
- Yumin Huang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
| | - John Hale
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY 10065 USA
| | - Yaomei Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Wei Li
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008 People’s Republic of China
| | - Shijie Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Jieying Zhang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, Changsha, 410078 People’s Republic of China
| | - Huizhi Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Xinhua Guo
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
| | - Jing Liu
- The State Key Laboratory of Medical Genetics and School of Life Sciences, Central South University, Changsha, 410078 People’s Republic of China
| | - Hongxia Yan
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY 10065 USA
| | - Karina Yazdanbakhsh
- Laboratory of Complement Biology, New York Blood Center, New York, NY 10065 USA
| | - Gang Huang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229 USA
| | | | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY 10065 USA
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| | - Ling Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
| | - Xiuli An
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000 Henan People’s Republic of China
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065 USA
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001 People’s Republic of China
| |
Collapse
|
47
|
Rogerson C, Bergamaschi D, O'Shaughnessy RFL. Uncovering mechanisms of nuclear degradation in keratinocytes: A paradigm for nuclear degradation in other tissues. Nucleus 2018; 9:56-64. [PMID: 29205081 PMCID: PMC5973266 DOI: 10.1080/19491034.2017.1412027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Eukaryotic nuclei are essential organelles, storing the majority of the cellular DNA, comprising the site of most DNA and RNA synthesis, controlling gene expression and therefore regulating cellular function. The majority of mammalian cells retain their nucleus throughout their lifetime, however, in three mammalian tissues the nucleus is entirely removed and its removal is essential for cell function. Lens fibre cells, erythroblasts and epidermal keratinocytes all lose their nucleus in the terminal differentiation pathways of these cell types. However, relatively little is known about the pathways that lead to complete nuclear removal and about how these pathways are regulated. In this review, we aim to discuss the current understanding of nuclear removal mechanisms in these three cell types and expand upon how recent studies into nuclear degradation in keratinocytes, an easily accessible experimental model, could contribute to a wider understanding of these molecular mechanisms in both health and pathology.
Collapse
Affiliation(s)
- Clare Rogerson
- a Centre for Cell Biology and Cutaneous Research , Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| | - Daniele Bergamaschi
- a Centre for Cell Biology and Cutaneous Research , Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| | - Ryan F L O'Shaughnessy
- a Centre for Cell Biology and Cutaneous Research , Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London , London , UK
| |
Collapse
|
48
|
|
49
|
Moras M, Lefevre SD, Ostuni MA. From Erythroblasts to Mature Red Blood Cells: Organelle Clearance in Mammals. Front Physiol 2017; 8:1076. [PMID: 29311991 PMCID: PMC5742207 DOI: 10.3389/fphys.2017.01076] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
Erythropoiesis occurs mostly in bone marrow and ends in blood stream. Mature red blood cells are generated from multipotent hematopoietic stem cells, through a complex maturation process involving several morphological changes to produce a highly functional specialized cells. In mammals, terminal steps involved expulsion of the nucleus from erythroblasts that leads to the formation of reticulocytes. In order to produce mature biconcave red blood cells, organelles and ribosomes are selectively eliminated from reticulocytes as well as the plasma membrane undergoes remodeling. The mechanisms involved in these last maturation steps are still under investigation. Enucleation involves dramatic chromatin condensation and establishment of the nuclear polarity, which is driven by a rearrangement of actin cytoskeleton and the clathrin-dependent generation of vacuoles at the nuclear-cytoplasmic junction. This process is favored by interaction between the erythroblasts and macrophages at the erythroblastic island. Mitochondria are eliminated by mitophagy. This is a macroautophagy pathway consisting in the engulfment of mitochondria into a double-membrane structure called autophagosome before degradation. Several mice knock-out models were developed to identify mitophagy-involved proteins during erythropoiesis, but whole mechanisms are not completely determined. Less is known concerning the clearance of other organelles, such as smooth and rough ER, Golgi apparatus and ribosomes. Understanding the modulators of organelles clearance in erythropoiesis may elucidate the pathogenesis of different dyserythropoietic diseases such as myelodysplastic syndrome, leukemia and anemia.
Collapse
Affiliation(s)
| | | | - Mariano A. Ostuni
- UMR-S1134 Integrated Biology of Red Blood Cell, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Institut National de la Transfusion Sanguine, Laboratoire d'Excellence GR-Ex, Paris, France
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Transcriptional regulators provide the molecular and biochemical basis for the cell specific properties and characteristics that follow from their central role in establishing tissue-restricted expression. Precise and sequential control of terminal cell divisions, nuclear condensation, and enucleation are defining characteristics within erythropoietic differentiation. This review is focused on KLF1, a central global regulator of this process. RECENT FINDINGS Studies in the past year have brought a number of proteins that are targets of KLF1 regulation into focus with respect to their roles in terminal erythroid differentiation. Many of these are involved in fine control of the cell cycle at both early (E2F2, Cyclin A2) and later (p18, p27, p19) stages of differentiation, or are directly involved in enucleation (p18, p27). Dramatic biophysical changes controlled at the nuclear lamin by caspase 3 enable histone release and nuclear condensation, whereas dematin association with structural proteins alters the timing of enucleation. Conditional ablation of mDia2 has established its role in late stage cell cycle and enucleation. SUMMARY Transcription factors such as KLF1, along with epigenetic modifiers, play crucial roles in establishing the proper onset and progression of terminal differentiation events. Studies from the past year show a remarkable multifaceted convergence on cell cycle control, and establish that the orthochromatic erythroblast stage is a critical nodal point for many of the effects on enucleation. These studies are relevant to understanding the underlying causes of anemia and hematologic disease where defective enucleation predicts a poor clinical outcome.
Collapse
|