1
|
Gao J, Luo F, Chen Q, Chen N, Wan J, Sun L, Cao Y, Ren H, Tu Y, Huang H, Cui F. The transferrin a signaling pathway mediates uranium-induced hematopoietic dysfunction. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 373:126077. [PMID: 40139297 DOI: 10.1016/j.envpol.2025.126077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/05/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE This study was designed to explore the toxic effects of Transferrin a(tfa)-mediated uranium exposure on the hematopoietic system. METHODS Zebrafish embryos were subjected to uranium nitrate solutions at concentrations of 50, 100, 250, and 500 μg/L for a defined period, followed by sample collection. The impact of uranium on hematopoietic system development in zebrafish was evaluated through hemoglobin staining, qRT-PCR, and in situ hybridization. RNA-Seq was utilized to detect differentially expressed genes (DEGs) in embryos exposed to 100 μg/L uranium, with subsequent bioinformatics analysis to confirm these DEGs. Furthermore, blood samples from patients with hematological disorders and impaired hematopoietic function were collected, and RNA-Seq was applied to identify DEGs. RESULTS Uranium exposure in zebrafish embryos led to reduced hemoglobin expression, with key transcription factors for primitive and definitive hematopoiesis being significantly downregulated at 100 μg/L uranium exposure. Overexpression of tfa resulted in a marked increase in hemoglobin content and upregulation of GATA1, a key factor in primitive hematopoiesis. Patients with hematopoietic dysfunction exhibited abnormalities in the tfa signaling pathway. CONCLUSION tfa plays a role in mediating the inhibitory effects of uranium on hematopoietic function.
Collapse
Affiliation(s)
- Jin Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Fajian Luo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Qiu Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Na Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Jun Wan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Liang Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Yiyao Cao
- Department of Occupational Health and Radiological Protection, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Hong Ren
- Department of Occupational Health and Radiological Protection, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Yu Tu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| | - Haiwen Huang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Fengmei Cui
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China.
| |
Collapse
|
2
|
Li M, Suzuki K, Wang M, Benner C, Ku M, Ma L, Kobari L, Kim NY, Montserrat N, Chang CJ, Liu G, Qu J, Xu J, Zhang Y, Aizawa E, Wu J, Douay L, Esteban CR, Belmonte JCI. Dynamic WNT signaling controls differentiation of hematopoietic progenitor cells from human pluripotent stem cells. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2816-0. [PMID: 40080269 DOI: 10.1007/s11427-024-2816-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/11/2024] [Indexed: 03/15/2025]
Abstract
Human pluripotent stem cells (hPSCs) can in theory give rise to any hematopoietic lineages, thereby offering opportunities for disease modeling, drug screening and cell therapies. However, gaps in our knowledge of the signaling requirements for the specification of human hematopoietic stem/progenitor cells (HSPCs), which lie at the apex of all hematopoietic lineages, greatly limit the potential of hPSC in hematological research and application. Transcriptomic analysis reveals aberrant regulation of WNT signaling during maturation of hPSC-derived hematopoietic progenitor cells (hPSC-HPCs), which results in higher mitochondria activity, misregulation of HOX genes, loss of self-renewal and precocious differentiation. These defects are partly due to the activation of the WNT target gene CDX2. Late-stage WNT inhibition improves the yield, self-renewal, multilineage differentiation, and transcriptional and metabolic profiles of hPSC-HPCs. Genome-wide mapping of transcription factor (TF) accessible chromatin reveals a significant overrepresentation of myeloid TF binding motifs in hPSC-HPCs, which could underlie their myeloid-biased lineage potential. Together our findings uncover a previously unappreciated dynamic requirement of the WNT signaling pathway during the specification of human HSPCs. Modulating the WNT pathway with small molecules normalizes the molecular differences between hPSC-HPCs and endogenous hematopoietic stem cells (HSCs), thereby representing a promising approach to improve the differentiation and function of hPSC-HPCs.
Collapse
Affiliation(s)
- Mo Li
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
- KAUST Center of Excellence for Smart Health (KCSH), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA.
| | - Keiichiro Suzuki
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Institute for Advanced Co-Creation Studies, Osaka University, Osaka, 560-8531, Japan
| | - Mengge Wang
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- KAUST Center of Excellence for Smart Health (KCSH), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Christopher Benner
- Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Manching Ku
- Next Generation Sequencing Core, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Li Ma
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Ladan Kobari
- Prolifération et Différentiation des Cellules Souches, UPMC Univ Paris 06, UMR_S938 CDR Saint-Antoine, Paris, F-75012, France
- Prolifération et Différentiation des Cellules Souches, INSERM, UMR_S938, Paris, F-75012, France
| | - Na Young Kim
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Nuria Montserrat
- Center for Regenerative Medicine in Barcelona, Barcelona, 08003, Spain
| | - Chan-Jung Chang
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Guanghui Liu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Qu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jinna Xu
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- KAUST Center of Excellence for Smart Health (KCSH), Thuwal, 23955-6900, Kingdom of Saudi Arabia
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Yingzi Zhang
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia
| | - Emi Aizawa
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Jun Wu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
| | - Luc Douay
- Prolifération et Différentiation des Cellules Souches, UPMC Univ Paris 06, UMR_S938 CDR Saint-Antoine, Paris, F-75012, France
- Unité d'Ingénierie et de Thérapie Cellulaire, EFS Ile de France, Créteil, F-94017, France
- Service d'Hématologie et immunologie biologique, AP-HP Hôpital Saint Antoine/Armand Trousseau, Paris, F-75012, France
| | - Concepcion Rodriguez Esteban
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA
- Altos Labs, San Diego, 92122, USA
| | - Juan Carlos Izpisua Belmonte
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Kingdom of Saudi Arabia.
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, 92037, USA.
- Altos Labs, San Diego, 92122, USA.
| |
Collapse
|
3
|
Hjazi A, Jasim SA, Al-Dhalimy AMB, Bansal P, Kaur H, Qasim MT, Mohammed IH, Deorari M, Jawad MA, Zwamel AH. HOXA9 versus HOXB9; particular focus on their controversial role in tumor pathogenesis. J Appl Genet 2024; 65:473-492. [PMID: 38753266 DOI: 10.1007/s13353-024-00868-x] [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: 02/26/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 08/09/2024]
Abstract
The Homeobox (HOX) gene family is essential to regulating cellular processes because it maintains the exact coordination required for tissue homeostasis, cellular differentiation, and embryonic development. The most distinctive feature of this class of genes is the presence of the highly conserved DNA region known as the homeobox, which is essential for controlling their regulatory activities. Important players in the intricate process of genetic regulation are the HOX genes. Many diseases, especially in the area of cancer, are linked to their aberrant functioning. Due to their distinctive functions in biomedical research-particularly in the complex process of tumor advancement-HOXA9 and HOXB9 have drawn particular attention. HOXA9 and HOXB9 are more significant than what is usually connected with HOX genes since they have roles in the intricate field of cancer and beyond embryonic processes. The framework for a focused study of the different effects of HOXA9 and HOXB9 in the context of tumor biology is established in this study.
Collapse
Affiliation(s)
- Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | | | | | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-Be) University, Bengaluru, Karnataka, 560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Maytham T Qasim
- College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Nasiriyah, 64001, Iraq
| | - Israa Hussein Mohammed
- College of Nursing, National University of Science and Technology, Dhi Qar, Nasiriyah, Iraq
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Mohammed Abed Jawad
- Department of Medical Laboratories Technology, Al-Nisour University College, Baghdad, Iraq
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
4
|
Wellik DM. Hox genes and patterning the vertebrate body. Curr Top Dev Biol 2024; 159:1-27. [PMID: 38729674 DOI: 10.1016/bs.ctdb.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The diversity of vertebrate body plans is dizzying, yet stunning for the many things they have in common. Vertebrates have inhabited virtually every part of the earth from its coldest to warmest climates. They locomote by swimming, flying, walking, slithering, or climbing, or combinations of these behaviors. And they exist in many different sizes, from the smallest of frogs, fish and lizards to giraffes, elephants, and blue whales. Despite these differences, vertebrates follow a remarkably similar blueprint for the establishment of their body plan. Within the relatively small amount of time required to complete gastrulation, the process through which the three germ layers, ectoderm, mesoderm, and endoderm are created, the embryo also generates its body axis and is simultaneously patterned. For the length of this axis, the genes that distinguish the neck from the rib cage or the trunk from the sacrum are the Hox genes. In vertebrates, there was evolutionary pressure to maintain this set of genes in the organism. Over the past decades, much has been learned regarding the regulatory mechanisms that ensure the appropriate expression of these genes along the main body axes. Genetic functions continue to be explored though much has been learned. Much less has been discerned on the identity of co-factors used by Hox proteins for the specificity of transcriptional regulation or what downstream targets and pathways are critical for patterning events, though there are notable exceptions. Current work in the field is demonstrating that Hox genes continue to function in many organs long after directing early patterning events. It is hopeful continued research will shed light on remaining questions regarding mechanisms used by this important and conserved set of transcriptional regulators.
Collapse
Affiliation(s)
- Deneen M Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States.
| |
Collapse
|
5
|
Kayvanjoo AH, Splichalova I, Bejarano DA, Huang H, Mauel K, Makdissi N, Heider D, Tew HM, Balzer NR, Greto E, Osei-Sarpong C, Baßler K, Schultze JL, Uderhardt S, Kiermaier E, Beyer M, Schlitzer A, Mass E. Fetal liver macrophages contribute to the hematopoietic stem cell niche by controlling granulopoiesis. eLife 2024; 13:e86493. [PMID: 38526524 PMCID: PMC11006421 DOI: 10.7554/elife.86493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 03/23/2024] [Indexed: 03/26/2024] Open
Abstract
During embryogenesis, the fetal liver becomes the main hematopoietic organ, where stem and progenitor cells as well as immature and mature immune cells form an intricate cellular network. Hematopoietic stem cells (HSCs) reside in a specialized niche, which is essential for their proliferation and differentiation. However, the cellular and molecular determinants contributing to this fetal HSC niche remain largely unknown. Macrophages are the first differentiated hematopoietic cells found in the developing liver, where they are important for fetal erythropoiesis by promoting erythrocyte maturation and phagocytosing expelled nuclei. Yet, whether macrophages play a role in fetal hematopoiesis beyond serving as a niche for maturing erythroblasts remains elusive. Here, we investigate the heterogeneity of macrophage populations in the murine fetal liver to define their specific roles during hematopoiesis. Using a single-cell omics approach combined with spatial proteomics and genetic fate-mapping models, we found that fetal liver macrophages cluster into distinct yolk sac-derived subpopulations and that long-term HSCs are interacting preferentially with one of the macrophage subpopulations. Fetal livers lacking macrophages show a delay in erythropoiesis and have an increased number of granulocytes, which can be attributed to transcriptional reprogramming and altered differentiation potential of long-term HSCs. Together, our data provide a detailed map of fetal liver macrophage subpopulations and implicate macrophages as part of the fetal HSC niche.
Collapse
Affiliation(s)
- Amir Hossein Kayvanjoo
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Iva Splichalova
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - David Alejandro Bejarano
- Quantitative Systems Biology, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Hao Huang
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Katharina Mauel
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Nikola Makdissi
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - David Heider
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Hui Ming Tew
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Nora Reka Balzer
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Eric Greto
- Department of Internal Medicine 3-Rheumatology and Immunology, Deutsches Zentrum für Immuntherapie (DZI) and FAU Profile Center Immunomedicine (FAU I-MED), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum ErlangenErlangenGermany
- Exploratory Research Unit, Optical Imaging Centre ErlangenErlangenGermany
| | - Collins Osei-Sarpong
- Immunogenomics & Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)BonnGermany
| | - Kevin Baßler
- Genomics & Immunoregulation, LIMES Institute, University of BonnBonnGermany
| | - Joachim L Schultze
- Genomics & Immunoregulation, LIMES Institute, University of BonnBonnGermany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)BonnGermany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of BonnBonnGermany
| | - Stefan Uderhardt
- Department of Internal Medicine 3-Rheumatology and Immunology, Deutsches Zentrum für Immuntherapie (DZI) and FAU Profile Center Immunomedicine (FAU I-MED), Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum ErlangenErlangenGermany
- Exploratory Research Unit, Optical Imaging Centre ErlangenErlangenGermany
| | - Eva Kiermaier
- Immune and Tumor Biology, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Marc Beyer
- Immunogenomics & Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)BonnGermany
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE)BonnGermany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of BonnBonnGermany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| | - Elvira Mass
- Developmental Biology of the Immune System, Life & Medical Sciences (LIMES) Institute, University of BonnBonnGermany
| |
Collapse
|
6
|
Aryal S, Lu R. HOXA9 Regulome and Pharmacological Interventions in Leukemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:405-430. [PMID: 39017854 DOI: 10.1007/978-3-031-62731-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
HOXA9, an important transcription factor (TF) in hematopoiesis, is aberrantly expressed in numerous cases of both acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) and is a strong indicator of poor prognosis in patients. HOXA9 is a proto-oncogene which is both sufficient and necessary for leukemia transformation. HOXA9 expression in leukemia correlates with patient survival outcomes and response to therapy. Chromosomal transformations (such as NUP98-HOXA9), mutations, epigenetic dysregulation (e.g., MLL- MENIN -LEDGF complex or DOT1L/KMT4), transcription factors (such as USF1/USF2), and noncoding RNA (such as HOTTIP and HOTAIR) regulate HOXA9 mRNA and protein during leukemia. HOXA9 regulates survival, self-renewal, and progenitor cell cycle through several of its downstream target TFs including LMO2, antiapoptotic BCL2, SOX4, and receptor tyrosine kinase FLT3 and STAT5. This dynamic and multilayered HOXA9 regulome provides new therapeutic opportunities, including inhibitors targeting DOT1L/KMT4, MENIN, NPM1, and ENL proteins. Recent findings also suggest that HOXA9 maintains leukemia by actively repressing myeloid differentiation genes. This chapter summarizes the recent advances understanding biochemical mechanisms underlying HOXA9-mediated leukemogenesis, the clinical significance of its abnormal expression, and pharmacological approaches to treat HOXA9-driven leukemia.
Collapse
Affiliation(s)
- Sajesan Aryal
- Department of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Rui Lu
- Department of Medicine, Division of Hematology/Oncology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
7
|
Xu Z, Yang J, Xin X, Liu C, Li L, Mei X, Li M. Merits and challenges of iPSC-derived organoids for clinical applications. Front Cell Dev Biol 2023; 11:1188905. [PMID: 37305682 PMCID: PMC10250752 DOI: 10.3389/fcell.2023.1188905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 04/18/2023] [Indexed: 06/13/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have entered an unprecedented state of development since they were first generated. They have played a critical role in disease modeling, drug discovery, and cell replacement therapy, and have contributed to the evolution of disciplines such as cell biology, pathophysiology of diseases, and regenerative medicine. Organoids, the stem cell-derived 3D culture systems that mimic the structure and function of organs in vitro, have been widely used in developmental research, disease modeling, and drug screening. Recent advances in combining iPSCs with 3D organoids are facilitating further applications of iPSCs in disease research. Organoids derived from embryonic stem cells, iPSCs, and multi-tissue stem/progenitor cells can replicate the processes of developmental differentiation, homeostatic self-renewal, and regeneration due to tissue damage, offering the potential to unravel the regulatory mechanisms of development and regeneration, and elucidate the pathophysiological processes involved in disease mechanisms. Herein, we have summarized the latest research on the production scheme of organ-specific iPSC-derived organoids, the contribution of these organoids in the treatment of various organ-related diseases, in particular their contribution to COVID-19 treatment, and have discussed the unresolved challenges and shortcomings of these models.
Collapse
Affiliation(s)
- Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
- Department of Clinical Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiaxu Yang
- Department of Neonatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xianyi Xin
- Department of Pediatric Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chengrun Liu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Xianglin Mei
- Department of pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| |
Collapse
|
8
|
Abstract
Hox genes encode evolutionarily conserved transcription factors that are essential for the proper development of bilaterian organisms. Hox genes are unique because they are spatially and temporally regulated during development in a manner that is dictated by their tightly linked genomic organization. Although their genetic function during embryonic development has been interrogated, less is known about how these transcription factors regulate downstream genes to direct morphogenetic events. Moreover, the continued expression and function of Hox genes at postnatal and adult stages highlights crucial roles for these genes throughout the life of an organism. Here, we provide an overview of Hox genes, highlighting their evolutionary history, their unique genomic organization and how this impacts the regulation of their expression, what is known about their protein structure, and their deployment in development and beyond.
Collapse
Affiliation(s)
- Katharine A. Hubert
- Program in Genetics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Deneen M. Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
9
|
Steens J, Klein D. HOX genes in stem cells: Maintaining cellular identity and regulation of differentiation. Front Cell Dev Biol 2022; 10:1002909. [PMID: 36176275 PMCID: PMC9514042 DOI: 10.3389/fcell.2022.1002909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Stem cells display a unique cell type within the body that has the capacity to self-renew and differentiate into specialized cell types. Compared to pluripotent stem cells, adult stem cells (ASC) such as mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) exhibit restricted differentiation capabilities that are limited to cell types typically found in the tissue of origin, which implicates that there must be a certain code or priming determined by the tissue of origin. HOX genes, a subset of homeobox genes encoding transcription factors that are generally repressed in undifferentiated pluripotent stem cells, emerged here as master regulators of cell identity and cell fate during embryogenesis, and in maintaining this positional identity throughout life as well as specifying various regional properties of respective tissues. Concurrently, intricate molecular circuits regulated by diverse stem cell-typical signaling pathways, balance stem cell maintenance, proliferation and differentiation. However, it still needs to be unraveled how stem cell-related signaling pathways establish and regulate ASC-specific HOX expression pattern with different temporal-spatial topography, known as the HOX code. This comprehensive review therefore summarizes the current knowledge of specific ASC-related HOX expression patterns and how these were integrated into stem cell-related signaling pathways. Understanding the mechanism of HOX gene regulation in stem cells may provide new ways to manipulate stem cell fate and function leading to improved and new approaches in the field of regenerative medicine.
Collapse
|
10
|
Sidhu I, Barwe SP, Kiick KL, Kolb EA, Gopalakrishnapillai A. A 3-D hydrogel based system for hematopoietic differentiation and its use in modeling down syndrome associated transient myeloproliferative disorder. Biomater Sci 2021; 9:6266-6281. [PMID: 34369483 PMCID: PMC8570143 DOI: 10.1039/d1bm00442e] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Induced pluripotent stem cells (iPSCs) provide an extraordinary tool for disease modeling owing to their potential to differentiate into the desired cell type. The differentiation of iPSCs is typically performed on 2-dimensional monolayers of stromal cell or animal tissue derived extracellular matrices. Recent advancements in disease modeling have utilized iPSCs in 3-dimensional (3D) cultures to study diseases such as muscular dystrophy, cardiomyopathy, and pulmonary fibrosis. However, these approaches are yet to be explored in modeling the hematological malignancies. Transient myeloproliferative disorder (TMD) is a preleukemic stage, which is induced in 10-20% of children with trisomy 21 possessing the pathognomonic mutation in the transcription factor GATA1. In this study, we established a synthetic 3D iPSC culture system for modeling TMD via hematopoietic differentiation of customized iPSCs. A chemically cross-linkable PEG hydrogel decorated with integrin binding peptide was found to be permissive of hematopoietic differentiation of iPSCs. It provided a cost-effective system for the generation of hematopoietic stem and progenitor cells (HSPCs) with higher yield of early HSPCs compared to traditional 2D culture on Matrigel coated dishes. Characterization of the HSPCs produced from the iPSC lines cultured in 3D showed that the erythroid population was reduced whereas the megakaryoid and myeloid populations were significantly increased in GATA1 mutant trisomic line compared to disomic or trisomic lines with wild-type GATA1, consistent with TMD characteristics. In conclusion, we have identified a cost-effective tunable 3D hydrogel system to model TMD.
Collapse
Affiliation(s)
- Ishnoor Sidhu
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
- University of Delaware, Newark, DE 19711, USA
| | - Sonali P Barwe
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
- University of Delaware, Newark, DE 19711, USA
| | | | - E Anders Kolb
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
| | - Anilkumar Gopalakrishnapillai
- Nemours Centers for Childhood Cancer Research and Cancer & Blood Disorders, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.
- University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
11
|
Specific Blood Cells Derived from Pluripotent Stem Cells: An Emerging Field with Great Potential in Clinical Cell Therapy. Stem Cells Int 2021; 2021:9919422. [PMID: 34434242 PMCID: PMC8380505 DOI: 10.1155/2021/9919422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/06/2021] [Accepted: 08/02/2021] [Indexed: 11/18/2022] Open
Abstract
Widely known for self-renewal and multilineage differentiation, stem cells can be differentiated into all specialized tissues and cells in the body. In the past few years, a number of researchers have focused on deriving hematopoietic stem cells (HSCs) from pluripotent stem cells (PSCs) as alternative sources for clinic. Existing findings demonstrated that it is feasible to obtain HSCs and certain mature blood lineages from PSCs, except for several issues to be addressed. This short review outlines the technologies used for hematopoietic differentiation in recent years. In addition, the therapeutic value of PSCs as a potential source of various blood cells is also discussed as well as its challenges and directions in future clinical applications.
Collapse
|
12
|
Abstract
Background and aim: The pathogenesis of β-thalassemia has been attributed to ineffective erythropoiesis. The function of Hox genes in normal haematopoiesis has been widely studied using gene expression analysis. The aim of this study is to evaluate the expression of HoxA9, and HoxA5 genes in beta-thalassemia.Materials and methods: Children with thalassemia major, thalassemia intermediate, and age and sex-matched healthy controls (n = 50/group) were enrolled. Detection of HoxA5 and HoxA9 mRNA expression was performed by real-time polymerase chain reaction (RT-PCR).Results: Expression of HoxA9 increased in a direct linear trend (median 0.5 in controls, 2.4 in intermediate disease, 4.1 in major disease, p = 0.001) and generally correlated with the red cell count, haematocrit, ferritin and levels of beta-globin. In those with thalassemia major, the relative change of HoxA9 was linked to transfusion history, the white blood cell count, ferritin, and beta-globin (all r > 0.5, p < 0.001). Levels of HoxA9 were superior to HoxA5 in differentiating controls from thalassemia intermedia, whilst both differentiated major from the intermediate disease.Conclusion: This study highlights the importance of HoxA genes in early identification of patients, at high risk of developing complications, as it allows specific measures to delay the progression of the disease. HoxA gene expression is a promising diagnostic and prognostic marker in patients with β-thalassemia.
Collapse
Affiliation(s)
- Eae Badr
- Department of Medical Biochemistry and Molecular Biology, Menoufia University, Shibin Al Kawm, Egypt
| | - Ie-T El-Sayed
- Department of Medical Biochemistry and Molecular Biology, Menoufia University, Shibin Al Kawm, Egypt.,Department of Chemistry, Biochemistry Section, Menoufia University, Shibin Al Kawm, Egypt
| | - Mkr Alasadi
- Department of Chemistry, Biochemistry Section, Menoufia University, Shibin Al Kawm, Egypt
| |
Collapse
|
13
|
Li W, Liu D, Zheng F, Zeng Z, Cai W, Luan S, Hong X, Tang D, Yin LH, Dai Y. Generation of Systemic Lupus Erythematosus Patient-Derived Induced Pluripotent Stem Cells from Blood. Stem Cells Dev 2021; 30:227-233. [PMID: 33397195 DOI: 10.1089/scd.2020.0194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease characterized by the production of multiple autoimmune antibodies and potentially involves any organ or tissue with a broad range of clinical manifestations. Conventional therapy still utilizes glucocorticoids and immunosuppressants. However, some patients show inadequate responses to glucocorticoids and immunosuppression, which may induce secondary immune dysfunction and severe infection as well as lead to an increased tumor risk. The lack of in vitro models has hampered progress in understanding and treating SLE. Patient-derived induced pluripotent stem cells (iPSCs) may provide a unique opportunity for modeling in vitro diseases as well as a platform for drug screening in individual patients. We isolated peripheral blood mononuclear cells from blood to explore the establishment of an in vitro model platform for SLE and directly purified CD34+ cells and seeded them for expansion. CD34+ cells were forced to express seven pluripotency factors, OCT4, SOX2, NANOG, LIN28, c-MYC, KLF4, and SV40LT, through transduction in lentiviral vectors. The morphological characteristics of induced pluripotent stem-like cells, such as prominent nucleoli and a high nucleus-to-cytoplasm ratio, were observed. The pluripotency of established SLE patient-derived iPSCs was confirmed by the expression of embryonic stem cell (ESC) markers and the ability of cells to differentiate into multiple cell lines. SLE patient-derived iPSCs exhibited human ESC properties, including morphology; growth characteristics; expression of pluripotency, genes, and surface markers; and teratoma formation. In conclusion, we generated SLE patient-derived iPSCs and validated their pluripotency. This study is a first but critical step that can provide a model platform for research aimed at understanding the SLE mechanism, which may lead to the discovery of new targets or compounds for the treatment of this disease.
Collapse
Affiliation(s)
- Weilong Li
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.,Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Dongzhou Liu
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Fengping Zheng
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Zhipeng Zeng
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Wanxia Cai
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Shaodong Luan
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Xiaoping Hong
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China
| | - Donge Tang
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.,Guangxi Key Laboratory of Metabolic Disease Research, Central Laboratory of Guilin NO. 924 Hospital, Guilin, China
| | - Liang-Hong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yong Dai
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, China.,Guangxi Key Laboratory of Metabolic Disease Research, Central Laboratory of Guilin NO. 924 Hospital, Guilin, China
| |
Collapse
|
14
|
Qiu J, Nordling S, Vasavada HH, Butcher EC, Hirschi KK. Retinoic Acid Promotes Endothelial Cell Cycle Early G1 State to Enable Human Hemogenic Endothelial Cell Specification. Cell Rep 2020; 33:108465. [PMID: 33264627 PMCID: PMC8105879 DOI: 10.1016/j.celrep.2020.108465] [Citation(s) in RCA: 9] [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: 02/18/2020] [Revised: 08/27/2020] [Accepted: 11/10/2020] [Indexed: 12/01/2022] Open
Abstract
Development of blood-forming (hemogenic) endothelial cells that give rise to hematopoietic stem and progenitor cells (HSPCs) is critical during embryogenesis to generate the embryonic and postnatal hematopoietic system. We previously demonstrated that the specification of murine hemogenic endothelial cells is promoted by retinoic acid (RA) signaling and requires downstream endothelial cell cycle control. Whether this mechanism is conserved in human hemogenic endothelial cell specification is unknown. Here, we present a protocol to derive primordial endothelial cells from human embryonic stem cells and promote their specification toward hemogenic endothelial cells. Furthermore, we demonstrate that RA treatment significantly increases human hemogenic endothelial cell specification. That is, RA promotes endothelial cell cycle arrest to enable RA-induced instructive signals to upregulate the genes needed for hematopoietic transition. These insights provide guidance for the ex vivo generation of autologous human hemogenic endothelial cells that are needed to produce human HSPCs for regenerative medicine applications.
Collapse
Affiliation(s)
- Jingyao Qiu
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06520, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sofia Nordling
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hema H Vasavada
- Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA; The Center for Molecular Biology and Medicine, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Karen K Hirschi
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06520, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
15
|
Águila S, Cuenca-Zamora E, Martínez C, Teruel-Montoya R. MicroRNAs in Platelets: Should I Stay or Should I Go? Platelets 2020. [DOI: 10.5772/intechopen.93181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this chapter, we discuss different topics always using the microRNA as the guiding thread of the review. MicroRNAs, member of small noncoding RNAs family, are an important element involved in gene expression. We cover different issues such as their importance in the differentiation and maturation of megakaryocytes (megakaryopoiesis), as well as the role in platelets formation (thrombopoiesis) focusing on the described relationship between miRNA and critical myeloid lineage transcription factors such as RUNX1, chemokines receptors as CRCX4, or central hormones in platelet homeostasis like TPO, as well as its receptor (MPL) and the TPO signal transduction pathway, that is JAK/STAT. In addition to platelet biogenesis, we review the microRNA participation in platelets physiology and function. This review also introduces the use of miRNAs as biomarkers of platelet function since the detection of pathogenic situations or response to therapy using these noncoding RNAs is getting increasing interest in disease management. Finally, this chapter describes the participation of platelets in cellular interplay, since extracellular vesicles have been demonstrated to have the ability to deliver microRNAs to others cells, modulating their function through intercellular communication, redefining the extracellular vesicles from the so-called “platelet dust” to become mediators of intercellular communication.
Collapse
|
16
|
Li J, Zhou Z, Sun HX, Ouyang W, Dong G, Liu T, Ge L, Zhang X, Liu C, Gu Y. Transcriptome Analyses of β-Thalassemia -28(A>G) Mutation Using Isogenic Cell Models Generated by CRISPR/Cas9 and Asymmetric Single-Stranded Oligodeoxynucleotides (assODNs). Front Genet 2020; 11:577053. [PMID: 33193694 PMCID: PMC7580707 DOI: 10.3389/fgene.2020.577053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/01/2020] [Indexed: 01/11/2023] Open
Abstract
β-thalassemia, caused by mutations in the human hemoglobin β (HBB) gene, is one of the most common genetic diseases in the world. The HBB -28(A>G) mutation is one of the five most common mutations in Chinese patients with β-thalassemia. However, few studies have been conducted to understand how this mutation affects the expression of pathogenesis-related genes, including globin genes, due to limited homozygote clinical materials. Therefore, we developed an efficient technique using CRISPR/Cas9 combined with asymmetric single-stranded oligodeoxynucleotides (assODNs) to generate a K562 cell model with HBB -28(A>G) named K562-28(A>G). Then, we systematically analyzed the differences between K562-28(A>G) and K562 at the transcriptome level by high-throughput RNA-seq before and after erythroid differentiation. We found that the HBB -28(A>G) mutation not only disturbed the transcription of HBB, but also decreased the expression of HBG, which may further aggravate the thalassemia phenotype and partially explain the more severe clinical outcome of β-thalassemia patients with the HBB -28(A>G) mutation. Moreover, we found that the K562-28(A>G) cell line is more sensitive to hypoxia and shows a defective erythrogenic program compared with K562 before differentiation. Importantly, all abovementioned abnormalities in K562-28(A>G) were reversed after correction of this mutation with CRISPR/Cas9 and assODNs, confirming the specificity of these phenotypes. Overall, this is the first time to analyze the effects of the HBB -28(A>G) mutation at the whole-transcriptome level based on isogenic cell lines, providing a landscape for further investigation of the mechanism of β-thalassemia with the HBB -28(A>G) mutation.
Collapse
Affiliation(s)
- Jing Li
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Ziheng Zhou
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Hai-Xi Sun
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
- Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Wenjie Ouyang
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Guoyi Dong
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Tianbin Liu
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Lei Ge
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Xiuqing Zhang
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, Shenzhen Key Laboratory of Genomics, BGI-Shenzhen, Shenzhen, China
| | - Chao Liu
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Ying Gu
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen, China
| |
Collapse
|
17
|
Vaschetto LM, Editor G. The Critical Role of Epigenetic Regulation in Developmental Programming of Higher Organisms. Curr Genomics 2019; 20:403-404. [PMID: 32476996 PMCID: PMC7235389 DOI: 10.2174/138920292006191206141546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Luis María Vaschetto
- Agronomy, Horticulture & Plant Science Department
- South Dakota State University
- Brookings, SD
- USA
| | - Guest Editor
- Agronomy, Horticulture & Plant Science Department
- South Dakota State University
- Brookings, SD
- USA
| |
Collapse
|