1
|
Haniffa M, Maartens A, Winheim E, Jardine L. Decoding the human prenatal immune system with single-cell multi-omics. Nat Rev Immunol 2024:10.1038/s41577-024-01099-1. [PMID: 39482372 DOI: 10.1038/s41577-024-01099-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 11/03/2024]
Abstract
The human immune system is made up of a huge variety of cell types each with unique functions. Local networks of resident immune cells are poised to sense and protect against pathogen entry, whereas more widespread innate and adaptive immune networks provide first rapid, then long-lasting and targeted responses. However, how we develop such a diverse and complex system remains unknown. Studying human development directly has been challenging in the past, but recent advances in single-cell and spatial genomics, together with the co-ordinated efforts of the Human Cell Atlas and other initiatives, have led to new studies that map the development of the human immune system in unprecedented detail. In this Review, we consider the timings, transitions, cell types and tissue microenvironments that are crucial for building the human immune system. We also compare and contrast the human system with model species and in vitro systems, and discuss how an understanding of prenatal immune system development will improve our knowledge of human disease.
Collapse
Affiliation(s)
- Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- National Institute for Health Research (NIHR) Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
- Department of Dermatology, Newcastle upon Tyne Hospitals Foundation Trust, Newcastle upon Tyne, UK.
| | - Aidan Maartens
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Elena Winheim
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
2
|
Ng ES, Sarila G, Li JY, Edirisinghe HS, Saxena R, Sun S, Bruveris FF, Labonne T, Sleebs N, Maytum A, Yow RY, Inguanti C, Motazedian A, Calvanese V, Capellera-Garcia S, Ma F, Nim HT, Ramialison M, Bonifer C, Mikkola HKA, Stanley EG, Elefanty AG. Long-term engrafting multilineage hematopoietic cells differentiated from human induced pluripotent stem cells. Nat Biotechnol 2024:10.1038/s41587-024-02360-7. [PMID: 39223325 DOI: 10.1038/s41587-024-02360-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/20/2024] [Indexed: 09/04/2024]
Abstract
Hematopoietic stem cells (HSCs) derived from human induced pluripotent stem cells (iPS cells) have important biomedical applications. We identified differentiation conditions that generate HSCs defined by robust long-term multilineage engraftment in immune-deficient NOD,B6.Prkdcscid Il2rgtm1Wjl/SzJ KitW41/W41 mice. We guided differentiating iPS cells, as embryoid bodies in a defined culture medium supplemented with retinyl acetate, through HOXA-patterned mesoderm to hemogenic endothelium specified by bone morphogenetic protein 4 and vascular endothelial growth factor (VEGF). Removal of VEGF facilitated an efficient endothelial-to-hematopoietic transition, evidenced by release into the culture medium of CD34+ blood cells, which were cryopreserved. Intravenous transplantation of two million thawed CD34+ cells differentiated from four independent iPS cell lines produced multilineage bone marrow engraftment in 25-50% of immune-deficient recipient mice. These functionally defined, multipotent CD34+ hematopoietic cells, designated iPS cell-derived HSCs (iHSCs), produced levels of engraftment similar to those achieved following umbilical cord blood transplantation. Our study provides a step toward the goal of generating HSCs for clinical translation.
Collapse
Grants
- GNT1117596,GNT1068866, GNT1129861,GNT2012535 Department of Health | National Health and Medical Research Council (NHMRC)
- GNT1164577, GNT2012936 Department of Health | National Health and Medical Research Council (NHMRC)
- GNT2012535 Department of Health | National Health and Medical Research Council (NHMRC)
- GNT1186019 Department of Health | National Health and Medical Research Council (NHMRC)
- GNT1068866, GNT1129861, GNT2012535 Department of Health | National Health and Medical Research Council (NHMRC)
- GNT1068866, GNT1129861, GNT1186019 Department of Health | National Health and Medical Research Council (NHMRC)
- GNT1079004, GNT1068866, GNT1129861, GNT1186019 Department of Health | National Health and Medical Research Council (NHMRC)
- RT3-07763 California Institute for Regenerative Medicine (CIRM)
- NNF21CC0073729 Novo Nordisk Fonden (Novo Nordisk Foundation)
- NIH 1RO1DK125097-01 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- IPD2 2018-06635 Vetenskapsrådet (Swedish Research Council)
- BB/R014809/1 RCUK | Biotechnology and Biological Sciences Research Council (BBSRC)
Collapse
Affiliation(s)
- Elizabeth S Ng
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia.
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia.
| | - Gulcan Sarila
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Jacky Y Li
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Hasindu S Edirisinghe
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Ritika Saxena
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Shicheng Sun
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Changping Laboratory, Beijing, China
| | - Freya F Bruveris
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Tanya Labonne
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Nerida Sleebs
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Alexander Maytum
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Raymond Y Yow
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Chantelle Inguanti
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Ali Motazedian
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Vincenzo Calvanese
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
- Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Sandra Capellera-Garcia
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hieu T Nim
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Mirana Ramialison
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Constanze Bonifer
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Hanna K A Mikkola
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | - Edouard G Stanley
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Andrew G Elefanty
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia.
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research Institute, Parkville, Victoria, Australia.
| |
Collapse
|
3
|
Bello AB, Canlas KKV, Kim D, Park H, Lee SH. Stepwise dual-release microparticles of BMP-4 and SCF in induced pluripotent stem cell spheroids enhance differentiation into hematopoietic stem cells. J Control Release 2024; 371:386-405. [PMID: 38844177 DOI: 10.1016/j.jconrel.2024.06.011] [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] [Received: 12/26/2023] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.
Collapse
Affiliation(s)
- Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul 04620, Republic of Korea; School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea
| | | | - Deogil Kim
- Department of Biomedical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
4
|
Kidder BL. Decoding the universal human chromatin landscape through teratoma-based profiling. Nucleic Acids Res 2024; 52:3589-3606. [PMID: 38281248 PMCID: PMC11039989 DOI: 10.1093/nar/gkae021] [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: 05/02/2023] [Revised: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 01/30/2024] Open
Abstract
Teratoma formation is key for evaluating differentiation of human pluripotent stem cells into embryonic germ layers and serves as a model for understanding stem cell differentiation and developmental processes. Its potential for insights into epigenome and transcriptome profiling is significant. This study integrates the analysis of the epigenome and transcriptome of hESC-generated teratomas, comparing transcriptomes between hESCs and teratomas. It employs cell type-specific expression patterns from single-cell data to deconvolve RNA-Seq data and identify cell types within teratomas. Our results provide a catalog of activating and repressive histone modifications, while also elucidating distinctive features of chromatin states. Construction of an epigenetic signature matrix enabled the quantification of diverse cell populations in teratomas and enhanced the ability to unravel the epigenetic landscape in heterogeneous tissue contexts. This study also includes a single cell multiome atlas of expression (scRNA-Seq) and chromatin accessibility (scATAC-Seq) of human teratomas, further revealing the complexity of these tissues. A histology-based digital staining tool further complemented the annotation of cell types in teratomas, enhancing our understanding of their cellular composition. This research is a valuable resource for examining teratoma epigenomic and transcriptomic landscapes and serves as a model for epigenetic data comparison.
Collapse
Affiliation(s)
- Benjamin L Kidder
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
5
|
Palma LG, Bigas A. Making Human Hematopoietic Stem Cells Without Transgenes. Cell Reprogram 2024; 26:43-45. [PMID: 38530081 DOI: 10.1089/cell.2024.0020] [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: 03/27/2024] Open
Abstract
Creating hematopoietic stem cells (HSCs) capable of multilineage engraft while possessing the ability to self-renew stands as a pivotal achievement within the field of regenerative medicine. However, achieving the generation of these cells without transgene expression or teratoma formation has not been fully accomplished. In a recent publication featured in Cell Stem Cell, Piau et al. document the production of functional HSCs derived from human-induced pluripotent stem cells (hiPSCs). They achieved this through a one-step differentiation protocol that notably does not require any transgene expression. hiPSCs-derived HSCs can engraft and self-renew upon serial transplantation and they are able to reconstitute lymphoid, myeloid, and erythroid compartments. This study presents a promising system to further study human HSC ontogeny, and it might represent a crucial step to obtain HSCs.
Collapse
Affiliation(s)
- Luis G Palma
- Research Program in Cancer, Hospital del Mar Research Institute, Barcelona, Spain
- Research Program in Lymphoid Malignancies, Research Institute Against Leukemia Josep Carreras, Barcelona, Spain
- CIBERONC, Barcelona, Spain
| | - Anna Bigas
- Research Program in Cancer, Hospital del Mar Research Institute, Barcelona, Spain
- Research Program in Lymphoid Malignancies, Research Institute Against Leukemia Josep Carreras, Barcelona, Spain
- CIBERONC, Barcelona, Spain
| |
Collapse
|
6
|
Welsh AM, Muljo SA. Post-transcriptional (re)programming of B lymphocyte development: From bench to bedside? Adv Immunol 2024; 161:85-108. [PMID: 38763703 DOI: 10.1016/bs.ai.2024.03.003] [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: 05/21/2024]
Abstract
Hematopoiesis, a process which generates blood and immune cells, changes significantly during mammalian development. Definitive hematopoiesis is marked by the emergence of long-term hematopoietic stem cells (HSCs). Here, we will focus on the post-transcriptional differences between fetal liver (FL) and adult bone marrow (ABM) HSCs. It remains unclear how or why exactly FL HSCs transition to ABM HSCs, but we aim to leverage their differences to revive an old idea: in utero HSC transplantation. Unexpectedly, the expression of certain RNA-binding proteins (RBPs) play an important role in HSC specification, and can be employed to convert or reprogram adult HSCs back to a fetal-like state. Among other features, FL HSCs have a broad differentiation capacity that includes the ability to regenerate both conventional B and T cells, as well as innate-like or unconventional lymphocytes such as B-1a and marginal zone B (MzB) cells. This chapter will focus on RNA binding proteins, namely LIN28B and IGF2BP3, that are expressed during fetal life and how they promote B-1a cell development. Furthermore, this chapter considers a potential clinical application of synthetic co-expression of LIN28B and IGF2BP3 in HSCs.
Collapse
Affiliation(s)
- Alia M Welsh
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stefan A Muljo
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States.
| |
Collapse
|
7
|
Thambyrajah R, Maqueda M, Neo WH, Imbach K, Guillén Y, Grases D, Fadlullah Z, Gambera S, Matteini F, Wang X, Calero-Nieto FJ, Esteller M, Florian MC, Porta E, Benedito R, Göttgens B, Lacaud G, Espinosa L, Bigas A. Cis inhibition of NOTCH1 through JAGGED1 sustains embryonic hematopoietic stem cell fate. Nat Commun 2024; 15:1604. [PMID: 38383534 PMCID: PMC10882055 DOI: 10.1038/s41467-024-45716-y] [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: 06/07/2023] [Accepted: 02/02/2024] [Indexed: 02/23/2024] Open
Abstract
Hematopoietic stem cells (HSCs) develop from the hemogenic endothelium (HE) in the aorta- gonads-and mesonephros (AGM) region and reside within Intra-aortic hematopoietic clusters (IAHC) along with hematopoietic progenitors (HPC). The signalling mechanisms that distinguish HSCs from HPCs are unknown. Notch signaling is essential for arterial specification, IAHC formation and HSC activity, but current studies on how Notch segregates these different fates are inconsistent. We now demonstrate that Notch activity is highest in a subset of, GFI1 + , HSC-primed HE cells, and is gradually lost with HSC maturation. We uncover that the HSC phenotype is maintained due to increasing levels of NOTCH1 and JAG1 interactions on the surface of the same cell (cis) that renders the NOTCH1 receptor from being activated. Forced activation of the NOTCH1 receptor in IAHC activates a hematopoietic differentiation program. Our results indicate that NOTCH1-JAG1 cis-inhibition preserves the HSC phenotype in the hematopoietic clusters of the embryonic aorta.
Collapse
Affiliation(s)
- Roshana Thambyrajah
- Program in Cancer Research. Institut Hospital del Mar d'Investigacions Mèdiques, CIBERONC, Barcelona, Spain.
- Josep Carreras Leukemia Research Institute, Barcelona, Spain.
- Centro de Investigacion Biomedica en Red (CIBER), Madrid, Spain.
| | - Maria Maqueda
- Program in Cancer Research. Institut Hospital del Mar d'Investigacions Mèdiques, CIBERONC, Barcelona, Spain
| | - Wen Hao Neo
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Kathleen Imbach
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Yolanda Guillén
- Program in Cancer Research. Institut Hospital del Mar d'Investigacions Mèdiques, CIBERONC, Barcelona, Spain
| | - Daniela Grases
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Zaki Fadlullah
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Stefano Gambera
- Molecular Genetics of Angiogenesis Group. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Francesca Matteini
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), Barcelona, Spain
| | - Xiaonan Wang
- Department of Haematology, Wellcome - MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
- School of Public Health, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fernando J Calero-Nieto
- Department of Haematology, Wellcome - MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Manel Esteller
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Centro de Investigacion Biomedica en Red (CIBER), Madrid, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain
- Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Maria Carolina Florian
- Centro de Investigacion Biomedica en Red (CIBER), Madrid, Spain
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), Barcelona, Spain
| | - Eduard Porta
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Berthold Göttgens
- Department of Haematology, Wellcome - MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Lluis Espinosa
- Program in Cancer Research. Institut Hospital del Mar d'Investigacions Mèdiques, CIBERONC, Barcelona, Spain
- Centro de Investigacion Biomedica en Red (CIBER), Madrid, Spain
| | - Anna Bigas
- Program in Cancer Research. Institut Hospital del Mar d'Investigacions Mèdiques, CIBERONC, Barcelona, Spain.
- Josep Carreras Leukemia Research Institute, Barcelona, Spain.
- Centro de Investigacion Biomedica en Red (CIBER), Madrid, Spain.
| |
Collapse
|
8
|
Abe T, Sarentonglaga B, Nagao Y. Advancements in medical research using fetal sheep: Implications for human health and treatment methods. Anim Sci J 2024; 95:e13945. [PMID: 38651196 DOI: 10.1111/asj.13945] [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: 01/24/2024] [Revised: 03/13/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Sheep are typically considered as industrial animals that provide wool and meals. However, they play a significant role in medical research in addition to their conventional use. Notably, sheep fetuses are resistant to surgical invasions and can endure numerous manipulations, such as needle puncture and cell transplantation, and surgical operations requiring exposure beyond the uterus. Based on these distinguishing characteristics, we established a chimeric sheep model capable of producing human/monkey pluripotent cell-derived blood cells via the fetal liver. Furthermore, sheep have become crucial as human fetal models, acting as platforms for developing and improving techniques for intrauterine surgery to address congenital disorders and clarifying the complex pharmacokinetic interactions between mothers and their fetuses. This study emphasizes the significant contributions of fetal sheep to advancing human disease understanding and treatment strategies, highlighting their unique characteristics that are not present in other animals.
Collapse
Affiliation(s)
- Tomoyuki Abe
- Open Science Laboratory, Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi, Japan
| | | | - Yoshikazu Nagao
- Department of Agriculture, Utsunomiya University, Tochigi, Japan
| |
Collapse
|
9
|
Zhang Y, Liu F. The evolving views of hematopoiesis: from embryo to adulthood and from in vivo to in vitro. J Genet Genomics 2024; 51:3-15. [PMID: 37734711 DOI: 10.1016/j.jgg.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
The hematopoietic system composed of hematopoietic stem and progenitor cells (HSPCs) and their differentiated lineages serves as an ideal model to uncover generic principles of cell fate transitions. From gastrulation onwards, there successively emerge primitive hematopoiesis (that produces specialized hematopoietic cells), pro-definitive hematopoiesis (that produces lineage-restricted progenitor cells), and definitive hematopoiesis (that produces multipotent HSPCs). These nascent lineages develop in several transient hematopoietic sites and finally colonize into lifelong hematopoietic sites. The development and maintenance of hematopoietic lineages are orchestrated by cell-intrinsic gene regulatory networks and cell-extrinsic microenvironmental cues. Owing to the progressive methodology (e.g., high-throughput lineage tracing and single-cell functional and omics analyses), our understanding of the developmental origin of hematopoietic lineages and functional properties of certain hematopoietic organs has been updated; meanwhile, new paradigms to characterize rare cell types, cell heterogeneity and its causes, and comprehensive regulatory landscapes have been provided. Here, we review the evolving views of HSPC biology during developmental and postnatal hematopoiesis. Moreover, we discuss recent advances in the in vitro induction and expansion of HSPCs, with a focus on the implications for clinical applications.
Collapse
Affiliation(s)
- Yifan Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Feng Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China; State Key Laboratory of Membrane Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
10
|
Marcoux P, Imeri J, Desterke C, Latsis T, Chaker D, Hugues P, Griscelli AB, Turhan AG. Impact of the overexpression of the tyrosine kinase receptor RET in the hematopoietic potential of induced pluripotent stem cells (iPSCs). Cytotherapy 2024; 26:63-72. [PMID: 37921725 DOI: 10.1016/j.jcyt.2023.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/12/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Previous studies have suggested that the tyrosine kinase receptor RET plays a significant role in the hematopoietic potential in mice and could also be used to expand cord-blood derived hematopoietic stem cells (HSCs). The role of RET in human iPSC-derived hematopoiesis has not been tested so far. METHODS To test the implication of RET on the hematopoietic potential of iPSCs, we activated its pathway with the lentiviral overexpression of RETWT or RETC634Y mutation in normal iPSCs. An iPSC derived from a patient harboring the RETC634Y mutation (iRETC634Y) and its CRISPR-corrected isogenic control iPSC (iRETCTRL) were also used. The hematopoietic potential was tested using 2D cultures and evaluated regarding the phenotype and the clonogenic potential of generated cells. RESULTS Hematopoietic differentiation from iPSCs with RET overexpression (WT or C634Y) led to a significant reduction in the number and in the clonogenic potential of primitive hematopoietic cells (CD34+/CD38-/CD49f+) as compared to control iPSCs. Similarly, the hematopoietic potential of iRETC634Y was reduced as compared to iRETCTRL. Transcriptomic analyses revealed a specific activated expression profile for iRETC634Y compared to its control with evidence of overexpression of genes which are part of the MAPK network with negative hematopoietic regulator activities. CONCLUSION RET activation in iPSCs is associated with an inhibitory activity in iPSC-derived hematopoiesis, potentially related to MAPK activation.
Collapse
Affiliation(s)
- Paul Marcoux
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | - Jusuf Imeri
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | - Christophe Desterke
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | | | - Diana Chaker
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; CITHERA, Centre for iPSC Therapies, INSERM UMS-45, Genopole Campus, Evry, France
| | - Patricia Hugues
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France
| | - Annelise Bennaceur Griscelli
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France; Department of Hematology, APHP Paris Saclay, Hôpital Bicetre, Le Kremlin Bicetre France; CITHERA, Centre for iPSC Therapies, INSERM UMS-45, Genopole Campus, Evry, France; Department of Hematology, APHP Paris Saclay, Hôpital Paul Brousse, Villejuif, France
| | - Ali G Turhan
- INSERM UMR-S-1310, Université Paris Saclay, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicetre France; Department of Hematology, APHP Paris Saclay, Hôpital Bicetre, Le Kremlin Bicetre France; CITHERA, Centre for iPSC Therapies, INSERM UMS-45, Genopole Campus, Evry, France; Department of Hematology, APHP Paris Saclay, Hôpital Paul Brousse, Villejuif, France.
| |
Collapse
|
11
|
Dijkhuis L, Johns A, Ragusa D, van den Brink SC, Pina C. Haematopoietic development and HSC formation in vitro: promise and limitations of gastruloid models. Emerg Top Life Sci 2023; 7:439-454. [PMID: 38095554 PMCID: PMC10754337 DOI: 10.1042/etls20230091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
Haematopoietic stem cells (HSCs) are the most extensively studied adult stem cells. Yet, six decades after their first description, reproducible and translatable generation of HSC in vitro remains an unmet challenge. HSC production in vitro is confounded by the multi-stage nature of blood production during development. Specification of HSC is a late event in embryonic blood production and depends on physical and chemical cues which remain incompletely characterised. The precise molecular composition of the HSC themselves is incompletely understood, limiting approaches to track their origin in situ in the appropriate cellular, chemical and mechanical context. Embryonic material at the point of HSC emergence is limiting, highlighting the need for an in vitro model of embryonic haematopoietic development in which current knowledge gaps can be addressed and exploited to enable HSC production. Gastruloids are pluripotent stem cell-derived 3-dimensional (3D) cellular aggregates which recapitulate developmental events in gastrulation and early organogenesis with spatial and temporal precision. Gastruloids self-organise multi-tissue structures upon minimal and controlled external cues, and are amenable to live imaging, screening, scaling and physicochemical manipulation to understand and translate tissue formation. In this review, we consider the haematopoietic potential of gastruloids and review early strategies to enhance blood progenitor and HSC production. We highlight possible strategies to achieve HSC production from gastruloids, and discuss the potential of gastruloid systems in illuminating current knowledge gaps in HSC specification.
Collapse
Affiliation(s)
- Liza Dijkhuis
- Department of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | - Ayona Johns
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| | - Denise Ragusa
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| | | | - Cristina Pina
- College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge UB8 3PH, U.K
- Centre for Genome Engineering and Maintenance, Brunel University London, Uxbridge UB8 3PH, U.K
| |
Collapse
|
12
|
Piau O, Brunet-Manquat M, L'Homme B, Petit L, Birebent B, Linard C, Moeckes L, Zuliani T, Lapillonne H, Benderitter M, Douay L, Chapel A, Guyonneau-Harmand L, Jaffredo T. Generation of transgene-free hematopoietic stem cells from human induced pluripotent stem cells. Cell Stem Cell 2023; 30:1610-1623.e7. [PMID: 38065068 DOI: 10.1016/j.stem.2023.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 09/25/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023]
Abstract
Hematopoietic stem cells (HSCs) are the rare cells responsible for the lifelong curative effects of hematopoietic cell (HC) transplantation. The demand for clinical-grade HSCs has increased significantly in recent decades, leading to major difficulties in treating patients. A promising but not yet achieved goal is the generation of HSCs from pluripotent stem cells. Here, we have obtained vector- and stroma-free transplantable HSCs by differentiating human induced pluripotent stem cells (hiPSCs) using an original one-step culture system. After injection into immunocompromised mice, cells derived from hiPSCs settle in the bone marrow and form a robust multilineage hematopoietic population that can be serially transplanted. Single-cell RNA sequencing shows that this repopulating activity is due to a hematopoietic population that is transcriptionally similar to human embryonic aorta-derived HSCs. Overall, our results demonstrate the generation of HSCs from hiPSCs and will help identify key regulators of HSC production during human ontogeny.
Collapse
Affiliation(s)
- Olivier Piau
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, CRSA, 75012 Paris, France; Sorbonne Université, CNRS UMR7622, Inserm U1156, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 9 Quai St-Bernard, 75005 Paris, France
| | - Mathias Brunet-Manquat
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, CRSA, 75012 Paris, France; EFS Ile de France, Unité d'Ingénierie et de Thérapie Cellulaire, 94017 Créteil, France
| | - Bruno L'Homme
- Laboratoire de radiobiologie des expositions médicales (LRMed), Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92262 Fontenay-aux-Roses, France
| | - Laurence Petit
- Sorbonne Université, CNRS UMR7622, Inserm U1156, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, 9 Quai St-Bernard, 75005 Paris, France
| | - Brigitte Birebent
- EFS Ile de France, Unité d'Ingénierie et de Thérapie Cellulaire, 94017 Créteil, France
| | - Christine Linard
- Laboratoire de radiobiologie des expositions médicales (LRMed), Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92262 Fontenay-aux-Roses, France
| | - Laetitia Moeckes
- Etablissement Français du Sang - Atlantic Bio GMP - 2, rue Aronnax, 44800 Saint-Herblain, France
| | - Thomas Zuliani
- Etablissement Français du Sang - Atlantic Bio GMP - 2, rue Aronnax, 44800 Saint-Herblain, France
| | - Hélène Lapillonne
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, CRSA, 75012 Paris, France; AP-HP, Hôpital St Antoine/Trousseau, Service d'Hématologie Biologique, 75012 Paris, France
| | - Marc Benderitter
- Laboratoire de radiobiologie des expositions médicales (LRMed), Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92262 Fontenay-aux-Roses, France
| | - Luc Douay
- AP-HP, Hôpital St Antoine/Trousseau, Service d'Hématologie Biologique, 75012 Paris, France
| | - Alain Chapel
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, CRSA, 75012 Paris, France; Laboratoire de radiobiologie des expositions médicales (LRMed), Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92262 Fontenay-aux-Roses, France
| | - Laurence Guyonneau-Harmand
- Sorbonne Université, INSERM UMR_S938, Centre de Recherche Saint Antoine, CRSA, 75012 Paris, France; EFS Ile de France, Unité d'Ingénierie et de Thérapie Cellulaire, 94017 Créteil, France.
| | - Thierry Jaffredo
- EFS Ile de France, Unité d'Ingénierie et de Thérapie Cellulaire, 94017 Créteil, France.
| |
Collapse
|
13
|
Zheng H, Chen Y, Luo Q, Zhang J, Huang M, Xu Y, Huo D, Shan W, Tie R, Zhang M, Qian P, Huang H. Generating hematopoietic cells from human pluripotent stem cells: approaches, progress and challenges. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:31. [PMID: 37656237 PMCID: PMC10474004 DOI: 10.1186/s13619-023-00175-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Human pluripotent stem cells (hPSCs) have been suggested as a potential source for the production of blood cells for clinical application. In two decades, almost all types of blood cells can be successfully generated from hPSCs through various differentiated strategies. Meanwhile, with a deeper understanding of hematopoiesis, higher efficiency of generating progenitors and precursors of blood cells from hPSCs is achieved. However, how to generate large-scale mature functional cells from hPSCs for clinical use is still difficult. In this review, we summarized recent approaches that generated both hematopoietic stem cells and mature lineage cells from hPSCs, and remarked their efficiency and mechanisms in producing mature functional cells. We also discussed the major challenges in hPSC-derived products of blood cells and provided some potential solutions. Our review summarized efficient, simple, and defined methodologies for developing good manufacturing practice standards for hPSC-derived blood cells, which will facilitate the translation of these products into the clinic.
Collapse
Affiliation(s)
- Haiqiong Zheng
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Yijin Chen
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Qian Luo
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Jie Zhang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Mengmeng Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Yulin Xu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Dawei Huo
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Wei Shan
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China
| | - Meng Zhang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China.
| | - Pengxu Qian
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China.
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310012, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310012, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310012, China.
| |
Collapse
|
14
|
Martin-Iglesias S, Herrera L, Santos S, Vesga MÁ, Eguizabal C, Lanceros-Mendez S, Silvan U. Analysis of the impact of handling and culture on the expansion and functionality of NK cells. Front Immunol 2023; 14:1225549. [PMID: 37638054 PMCID: PMC10451065 DOI: 10.3389/fimmu.2023.1225549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/13/2023] [Indexed: 08/29/2023] Open
Abstract
Natural killer (NK) cells are lymphocytes of the innate immune system that play a key role in the elimination of tumor and virus-infected cells. Unlike T cells, NK cell activation is governed by their direct interaction with target cells via the inhibitory and activating receptors present on their cytoplasmic membrane. The simplicity of this activation mechanism has allowed the development of immunotherapies based on the transduction of NK cells with CAR (chimeric antigen receptor) constructs for the treatment of cancer. Despite the advantages of CAR-NK therapy over CAR-T, including their inability to cause graft-versus-host disease in allogenic therapies, a deeper understanding of the impact of their handling is needed in order to increase their functionality and applicability. With that in mind, the present work critically examines the steps required for NK cell isolation, expansion and storage, and analyze the response of the NK cells to these manipulations. The results show that magnetic-assisted cell sorting, traditionally used for NK isolation, increases the CD16+ population of NK cultures only if the protocol includes both, antibody incubation and passage through the isolation column. Furthermore, based on the importance of surface potential on cellular responses, the influence of surfaces with different net surface charge on NK cells has been evaluated, showing that NK cells displayed higher proliferation rates on charged surfaces than on non-charged ones. The present work highlights the relevance of NK cells manipulation for improving the applicability and effectiveness of NK cell-based therapies.
Collapse
Affiliation(s)
- Sara Martin-Iglesias
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Spain
| | - Lara Herrera
- Cell Therapy, Stem Cells and Tissues Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Research Unit, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain
| | - Silvia Santos
- Cell Therapy, Stem Cells and Tissues Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Research Unit, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Miguel Ángel Vesga
- Cell Therapy, Stem Cells and Tissues Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Research Unit, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Cristina Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Research Unit, Basque Centre for Blood Transfusion and Human Tissues, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Red de Inmunoterapia del Cáncer “REINCA” (RED2022-134831-T), Madrid, Spain
| | - Senentxu Lanceros-Mendez
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Basque Foundation for Science, Ikerbasque, Bilbao, Spain
| | - Unai Silvan
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Basque Foundation for Science, Ikerbasque, Bilbao, Spain
| |
Collapse
|
15
|
Li Y, Ding J, Araki D, Zou J, Larochelle A. Modulation of WNT, Activin/Nodal, and MAPK Signaling Pathways Increases Arterial Hemogenic Endothelium and Hematopoietic Stem/Progenitor Cell Formation During Human iPSC Differentiation. Stem Cells 2023; 41:685-697. [PMID: 37220178 PMCID: PMC10346406 DOI: 10.1093/stmcls/sxad040] [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: 02/21/2023] [Accepted: 05/03/2023] [Indexed: 05/25/2023]
Abstract
Several differentiation protocols enable the emergence of hematopoietic stem and progenitor cells (HSPCs) from human-induced pluripotent stem cells (iPSCs), yet optimized schemes to promote the development of HSPCs with self-renewal, multilineage differentiation, and engraftment potential are lacking. To improve human iPSC differentiation methods, we modulated WNT, Activin/Nodal, and MAPK signaling pathways by stage-specific addition of small-molecule regulators CHIR99021, SB431542, and LY294002, respectively, and measured the impact on hematoendothelial formation in culture. Manipulation of these pathways provided a synergy sufficient to enhance formation of arterial hemogenic endothelium (HE) relative to control culture conditions. Importantly, this approach significantly increased production of human HSPCs with self-renewal and multilineage differentiation properties, as well as phenotypic and molecular evidence of progressive maturation in culture. Together, these findings provide a stepwise improvement in human iPSC differentiation protocols and offer a framework for manipulating intrinsic cellular cues to enable de novo generation of human HSPCs with functionality in vivo.
Collapse
Affiliation(s)
- Yongqin Li
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jianyi Ding
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Daisuke Araki
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jizhong Zou
- iPSC Core Facility, NHLBI, NIH, Bethesda, MD, USA
| | - Andre Larochelle
- Cellular and Molecular Therapeutics Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
16
|
Establishment of isogenic induced pluripotent stem cells with or without pathogenic mutation for understanding the pathogenesis of myeloproliferative neoplasms. Exp Hematol 2023; 118:12-20. [PMID: 36511286 DOI: 10.1016/j.exphem.2022.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Identification and functional characterization of disease-associated genetic traits are crucial for understanding the pathogenesis of hematologic malignancies. Various in vitro and in vivo models, including cell lines, primary cells, and animal models, have been established to examine these genetic alterations. However, their nonphysiologic conditions, diverse genetic backgrounds, and species-specific differences often limit data interpretation. To evaluate somatic mutations in myeloproliferative neoplasms (MPNs), we used CRISPR/Cas9 combined with the piggyBac transposon system to establish isogenic induced pluripotent stem (iPS) cell lines with or without JAK2V617F mutation, a driver mutation of MPNs. We induced hematopoietic stem/progenitor cells (HSPCs) from these iPS cells and observed phenotypic differences during hematopoiesis using fluorescence-activated cell sorting analysis. HSPCs with pathogenic mutations exhibited cell-autonomous erythropoiesis and megakaryopoiesis, which are hallmarks in the bone marrow of patients with MPNs. Furthermore, we used these HSPCs as a model to validate therapeutic compounds and showed that interferon alpha selectively inhibited erythropoiesis and megakaryopoiesis in mutant HSPCs. These results demonstrate that genome editing is feasible for establishing isogenic iPS cells, studying genetic elements to understand the pathogenesis of MPNs, and evaluating therapeutic compounds against MPNs.
Collapse
|
17
|
Ding J, Li Y, Larochelle A. De Novo Generation of Human Hematopoietic Stem Cells from Pluripotent Stem Cells for Cellular Therapy. Cells 2023; 12:321. [PMID: 36672255 PMCID: PMC9857267 DOI: 10.3390/cells12020321] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
The ability to manufacture human hematopoietic stem cells (HSCs) in the laboratory holds enormous promise for cellular therapy of human blood diseases. Several differentiation protocols have been developed to facilitate the emergence of HSCs from human pluripotent stem cells (PSCs). Most approaches employ a stepwise addition of cytokines and morphogens to recapitulate the natural developmental process. However, these protocols globally lack clinical relevance and uniformly induce PSCs to produce hematopoietic progenitors with embryonic features and limited engraftment and differentiation capabilities. This review examines how key intrinsic cues and extrinsic environmental inputs have been integrated within human PSC differentiation protocols to enhance the emergence of definitive hematopoiesis and how advances in genomics set the stage for imminent breakthroughs in this field.
Collapse
Affiliation(s)
| | | | - Andre Larochelle
- Cellular and Molecular Therapeutics Branch, National Heart Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Bigas A, Galán Palma L, Kartha GM, Giorgetti A. Using Pluripotent Stem Cells to Understand Normal and Leukemic Hematopoietic Development. Stem Cells Transl Med 2022; 11:1123-1134. [PMID: 36398586 PMCID: PMC9672852 DOI: 10.1093/stcltm/szac071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2023] Open
Abstract
Several decades have passed since the generation of the first embryonic stem cell (ESC) lines both in mice and in humans. Since then, stem cell biologists have tried to understand their potential biological and clinical uses for their implementation in regenerative medicine. The hematopoietic field was a pioneer in establishing the potential use for the development of blood cell products and clinical applications; however, early expectations have been truncated by the difficulty in generating bonafide hematopoietic stem cells (HSCs). Despite some progress in understanding the origin of HSCs during embryonic development, the reproduction of this process in vitro is still not possible, but the knowledge acquired in the embryo is slowly being implemented for mouse and human pluripotent stem cells (PSCs). In contrast, ESC-derived hematopoietic cells may recapitulate some leukemic transformation processes when exposed to oncogenic drivers. This would be especially useful to model prenatal leukemia development or other leukemia-predisposing syndromes, which are difficult to study. In this review, we will review the state of the art of the use of PSCs as a model for hematopoietic and leukemia development.
Collapse
Affiliation(s)
- Anna Bigas
- Program in Cancer Research, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), CIBERONC, Barcelona, Spain
- Josep Carreras Leukemia Research Institute (IJC), Barcelona, Spain
| | - Luis Galán Palma
- Program in Cancer Research, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), CIBERONC, Barcelona, Spain
- Josep Carreras Leukemia Research Institute (IJC), Barcelona, Spain
| | - Gayathri M Kartha
- Program in Cancer Research, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), CIBERONC, Barcelona, Spain
- Josep Carreras Leukemia Research Institute (IJC), Barcelona, Spain
| | - Alessandra Giorgetti
- Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Barcelona University, Barcelona, Spain
| |
Collapse
|
19
|
Abstract
Hematopoietic stem cell (HSC) regeneration is the remarkable process by which extremely rare, normally inactive cells of the bone marrow can replace an entire organ if called to do so by injury or harnessed by transplantation. HSC research is arguably the first quantitative single-cell science and the foundation of adult stem cell biology. Bone marrow transplant is the oldest and most refined technique of regenerative medicine. Here we review the intertwined history of the discovery of HSCs and bone marrow transplant, the molecular and cellular mechanisms of HSC self-renewal, and the use of HSCs and their derivatives for cell therapy.
Collapse
Affiliation(s)
- Mitch Biermann
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Tannishtha Reya
- Department of Medicine, University of California San Diego, La Jolla, California 92093
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093
| |
Collapse
|
20
|
Challenges in Cell Fate Acquisition to Scid-Repopulating Activity from Hemogenic Endothelium of hiPSCs Derived from AML Patients Using Forced Transcription Factor Expression. Cells 2022; 11:cells11121915. [PMID: 35741044 PMCID: PMC9221973 DOI: 10.3390/cells11121915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 12/10/2022] Open
Abstract
The generation of human hematopoietic stem cells (HSCs) from human pluripotent stem cells (hPSCs) represents a major goal in regenerative medicine and is believed would follow principles of early development. HSCs arise from a type of endothelial cell called a “hemogenic endothelium” (HE), and human HSCs are experimentally detected by transplantation into SCID or other immune-deficient mouse recipients, termed SCID-Repopulating Cells (SRC). Recently, SRCs were detected by forced expression of seven transcription factors (TF) (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1, and SPI1) in hPSC-derived HE, suggesting these factors are deficient in hPSC differentiation to HEs required to generate HSCs. Here we derived PECAM-1-, Flk-1-, and VE-cadherin-positive endothelial cells that also lack CD45 expression (PFVCD45−) which are solely responsible for hematopoietic output from iPSC lines reprogrammed from AML patients. Using HEs derived from AML patient iPSCs devoid of somatic leukemic aberrations, we sought to generate putative SRCs by the forced expression of 7TFs to model autologous HSC transplantation. The expression of 7TFs in hPSC-derived HE cells from an enhanced hematopoietic progenitor capacity was present in vitro, but failed to acquire SRC activity in vivo. Our findings emphasize the benefits of forced TF expression, along with the continued challenges in developing HSCs for autologous-based therapies from hPSC sources.
Collapse
|
21
|
Guo R, Li W, Li Y, Li Y, Jiang Z, Song Y. Generation and clinical potential of functional T lymphocytes from gene-edited pluripotent stem cells. Exp Hematol Oncol 2022; 11:27. [PMID: 35568954 PMCID: PMC9107657 DOI: 10.1186/s40164-022-00285-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 04/26/2022] [Indexed: 12/16/2022] Open
Abstract
Engineered T cells have been shown to be highly effective in cancer immunotherapy, although T cell exhaustion presents a challenge for their long-term function. Additional T-cell sources must be exploited to broaden the application of engineered T cells for immune defense and reconstitution. Unlimited sources of pluripotent stem cells (PSCs) have provided a potential opportunity to generate precise-engineered therapeutic induced T (iT) cells. Single-cell transcriptome analysis of PSC-derived induced hematopoietic stem and progenitor cells (iHSPC)/iT identified the developmental pathways and possibilities of generating functional T cell from PSCs. To date, the PSC-to-iT platforms encounter several problems, including low efficiency of conventional T subset specification, limited functional potential, and restrictions on large-scale application, because of the absence of a thymus-like organized microenvironment. The updated PSC-to-iT platforms, such as the three-dimensional (3D) artificial thymic organoid (ATO) co-culture system and Runx1/Hoxa9-enforced iT lymphopoiesis, provide fresh perspectives for coordinating culture conditions and transcription factors, which may greatly improve the efficiency of T-cell generation greatly. In addition, the improved PSC-to-iT platform coordinating gene editing technologies will provide various functional engineered unconventional or conventional T cells. Furthermore, the clinical applications of PSC-derived immune cells are accelerating from bench to bedside.
Collapse
Affiliation(s)
- Rongqun Guo
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yadan Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.,Academy of Medical Science, Henan Medical College of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yingmei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
22
|
Wen B, Wang G, Li E, Kolesnichenko OA, Tu Z, Divanovic S, Kalin TV, Kalinichenko VV. In vivo generation of bone marrow from embryonic stem cells in interspecies chimeras. eLife 2022; 11:74018. [PMID: 36178184 PMCID: PMC9578712 DOI: 10.7554/elife.74018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 09/29/2022] [Indexed: 01/07/2023] Open
Abstract
Generation of bone marrow (BM) from embryonic stem cells (ESCs) promises to accelerate the development of future cell therapies for life-threatening disorders. However, such approach is limited by technical challenges to produce a mixture of functional BM progenitor cells able to replace all hematopoietic cell lineages. Herein, we used blastocyst complementation to simultaneously produce BM cell lineages from mouse ESCs in a rat. Based on fluorescence-activated cell sorting analysis and single-cell RNA sequencing, mouse ESCs differentiated into multiple hematopoietic and stromal cell types that were indistinguishable from normal mouse BM cells based on gene expression signatures and cell surface markers. Receptor-ligand interactions identified Cxcl12-Cxcr4, Lama2-Itga6, App-Itga6, Comp-Cd47, Col1a1-Cd44, and App-Il18rap as major signaling pathways between hematopoietic progenitors and stromal cells. Multiple hematopoietic progenitors, including hematopoietic stem cells (HSCs) in mouse-rat chimeras derived more efficiently from mouse ESCs, whereas chondrocytes predominantly derived from rat cells. In the dorsal aorta and fetal liver of mouse-rat chimeras, mouse HSCs emerged and expanded faster compared to endogenous rat cells. Sequential BM transplantation of ESC-derived cells from mouse-rat chimeras rescued lethally irradiated syngeneic mice and demonstrated long-term reconstitution potential of donor HSCs. Altogether, a fully functional BM was generated from mouse ESCs using rat embryos as 'bioreactors'.
Collapse
Affiliation(s)
- Bingqiang Wen
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Guolun Wang
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Enhong Li
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Olena A Kolesnichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Zhaowei Tu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States
| | - Senad Divanovic
- Division of Immunobiology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine of the University of CincinnatiCincinnatiUnited States
| | - Tanya V Kalin
- Department of Pediatrics, College of Medicine of the University of CincinnatiCincinnatiUnited States,Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine of the University of CincinnatiCincinnatiUnited States,Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States,Division of Developmental Biology, Cincinnati Children’s Hospital Medical CenterCincinnatiUnited States
| |
Collapse
|
23
|
Induced Pluripotent Stem Cells as a Tool for Modeling Hematologic Disorders and as a Potential Source for Cell-Based Therapies. Cells 2021; 10:cells10113250. [PMID: 34831472 PMCID: PMC8623953 DOI: 10.3390/cells10113250] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022] Open
Abstract
The breakthrough in human induced pluripotent stem cells (hiPSCs) has revolutionized the field of biomedical and pharmaceutical research and opened up vast opportunities for drug discovery and regenerative medicine, especially when combined with gene-editing technology. Numerous healthy and patient-derived hiPSCs for human disease modeling have been established, enabling mechanistic studies of pathogenesis, platforms for preclinical drug screening, and the development of novel therapeutic targets/approaches. Additionally, hiPSCs hold great promise for cell-based therapy, serving as an attractive cell source for generating stem/progenitor cells or functional differentiated cells for degenerative diseases, due to their unlimited proliferative capacity, pluripotency, and ethical acceptability. In this review, we provide an overview of hiPSCs and their utility in the study of hematologic disorders through hematopoietic differentiation. We highlight recent hereditary and acquired genetic hematologic disease modeling with patient-specific iPSCs, and discuss their applications as instrumental drug screening tools. The clinical applications of hiPSCs in cell-based therapy, including the next-generation cancer immunotherapy, are provided. Lastly, we discuss the current challenges that need to be addressed to fulfill the validity of hiPSC-based disease modeling and future perspectives of hiPSCs in the field of hematology.
Collapse
|
24
|
The Race of CAR Therapies: CAR-NK Cells for Fighting B-Cell Hematological Cancers. Cancers (Basel) 2021; 13:cancers13215418. [PMID: 34771581 PMCID: PMC8582420 DOI: 10.3390/cancers13215418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Over the last few years, CAR-T cells have arisen as one of the most promising immunotherapies against relapsed or refractory hematological cancers. Despite their good results in clinical trials, there are some limitations to overcome, such as undesirable side-effects or the restraints of an autologous treatment. Therefore, CAR-NK cells have emerged as a good alternative for these kinds of treatments. This review discusses the advantages of CAR-NK cells compared to CAR-T cells, as well as the different sources and strategies in order to obtain these CAR-NK cells. Abstract Acute lymphoblastic leukemia (ALL) and Chronic lymphocytic leukemia (CLL) are the most common leukemias in children and elderly people, respectively. Standard therapies, such as chemotherapy, are only effective in 40% of ALL adult patients with a five-year survival rate and therefore new alternatives need to be used, such as immunotherapy targeting specific receptors of malignant cells. Among all the options, CAR (Chimeric antigen receptor)-based therapy has arisen as a new opportunity for refractory or relapsed hematological cancer patients. CARs were designed to be used along with T lymphocytes, creating CAR-T cells, but they are presenting such encouraging results that they are already in use as drugs. Nonetheless, their side-effects and the fact that it is not possible to infuse an allogenic CAR-T product without causing graft-versus-host-disease, have meant using a different cell source to solve these problems, such as Natural Killer (NK) cells. Although CAR-based treatment is a high-speed race led by CAR-T cells, CAR-NK cells are slowly (but surely) consolidating their position; their demonstrated efficacy and the lack of undesirable side-effects is opening a new door for CAR-based treatments. CAR-NKs are now in the field to stay.
Collapse
|
25
|
Xiang Y, Sugimura R. Single-Cell Approaches to Deconvolute the Development of HSCs. Cells 2021; 10:2876. [PMID: 34831099 PMCID: PMC8616492 DOI: 10.3390/cells10112876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/19/2021] [Accepted: 10/23/2021] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells (HSCs) play a core role in blood development. The ability to efficiently produce HSCs from various pluripotent stem cell sources is the Holy Grail in the hematology field. However, in vitro or in vivo HSC production remains low, which may be attributable to the lack of understanding of hematopoiesis. Here, we review the recent progress in this area and introduce advanced technologies, such as single-cell RNA-seq, spatial transcriptomics, and molecular barcoding, which may help to acquire missing information about HSC generation. We finally discuss unresolved questions, the answers to which may be conducive to HSC production, providing a promising path toward HSC-based immunotherapies.
Collapse
Affiliation(s)
| | - Ryohichi Sugimura
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, University of Hong Kong, Hong Kong SAR, China;
| |
Collapse
|
26
|
Induced Pluripotent Stem Cells to Model Juvenile Myelomonocytic Leukemia: New Perspectives for Preclinical Research. Cells 2021; 10:cells10092335. [PMID: 34571984 PMCID: PMC8465353 DOI: 10.3390/cells10092335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 11/16/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a malignant myeloproliferative disorder arising in infants and young children. The origin of this neoplasm is attributed to an early deregulation of the Ras signaling pathway in multipotent hematopoietic stem/progenitor cells. Since JMML is notoriously refractory to conventional cytostatic therapy, allogeneic hematopoietic stem cell transplantation remains the mainstay of curative therapy for most cases. However, alternative therapeutic approaches with small epigenetic molecules have recently entered the stage and show surprising efficacy at least in specific subsets of patients. Hence, the establishment of preclinical models to test novel agents is a priority. Induced pluripotent stem cells (IPSCs) offer an opportunity to imitate JMML ex vivo, after attempts to generate immortalized cell lines from primary JMML material have largely failed in the past. Several research groups have previously generated patient-derived JMML IPSCs and successfully differentiated these into myeloid cells with extensive phenotypic similarities to primary JMML cells. With infinite self-renewal and the capability to differentiate into multiple cell types, JMML IPSCs are a promising resource to advance the development of treatment modalities targeting specific vulnerabilities. This review discusses current reprogramming techniques for JMML stem/progenitor cells, related clinical applications, and the challenges involved.
Collapse
|
27
|
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: 0] [Impact Index Per Article: 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
|
28
|
Fidanza A, Forrester LM. Progress in the production of haematopoietic stem and progenitor cells from human pluripotent stem cells. ACTA ACUST UNITED AC 2021; 13:100050. [PMID: 34405125 PMCID: PMC8350141 DOI: 10.1016/j.regen.2021.100050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022]
Abstract
Cell therapies are currently used to treat many haematological diseases. These treatments range from the long-term reconstitution of the entire haematopoietic system using the most potent haematopoietic stem cells (HSCs) to the short-term rescue with mature functional end cells such as oxygen-carrying red blood cells and cells of the immune system that can fight infection and repair tissue. Limitations in supply and the risk of transmitting infection has prompted the design of protocols to produce some of these cell types from human pluripotent stem cells (hPSCs). Although it has proven challenging to generate the most potent HSCs directly from hPSCs, significant progress has been made in the development of differentiation protocols that can successfully produce haematopoietic progenitor cells and most of the mature cell lineages. We review the key steps used in the production of haematopoietic stem and progenitor cells (HSPCs) from hPSCs and the cell surface markers and reporter strategies that have been used to define specific transitions. Most studies have relied on the use of known markers that define HSPC production in vivo but more recently single cell RNA sequencing has allowed a less biased approach to their characterisation. Transcriptional profiling has identified new markers for naïve and committed hPSC-derived HSPC populations and trajectory analyses has provided novel insights into their lineage potential. Direct comparison of in vitro- and in vivo-derived RNA single cell sequencing datasets has highlights similarities and differences between the two systems and this deeper understanding will be key to the design and the tracking of improved and more efficient differentiation protocols.
Collapse
Affiliation(s)
- Antonella Fidanza
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Lesley M Forrester
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| |
Collapse
|
29
|
Li H, Gao L, Du J, Ma T, Ye Z, Li Z. To Better Generate Organoids, What Can We Learn From Teratomas? Front Cell Dev Biol 2021; 9:700482. [PMID: 34336851 PMCID: PMC8324104 DOI: 10.3389/fcell.2021.700482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/21/2021] [Indexed: 12/12/2022] Open
Abstract
The genomic profile of animal models is not completely matched with the genomic profile of humans, and 2D cultures do not represent the cellular heterogeneity and tissue architecture found in tissues of their origin. Derived from 3D culture systems, organoids establish a crucial bridge between 2D cell cultures and in vivo animal models. Organoids have wide and promising applications in developmental research, disease modeling, drug screening, precision therapy, and regenerative medicine. However, current organoids represent only single or partial components of a tissue, which lack blood vessels, native microenvironment, communication with near tissues, and a continuous dorsal-ventral axis within 3D culture systems. Although efforts have been made to solve these problems, unfortunately, there is no ideal method. Teratoma, which has been frequently studied in pathological conditions, was recently discovered as a new in vivo model for developmental studies. In contrast to organoids, teratomas have vascularized 3D structures and regions of complex tissue-like organization. Studies have demonstrated that teratomas can be used to mimic multilineage human development, enrich specific somatic progenitor/stem cells, and even generate brain organoids. These results provide unique opportunities to promote our understanding of the vascularization and maturation of organoids. In this review, we first summarize the basic characteristics, applications, and limitations of both organoids and teratomas and further discuss the possibility that in vivo teratoma systems can be used to promote the vascularization and maturation of organoids within an in vitro 3D culture system.
Collapse
Affiliation(s)
- Hongyu Li
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lixiong Gao
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jinlin Du
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tianju Ma
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zi Ye
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhaohui Li
- Department of Ophthalmology, The Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
30
|
Gaggi G, Di Credico A, Izzicupo P, Iannetti G, Di Baldassarre A, Ghinassi B. Chemical and Biological Molecules Involved in Differentiation, Maturation, and Survival of Dopaminergic Neurons in Health and Parkinson's Disease: Physiological Aspects and Clinical Implications. Biomedicines 2021; 9:biomedicines9070754. [PMID: 34209807 PMCID: PMC8301385 DOI: 10.3390/biomedicines9070754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is one of the most common neurodegenerative disease characterized by a specific and progressive loss of dopaminergic (DA) neurons and dopamine, causing motor dysfunctions and impaired movements. Unfortunately, available therapies can partially treat the motor symptoms, but they have no effect on non-motor features. In addition, the therapeutic effect reduces gradually, and the prolonged use of drugs leads to a significative increase in the number of adverse events. For these reasons, an alternative approach that allows the replacement or the improved survival of DA neurons is very appealing for the treatment of PD patients and recently the first human clinical trials for DA neurons replacement have been set up. Here, we review the role of chemical and biological molecules that are involved in the development, survival and differentiation of DA neurons. In particular, we review the chemical small molecules used to differentiate different type of stem cells into DA neurons with high efficiency; the role of microRNAs and long non-coding RNAs both in DA neurons development/survival as far as in the pathogenesis of PD; and, finally, we dissect the potential role of exosomes carrying biological molecules as treatment of PD.
Collapse
Affiliation(s)
- Giulia Gaggi
- Beth Israel Deaconess Medical Center, Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA;
| | - Andrea Di Credico
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| | - Pascal Izzicupo
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| | | | - Angela Di Baldassarre
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
- Correspondence:
| | - Barbara Ghinassi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (A.D.C.); (P.I.); (B.G.)
| |
Collapse
|
31
|
Anurogo D, Yuli Prasetyo Budi N, Thi Ngo MH, Huang YH, Pawitan JA. Cell and Gene Therapy for Anemia: Hematopoietic Stem Cells and Gene Editing. Int J Mol Sci 2021; 22:ijms22126275. [PMID: 34200975 PMCID: PMC8230702 DOI: 10.3390/ijms22126275] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/23/2022] Open
Abstract
Hereditary anemia has various manifestations, such as sickle cell disease (SCD), Fanconi anemia, glucose-6-phosphate dehydrogenase deficiency (G6PDD), and thalassemia. The available management strategies for these disorders are still unsatisfactory and do not eliminate the main causes. As genetic aberrations are the main causes of all forms of hereditary anemia, the optimal approach involves repairing the defective gene, possibly through the transplantation of normal hematopoietic stem cells (HSCs) from a normal matching donor or through gene therapy approaches (either in vivo or ex vivo) to correct the patient’s HSCs. To clearly illustrate the importance of cell and gene therapy in hereditary anemia, this paper provides a review of the genetic aberration, epidemiology, clinical features, current management, and cell and gene therapy endeavors related to SCD, thalassemia, Fanconi anemia, and G6PDD. Moreover, we expound the future research direction of HSC derivation from induced pluripotent stem cells (iPSCs), strategies to edit HSCs, gene therapy risk mitigation, and their clinical perspectives. In conclusion, gene-corrected hematopoietic stem cell transplantation has promising outcomes for SCD, Fanconi anemia, and thalassemia, and it may overcome the limitation of the source of allogenic bone marrow transplantation.
Collapse
Affiliation(s)
- Dito Anurogo
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Makassar, Makassar 90221, Indonesia
| | - Nova Yuli Prasetyo Budi
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Mai-Huong Thi Ngo
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yen-Hua Huang
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (D.A.); (N.Y.P.B.); (M.-H.T.N.)
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Comprehensive Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (Y.-H.H.); (J.A.P.); Tel.: +886-2-2736-1661 (ext. 3150) (Y.-H.H.); +62-812-9535-0097 (J.A.P.)
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Stem Cell Medical Technology Integrated Service Unit, Cipto Mangunkusumo Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Stem Cell and Tissue Engineering Research Center, Indonesia Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Correspondence: (Y.-H.H.); (J.A.P.); Tel.: +886-2-2736-1661 (ext. 3150) (Y.-H.H.); +62-812-9535-0097 (J.A.P.)
| |
Collapse
|
32
|
Running the full human developmental clock in interspecies chimeras using alternative human stem cells with expanded embryonic potential. NPJ Regen Med 2021; 6:25. [PMID: 34001907 PMCID: PMC8128894 DOI: 10.1038/s41536-021-00135-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 04/20/2021] [Indexed: 02/08/2023] Open
Abstract
Human pluripotent stem cells (hPSCs) can generate specialized cell lineages that have great potential for regenerative therapies and disease modeling. However, the developmental stage of the lineages generated from conventional hPSC cultures in vitro are embryonic in phenotype, and may not possess the cellular maturity necessary for corrective regenerative function in vivo in adult recipients. Here, we present the scientific evidence for how adult human tissues could generate human–animal interspecific chimeras to solve this problem. First, we review the phenotypes of the embryonic lineages differentiated from conventional hPSC in vitro and through organoid technologies and compare their functional relevance to the tissues generated during normal human in utero fetal and adult development. We hypothesize that the developmental incongruence of embryo-stage hPSC-differentiated cells transplanted into a recipient adult host niche is an important mechanism ultimately limiting their utility in cell therapies and adult disease modeling. We propose that this developmental obstacle can be overcome with optimized interspecies chimeras that permit the generation of adult-staged, patient-specific whole organs within animal hosts with human-compatible gestational time-frames. We suggest that achieving this goal may ultimately have to await the derivation of alternative, primitive totipotent-like stem cells with improved embryonic chimera capacities. We review the scientific challenges of deriving alternative human stem cell states with expanded embryonic potential, outline a path forward for conducting this emerging research with appropriate ethical and regulatory oversight, and defend the case of why current federal funding restrictions on this important category of biomedical research should be liberalized.
Collapse
|
33
|
Lange L, Morgan M, Schambach A. The hemogenic endothelium: a critical source for the generation of PSC-derived hematopoietic stem and progenitor cells. Cell Mol Life Sci 2021; 78:4143-4160. [PMID: 33559689 PMCID: PMC8164610 DOI: 10.1007/s00018-021-03777-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/16/2020] [Accepted: 01/15/2021] [Indexed: 12/02/2022]
Abstract
In vitro generation of hematopoietic cells and especially hematopoietic stem cells (HSCs) from human pluripotent stem cells (PSCs) are subject to intensive research in recent decades, as these cells hold great potential for regenerative medicine and autologous cell replacement therapies. Despite many attempts, in vitro, de novo generation of bona fide HSCs remains challenging, and we are still far away from their clinical use, due to insufficient functionality and quantity of the produced HSCs. The challenges of generating PSC-derived HSCs are already apparent in early stages of hemato-endothelial specification with the limitation of recapitulating complex, dynamic processes of embryonic hematopoietic ontogeny in vitro. Further, these current shortcomings imply the incompleteness of our understanding of human ontogenetic processes from embryonic mesoderm over an intermediate, specialized hemogenic endothelium (HE) to their immediate progeny, the HSCs. In this review, we examine the recent investigations of hemato-endothelial ontogeny and recently reported progress for the conversion of PSCs and other promising somatic cell types towards HSCs with the focus on the crucial and inevitable role of the HE to achieve the long-standing goal—to generate therapeutically applicable PSC-derived HSCs in vitro.
Collapse
Affiliation(s)
- Lucas Lange
- Institute of Experimental Hematology, Hannover Medical School, 30625, Hannover, Germany.,REBIRTH, Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625, Hannover, Germany
| | - Michael Morgan
- Institute of Experimental Hematology, Hannover Medical School, 30625, Hannover, Germany.,REBIRTH, Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625, Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625, Hannover, Germany. .,REBIRTH, Research Center for Translational Regenerative Medicine, Hannover Medical School, 30625, Hannover, Germany. .,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
34
|
Generation and manipulation of human iPSC-derived platelets. Cell Mol Life Sci 2021; 78:3385-3401. [PMID: 33439272 PMCID: PMC7804213 DOI: 10.1007/s00018-020-03749-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/01/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
The discovery of iPSCs has led to the ex vivo production of differentiated cells for regenerative medicine. In the case of transfusion products, the derivation of platelets from iPSCs is expected to complement our current blood-donor supplied transfusion system through donor-independent production with complete pathogen-free assurance. This derivation can also overcome alloimmune platelet transfusion refractoriness by resulting in autologous, HLA-homologous or HLA-deficient products. Several developments were necessary to produce a massive number of platelets required for a single transfusion. First, expandable megakaryocytes were established from iPSCs through transgene expression. Second, a turbulent-type bioreactor with improved platelet yield and quality was developed. Third, novel drugs that enabled efficient feeder cell-free conditions were developed. Fourth, the platelet-containing suspension was purified and resuspended in an appropriate buffer. Finally, the platelet product needed to be assured for competency and safety including non-tumorigenicity through in vitro and in vivo preclinical tests. Based on these advancements, a clinical trial has started. The generation of human iPSC-derived platelets could evolve transfusion medicine to the next stage and assure a ubiquitous, safe supply of platelet products. Further, considering the feasibility of gene manipulations in iPSCs, other platelet products may bring forth novel therapeutic measures.
Collapse
|
35
|
Deslauriers AG, Kotini AG, Papapetrou EP. Modeling Leukemia Stem Cells with Patient-Derived Induced Pluripotent Stem Cells. Methods Mol Biol 2021; 2185:411-422. [PMID: 33165864 PMCID: PMC7821974 DOI: 10.1007/978-1-0716-0810-4_26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Patient-derived induced pluripotent stem cells (iPSCs) have recently provided a new way to model acute myeloid leukemia (AML) and other myeloid malignancies. Here, we describe methods for the generation of patient-derived iPSCs from leukemia cells and for their subsequent directed in vitro differentiation into hematopoietic cells that recapitulate features of leukemia stem cells (LSCs) and leukemic blasts.
Collapse
Affiliation(s)
- André G Deslauriers
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Biotech Research and Innovation Center, University of Copenhagen, København, Denmark
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andriana G Kotini
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
36
|
Zhang X, Li Z, Liu Y, Gai Z. Great Expectations: Induced pluripotent stem cell technologies in neurodevelopmental impairments. Int J Med Sci 2021; 18:459-473. [PMID: 33390815 PMCID: PMC7757149 DOI: 10.7150/ijms.51842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
Somatic cells such as skin fibroblasts, umbilical cord blood, peripheral blood, urinary epithelial cells, etc., are transformed into induced pluripotent stem cells (iPSCs) by reprogramming technology, a milestone in the stem-cell research field. IPSCs are similar to embryonic stem cells (ESCs), exhibiting the potential to differentiate into various somatic cells. Still, the former avoid problems of immune rejection and medical ethics in the study of ESCs and clinical trials. Neurodevelopmental disorders are chronic developmental brain dysfunctions that affect cognition, exercise, social adaptability, behavior, etc. Due to various inherited or acquired causes, they seriously affect the physical and psychological health of infants and children. These include generalized stunting / mental disability (GDD/ID), Epilepsy, autism spectrum disease (ASD), and attention deficit hyperactivity disorder (ADHD). Most neurodevelopmental disorders are challenging to cure. Establishing a neurodevelopmental disorder system model is essential for researching and treating neurodevelopmental disorders. At this stage, the scarcity of samples is a bigger problem for studying neurological diseases based on the donor, ethics, etc. Some iPSCs are reprogrammed from somatic cells that carry disease-causing mutations. They differentiate into nerve cells by induction, which has the original characteristics of diseases. Disease-specific iPSCs are used to study the mechanism and pathogenesis of neurodevelopmental disorders. The process provided samples and the impetus for developing drugs and developing treatment plans for neurodevelopmental disorders. Here, this article mainly introduced the development of iPSCs, the currently established iPSCs disease models, and artificial organoids related to neurodevelopmental impairments. This technology will promote our understanding of neurodevelopmental impairments and bring great expectations to children with neurological disorders.
Collapse
Affiliation(s)
- Xue Zhang
- Pediatric Research Institute, Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan 250022, China.,Jinan Pediatric Research Institute, Jinan Children's Hospital, Ji'nan 250022, China.,Neonatal Intensive Care Unit, Children's Medical Center, The Second Hospital of Shandong University, Ji'nan 250033, China
| | - Zilong Li
- Pediatric Research Institute, Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan 250022, China.,Jinan Pediatric Research Institute, Jinan Children's Hospital, Ji'nan 250022, China
| | - Yi Liu
- Pediatric Research Institute, Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan 250022, China.,Jinan Pediatric Research Institute, Jinan Children's Hospital, Ji'nan 250022, China
| | - Zhongtao Gai
- Pediatric Research Institute, Qilu Children's Hospital, Cheeloo College of Medicine, Shandong University, Ji'nan 250022, China.,Jinan Pediatric Research Institute, Jinan Children's Hospital, Ji'nan 250022, China
| |
Collapse
|
37
|
Spyrou N, Papapetrou EP. Studying leukemia stem cell properties and vulnerabilities with human iPSCs. Stem Cell Res 2020; 50:102117. [PMID: 33388708 PMCID: PMC8190184 DOI: 10.1016/j.scr.2020.102117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/16/2020] [Accepted: 12/04/2020] [Indexed: 12/17/2022] Open
Abstract
The reprogramming of cancer cells into induced pluripotent stem cells (iPSCs) can capture entire cancer genomes, and thus create genetically faithful models of human cancers. By providing stringent genetically clonal conditions, iPSC modeling can also unveil non-genetic sources of cancer heterogeneity and provide a unique opportunity to study them separately from genetic sources, as we recently showed in an iPSC-based model of acute myeloid leukemia (AML). Genetically clonal iPSCs, derived from a patient with AML, reproduce, upon hematopoietic differentiation, phenotypic and functional heterogeneity with all the hallmarks of a leukemia stem cell (LSC) hierarchy. Here we discuss the lessons that can be learned about the LSC state, its plasticity, stability and genetic and epigenetic determinants from iPSC modeling. We also discuss the practical and translational implications of exploiting AML-iPSCs to prospectively isolate large numbers of iLSCs for large-scale experiments, such as screens, and for discovery of new therapeutic targets specific to AML LSCs.
Collapse
Affiliation(s)
- Nikolaos Spyrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eirini P Papapetrou
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
38
|
Demirci S, Leonard A, Tisdale JF. Hematopoietic stem cells from pluripotent stem cells: Clinical potential, challenges, and future perspectives. Stem Cells Transl Med 2020; 9:1549-1557. [PMID: 32725882 PMCID: PMC7695636 DOI: 10.1002/sctm.20-0247] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
The generation of hematopoietic stem cells (HSCs) from induced pluripotent stem cells (iPSCs) is an active and promising area of research; however, generating engraftable HSCs remains a major obstacle. Ex vivo HSC derivation from renewable sources such as iPSCs offers an experimental tool for studying developmental hematopoiesis, disease modeling, and drug discovery, and yields tremendous therapeutic potential for malignant and nonmalignant hematological disorders. Although initial attempts mostly recapitulated yolk sac primitive/definitive hematopoiesis with inability to engraft, recent advances suggest the feasibility of engraftable HSC derivation from iPSCs utilizing ectopic transcription factor expression. Strategic development for de novo HSC generation includes further investigations of HSC ontogeny, and elucidation of critical signaling pathways, epigenetic modulations, HSC and iPSC microenvironment, and cell-cell interactions that contribute to stem cell biology and function.
Collapse
Affiliation(s)
- Selami Demirci
- Cellular and Molecular Therapeutics BranchNational Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - Alexis Leonard
- Cellular and Molecular Therapeutics BranchNational Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH)BethesdaMarylandUSA
| | - John F. Tisdale
- Cellular and Molecular Therapeutics BranchNational Heart Lung and Blood Institutes (NHLBI), National Institutes of Health (NIH)BethesdaMarylandUSA
| |
Collapse
|
39
|
McDonald D, Wu Y, Dailamy A, Tat J, Parekh U, Zhao D, Hu M, Tipps A, Zhang K, Mali P. Defining the Teratoma as a Model for Multi-lineage Human Development. Cell 2020; 183:1402-1419.e18. [PMID: 33152263 PMCID: PMC7704916 DOI: 10.1016/j.cell.2020.10.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 06/06/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022]
Abstract
We propose that the teratoma, a recognized standard for validating pluripotency in stem cells, could be a promising platform for studying human developmental processes. Performing single-cell RNA sequencing (RNA-seq) of 179,632 cells across 23 teratomas from 4 cell lines, we found that teratomas reproducibly contain approximately 20 cell types across all 3 germ layers, that inter-teratoma cell type heterogeneity is comparable with organoid systems, and teratoma gut and brain cell types correspond well to similar fetal cell types. Furthermore, cellular barcoding confirmed that injected stem cells robustly engraft and contribute to all lineages. Using pooled CRISPR-Cas9 knockout screens, we showed that teratomas can enable simultaneous assaying of the effects of genetic perturbations across all germ layers. Additionally, we demonstrated that teratomas can be sculpted molecularly via microRNA (miRNA)-regulated suicide gene expression to enrich for specific tissues. Taken together, teratomas are a promising platform for modeling multi-lineage development, pan-tissue functional genetic screening, and tissue engineering.
Collapse
Affiliation(s)
- Daniella McDonald
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA 92093, USA
| | - Yan Wu
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Amir Dailamy
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Justin Tat
- Department of Biological Sciences, University of California, San Diego, San Diego, CA 92093, USA
| | - Udit Parekh
- Department of Electrical and Computer Engineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Dongxin Zhao
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Michael Hu
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA
| | - Ann Tipps
- School of Medicine, University of California, San Diego, San Diego, CA 92103, USA
| | - Kun Zhang
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA 92093, USA.
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, San Diego, CA 92093, USA; Biomedical Sciences Graduate Program, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
40
|
Demirci S, Haro-Mora JJ, Leonard A, Drysdale C, Malide D, Keyvanfar K, Essawi K, Vizcardo R, Tamaoki N, Restifo NP, Tisdale JF, Uchida N. Definitive hematopoietic stem/progenitor cells from human embryonic stem cells through serum/feeder-free organoid-induced differentiation. Stem Cell Res Ther 2020; 11:493. [PMID: 33234163 PMCID: PMC7688003 DOI: 10.1186/s13287-020-02019-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Background Ex vivo production of hematopoietic stem/precursor cells (HSPCs) represents a promising versatile approach for blood disorders. Methods To derive definitive HSPCs from human embryonic stem cells (ESCs), we differentiated mesodermally specified embryoid bodies (EBs) on gelatin-coated plates in serum/feeder-free conditions. Results Seven-day EB maturation followed by an 8-day differentiation period on OP9 cells provided the highest number of definitive (CD34+ CD235a−, 69%, p < 0.01) and lowest number of primitive (CD34− CD235a+, 1.55%, p < 0.01) precursor cells along with the highest colony-forming units (149.8 ± 11.6, p < 0.01) in feeder-free conditions. Maximal HSPC fraction (CD34+ CD38− CD45RA− CD49f+ CD90+) was 7.6–8.9% after 10 days of hematopoietic differentiation with 14.5% adult β-globin expression following RBC differentiation. Myeloid and erythroid colonies were restricted strictly to the CD34+ CD43+ fraction (370.5 ± 65.7, p < 0.001), while the CD34− CD43+ fraction produced only a small number of colonies (21.6 ± 11.9). In addition, we differentiated the CD34+ CD43+ cells towards T-lymphocytes using the OP9/DLL1 co-culture system demonstrating double-positive T cells (CD4+ CD8+) with CD3+ expression displaying a broad T cell receptor (TCR) repertoire. Confocal imaging of organoid-like structures revealed a close association of CD31+ cells with CD34+ and CD43+ cells, suggesting a potential emergence of HSPCs through endothelial to hematopoietic transition. Furthermore, fluorescently labeled organoids exhibited the emergence of spherical non-attached cells from rare progenitors at the border of the organoid center. Conclusions In summary, definitive HSPCs can be derived from ESCs through a dynamic cellular process from an organoid-like structure, where erythroid progeny are capable of producing adult hemoglobin and lymphoid progeny shows a diverse TCR repertoire.
Collapse
Affiliation(s)
- Selami Demirci
- Sickle Cell Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bldg. 10, 9N112, Bethesda, MD, 20892, USA
| | - Juan J Haro-Mora
- Sickle Cell Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bldg. 10, 9N112, Bethesda, MD, 20892, USA
| | - Alexis Leonard
- Sickle Cell Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bldg. 10, 9N112, Bethesda, MD, 20892, USA
| | - Claire Drysdale
- Sickle Cell Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bldg. 10, 9N112, Bethesda, MD, 20892, USA
| | - Daniela Malide
- Light Microscopy Core Facility, NHLBI, NIH, Bethesda, MD, USA
| | | | - Khaled Essawi
- Sickle Cell Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bldg. 10, 9N112, Bethesda, MD, 20892, USA
| | - Raul Vizcardo
- National Cancer Institute, Center for Cancer Research, NIH, Bethesda, MD, USA
| | - Naritaka Tamaoki
- National Cancer Institute, Center for Cancer Research, NIH, Bethesda, MD, USA
| | - Nicholas P Restifo
- National Cancer Institute, Center for Cancer Research, NIH, Bethesda, MD, USA
| | - John F Tisdale
- Sickle Cell Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bldg. 10, 9N112, Bethesda, MD, 20892, USA.
| | - Naoya Uchida
- Sickle Cell Branch, National Heart Lung and Blood Institute (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health (NIH), 9000 Rockville Pike, Bldg. 10, 9N112, Bethesda, MD, 20892, USA
| |
Collapse
|
41
|
Donada A, Basso-Valentina F, Arkoun B, Monte-Mor B, Plo I, Raslova H. Induced pluripotent stem cells and hematological malignancies: A powerful tool for disease modeling and drug development. Stem Cell Res 2020; 49:102060. [PMID: 33142254 DOI: 10.1016/j.scr.2020.102060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 01/12/2023] Open
Abstract
The derivation of human pluripotent stem cell (iPSC) lines by in vitro reprogramming of somatic cells revolutionized research: iPSCs have been used for disease modeling, drug screening and regenerative medicine for many disorders, especially when combined with cutting-edge genome editing technologies. In hematology, malignant transformation is often a multi-step process, that starts with either germline or acquired genetic alteration, followed by progressive acquisition of mutations combined with the selection of one or more pre-existing clones. iPSCs are an excellent model to study the cooperation between different genetic alterations and to test relevant therapeutic drugs. In this review, we will describe the use of iPSCs for pathophysiological studies and drug testing in inherited and acquired hematological malignancies.
Collapse
Affiliation(s)
- A Donada
- INSERM, UMR1287, Université Paris Sud, Université Paris Saclay, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France
| | - F Basso-Valentina
- INSERM, UMR1287, Université Paris Sud, Université Paris Saclay, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France
| | - B Arkoun
- INSERM, UMR1287, Université Paris Sud, Université Paris Saclay, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France
| | - B Monte-Mor
- Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - I Plo
- INSERM, UMR1287, Université Paris Sud, Université Paris Saclay, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France
| | - H Raslova
- INSERM, UMR1287, Université Paris Sud, Université Paris Saclay, Gustave Roussy, Equipe Labellisée LNCC, Villejuif, France.
| |
Collapse
|
42
|
Beauchemin H, Möröy T. Multifaceted Actions of GFI1 and GFI1B in Hematopoietic Stem Cell Self-Renewal and Lineage Commitment. Front Genet 2020; 11:591099. [PMID: 33193732 PMCID: PMC7649360 DOI: 10.3389/fgene.2020.591099] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022] Open
Abstract
Growth factor independence 1 (GFI1) and the closely related protein GFI1B are small nuclear proteins that act as DNA binding transcriptional repressors. Both recognize the same consensus DNA binding motif via their C-terminal zinc finger domains and regulate the expression of their target genes by recruiting chromatin modifiers such as histone deacetylases (HDACs) and demethylases (LSD1) by using an N-terminal SNAG domain that comprises only 20 amino acids. The only region that is different between both proteins is the region that separates the zinc finger domains and the SNAG domain. Both proteins are co-expressed in hematopoietic stem cells (HSCs) and, to some extent, in multipotent progenitors (MPPs), but expression is specified as soon as early progenitors and show signs of lineage bias. While expression of GFI1 is maintained in lymphoid primed multipotent progenitors (LMPPs) that have the potential to differentiate into both myeloid and lymphoid cells, GFI1B expression is no longer detectable in these cells. By contrast, GFI1 expression is lost in megakaryocyte precursors (MKPs) and in megakaryocyte-erythrocyte progenitors (MEPs), which maintain a high level of GFI1B expression. Consequently, GFI1 drives myeloid and lymphoid differentiation and GFI1B drives the development of megakaryocytes, platelets, and erythrocytes. How such complementary cell type- and lineage-specific functions of GFI1 and GFI1B are maintained is still an unresolved question in particular since they share an almost identical structure and very similar biochemical modes of actions. The cell type-specific accessibility of GFI1/1B binding sites may explain the fact that very similar transcription factors can be responsible for very different transcriptional programming. An additional explanation comes from recent data showing that both proteins may have additional non-transcriptional functions. GFI1 interacts with a number of proteins involved in DNA repair and lack of GFI1 renders HSCs highly susceptible to DNA damage-induced death and restricts their proliferation. In contrast, GFI1B binds to proteins of the beta-catenin/Wnt signaling pathway and lack of GFI1B leads to an expansion of HSCs and MKPs, illustrating the different impact that GFI1 or GFI1B has on HSCs. In addition, GFI1 and GFI1B are required for endothelial cells to become the first blood cells during early murine development and are among those transcription factors needed to convert adult endothelial cells or fibroblasts into HSCs. This role of GFI1 and GFI1B bears high significance for the ongoing effort to generate hematopoietic stem and progenitor cells de novo for the autologous treatment of blood disorders such as leukemia and lymphoma.
Collapse
Affiliation(s)
| | - Tarik Möröy
- Institut de recherches cliniques de Montréal, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University, Montreal, QC, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
43
|
Al-Amoodi AS, Sakashita K, Ali AJ, Zhou R, Lee JM, Tehseen M, Li M, Belmonte JCI, Kusakabe T, Merzaban JS. Using Eukaryotic Expression Systems to Generate Human α1,3-Fucosyltransferases That Effectively Create Selectin-Binding Glycans on Stem Cells. Biochemistry 2020; 59:3757-3771. [PMID: 32901486 DOI: 10.1021/acs.biochem.0c00523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recruitment of circulating cells toward target sites is primarily dependent on selectin/ligand adhesive interactions. Glycosyltransferases are involved in the creation of selectin ligands on proteins and lipids. α1,3-Fucosylation is imperative for the creation of selectin ligands, and a number of fucosyltransferases (FTs) can modify terminal lactosamines on cells to create these ligands. One FT, fucosyltransferase VI (FTVI), adds a fucose in an α1,3 configuration to N-acetylglucosamine to generate sialyl Lewis X (sLex) epitopes on proteins of live cells and enhances their ability to bind E-selectin. Although a number of recombinant human FTVIs have been purified, apart from limited commercial enzymes, they were not characterized for their activity on live cells. Here we focused on establishing a robust method for producing FTVI that is active on living cells (hematopoietic cells and mesenchymal stromal cells). To this end, we used two expression systems, Bombyx mori (silkworm) and Pichia pastoris (yeast), to produce significant amounts of N-terminally tagged FTVI and demonstrated that these enzymes have superior activity when compared to currently available commercial enzymes that are produced from various expression systems. Overall, we outline a scheme for obtaining large amounts of highly active FTVI that can be used for the application of FTVI in enhancing the engraftment of cells lacking the sLex epitopes.
Collapse
Affiliation(s)
- Asma S Al-Amoodi
- Laboratory of Cell Migration and Signaling, Division of Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, Jeddah 23955, Saudi Arabia
| | - Kosuke Sakashita
- Laboratory of Cell Migration and Signaling, Division of Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, Jeddah 23955, Saudi Arabia
| | - Amal J Ali
- Laboratory of Cell Migration and Signaling, Division of Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, Jeddah 23955, Saudi Arabia
| | - Ruoyu Zhou
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Jae Man Lee
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Muhammad Tehseen
- Laboratory of DNA Replication and Recombination, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal 23955, Saudi Arabia
| | - Mo Li
- Laboratory of Stem Cell and Regeneration, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955, Saudi Arabia
| | - Juan Carlos I Belmonte
- Gene Expression Laboratory, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Takahiro Kusakabe
- Laboratory of Insect Genome Science, Kyushu University Graduate School of Bioresource and Bioenvironmental Sciences, Motooka 744, Nishi-ku, Fukuoka 819-0395, Japan
| | - Jasmeen S Merzaban
- Laboratory of Cell Migration and Signaling, Division of Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, Jeddah 23955, Saudi Arabia
| |
Collapse
|
44
|
Mace EM, Paust S, Conte MI, Baxley RM, Schmit MM, Patil SL, Guilz NC, Mukherjee M, Pezzi AE, Chmielowiec J, Tatineni S, Chinn IK, Akdemir ZC, Jhangiani SN, Muzny DM, Stray-Pedersen A, Bradley RE, Moody M, Connor PP, Heaps AG, Steward C, Banerjee PP, Gibbs RA, Borowiak M, Lupski JR, Jolles S, Bielinsky AK, Orange JS. Human NK cell deficiency as a result of biallelic mutations in MCM10. J Clin Invest 2020; 130:5272-5286. [PMID: 32865517 PMCID: PMC7524476 DOI: 10.1172/jci134966] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
Human natural killer cell deficiency (NKD) arises from inborn errors of immunity that lead to impaired NK cell development, function, or both. Through the understanding of the biological perturbations in individuals with NKD, requirements for the generation of terminally mature functional innate effector cells can be elucidated. Here, we report a cause of NKD resulting from compound heterozygous mutations in minichromosomal maintenance complex member 10 (MCM10) that impaired NK cell maturation in a child with fatal susceptibility to CMV. MCM10 has not been previously associated with monogenic disease and plays a critical role in the activation and function of the eukaryotic DNA replisome. Through evaluation of patient primary fibroblasts, modeling patient mutations in fibroblast cell lines, and MCM10 knockdown in human NK cell lines, we have shown that loss of MCM10 function leads to impaired cell cycle progression and induction of DNA damage-response pathways. By modeling MCM10 deficiency in primary NK cell precursors, including patient-derived induced pluripotent stem cells, we further demonstrated that MCM10 is required for NK cell terminal maturation and acquisition of immunological system function. Together, these data define MCM10 as an NKD gene and provide biological insight into the requirement for the DNA replisome in human NK cell maturation and function.
Collapse
Affiliation(s)
- Emily M. Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Silke Paust
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California, USA
| | - Matilde I. Conte
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Ryan M. Baxley
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Megan M. Schmit
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sagar L. Patil
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Nicole C. Guilz
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Malini Mukherjee
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics
| | - Ashley E. Pezzi
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics
| | - Jolanta Chmielowiec
- Center for Cell and Gene Therapy, and
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, Texas, USA
| | - Swetha Tatineni
- Department of Pediatrics
- Department of BioSciences, Rice University, Houston, Texas, USA
| | - Ivan K. Chinn
- Department of Pediatrics
- Department of Molecular and Human Genetics and
| | | | - Shalini N. Jhangiani
- Department of Molecular and Human Genetics and
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Donna M. Muzny
- Department of Molecular and Human Genetics and
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Asbjørg Stray-Pedersen
- Norwegian National Unit for Newborn Screening, Division of Pediatric and Adolescent Medicine, Oslo, Norway
| | - Rachel E. Bradley
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, Wales
| | - Mo Moody
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, Wales
| | - Philip P. Connor
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, Wales
| | - Adrian G. Heaps
- Department of Virology and Immunology, North Cumbria University Hospitals, Carlisle, United Kingdom
| | - Colin Steward
- Department of Paediatric Haematology, Oncology and Bone Marrow Transplantation, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Pinaki P. Banerjee
- Center for Human Immunobiology, Texas Children’s Hospital, Houston, Texas, USA
- Department of Pediatrics
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics and
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Malgorzata Borowiak
- Center for Cell and Gene Therapy, and
- Molecular and Cellular Biology Department, Baylor College of Medicine, Houston, Texas, USA
- Adam Mickiewicz University, Poznan, Poland
- McNair Medical Institute, Baylor College of Medicine, Houston, Texas, USA
| | - James R. Lupski
- Department of Pediatrics
- Department of Molecular and Human Genetics and
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, Wales
| | - Anja K. Bielinsky
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Jordan S. Orange
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
45
|
Gaggi G, Di Credico A, Izzicupo P, Alviano F, Di Mauro M, Di Baldassarre A, Ghinassi B. Human Mesenchymal Stromal Cells Unveil an Unexpected Differentiation Potential toward the Dopaminergic Neuronal Lineage. Int J Mol Sci 2020; 21:ijms21186589. [PMID: 32916865 PMCID: PMC7555006 DOI: 10.3390/ijms21186589] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 08/29/2020] [Accepted: 09/08/2020] [Indexed: 12/12/2022] Open
Abstract
Degeneration of dopaminergic neurons represents the cause of many neurodegenerative diseases, with increasing incidence worldwide. The replacement of dead cells with new healthy ones may represent an appealing therapeutic approach to these pathologies, but currently, only pluripotent stem cells can generate dopaminergic neurons with high efficiency. However, with the use of these cells arises safety and/or ethical issues. Human mesenchymal stromal cells (hFM-MSCs) are perinatal stem cells that can be easily isolated from the amniochorionic membrane after delivery. Generally considered multipotent, their real differentiative potential is not completely elucidated. The aim of this study was to analyze their stemness characteristics and to evaluate whether they may overcome their mesenchymal fate, generating dopaminergic neurons. We demonstrated that hFM-MSCs expressed embryonal genes OCT4, NANOG, SOX2, KLF4, OVOL1, and ESG1, suggesting they have some features of pluripotency. Moreover, hFM-MSCs that underwent a dopaminergic differentiation protocol gradually increased the transcription of dopaminergic markers LMX1b, NURR1, PITX3, and DAT. We finally obtained a homogeneous population of cells resembling the morphology of primary midbrain dopaminergic neurons that expressed the functional dopaminergic markers TH, DAT, and Nurr1. In conclusion, our results suggested that hFM-MSCs retain the expression of pluripotency genes and are able to differentiate not only into mesodermal cells, but also into neuroectodermal dopaminergic neuron-like cells.
Collapse
Affiliation(s)
- Giulia Gaggi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Andrea Di Credico
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Pascal Izzicupo
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| | - Francesco Alviano
- Department of Experimental Diagnostic and Speciality Medicine, Unit of Histology, Embriology and Applied Biology, University of Bologna, 40126 Bologna, Italy;
| | - Michele Di Mauro
- Cardio-Thoracic Surgery Unit, Heart and Vascular Centre, Maastricht University Medical Centre (MUMC), Cardiovascular Research Institute Maastricht (CARIM), 6202 Maastricht, The Netherlands;
| | - Angela Di Baldassarre
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
- Correspondence:
| | - Barbara Ghinassi
- Human Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G. D’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (B.G.)
| |
Collapse
|
46
|
Gaggi G, Di Credico A, Izzicupo P, Sancilio S, Di Mauro M, Iannetti G, Dolci S, Amabile G, Di Baldassarre A, Ghinassi B. Decellularized Extracellular Matrices and Cardiac Differentiation: Study on Human Amniotic Fluid-Stem Cells. Int J Mol Sci 2020; 21:E6317. [PMID: 32878275 PMCID: PMC7504221 DOI: 10.3390/ijms21176317] [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: 07/30/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Cell therapy with a variety of stem populations is increasingly being investigated as a promising regenerative strategy for cardiovascular (CV) diseases. Their combination with adequate scaffolds represents an improved therapeutic approach. Recently, several biomaterials were investigated as scaffolds for CV tissue repair, with decellularized extracellular matrices (dECMs) arousing increasing interest for cardiac tissue engineering applications. The aim of this study was to analyze whether dECMs support the cardiac differentiation of CardiopoieticAF stem cells. These perinatal stem cells, which can be easily isolated without ethical or safety limitations, display a high cardiac differentiative potential. Differentiation was previously achieved by culturing them on Matrigel, but this 3D scaffold is not transplantable. The identification of a new transplantable scaffold able to support CardiopoieticAF stem cell cardiac differentiation is pivotal prior to encouraging translation of in vitro studies in animal model preclinical investigations. Our data demonstrated that decellularized extracellular matrices already used in cardiac surgery (the porcine CorTMPATCH and the equine MatrixPatchTM) can efficiently support the proliferation and cardiac differentiation of CardiopoieticAF stem cells and represent a useful cellular scaffold to be transplanted with stem cells in animal hosts.
Collapse
Affiliation(s)
- Giulia Gaggi
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Andrea Di Credico
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Pascal Izzicupo
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Silvia Sancilio
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Michele Di Mauro
- Cardio-Thoracic Surgery Unit, Heart and Vascular Centre, Maastricht University Medical Centre (MUMC), Cardiovascular Research Institute Maastricht (CARIM), 6202 Maastricht, The Netherlands;
| | | | - Susanna Dolci
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | | | - Angela Di Baldassarre
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| | - Barbara Ghinassi
- Haman Anatomy and Cell Differentiation Lab, Department of Medicine and Aging Sciences, University “G.d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (G.G.); (A.D.C.); (P.I.); (S.S.); (A.D.B.)
| |
Collapse
|
47
|
Li X, Ma X, Chen Y, Peng D, Wang H, Chen S, Xiao Y, Li L, Zhou H, Cheng F, Gao Y, Chang J, Cheng T, Liu L. Coinhibition of activated p38 MAPKα and mTORC1 potentiates stemness maintenance of HSCs from SR1-expanded human cord blood CD34 + cells via inhibition of senescence. Stem Cells Transl Med 2020; 9:1604-1616. [PMID: 32602209 PMCID: PMC7695631 DOI: 10.1002/sctm.20-0129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 01/10/2023] Open
Abstract
The stemness of ex vivo expanded hematopoietic stem cells (HSCs) is usually compromised by current methods. To explore the failure mechanism of stemness maintenance of human HSCs, which were expanded from human umbilical cord blood (hUCB) CD34+ cells, by differentiation inhibitor Stem Regenin 1 (SR1), an antagonist of aryl hydrocarbon receptor, we investigated the activity of p38 mitogen‐activated protein kinase α (p38 MAPKα, p38α) and mammalian target of rapamycin complex 1 (mTORC1), and their effect on SR1‐expanded hUCB CD34+ cells. Our results showed that cellular senescence occurred in the SR1‐expanded hUCB CD34+ cells in which p38α and mTORC1 were successively activated. Furthermore, their coinhibition resulted in a further decrease in hUCB CD34+ cell senescence without an effect on apoptosis, promoted the maintenance of expanded phenotypic HSCs without differentiation inhibition, increased the hematopoietic reconstitution ability of multiple lineages, and potentiated the long‐term self‐renewal capability of HSCs from SR1‐expanded hUCB CD34+ cells in NOD/Shi‐scid/IL‐2Rγnull mice. Our mechanistic study revealed that senescence inhibition by our strategy was mainly attributed to downregulation of the splicesome, proteasome formation, and pyrimidine metabolism signaling pathways. These results suggest that coinhibition of activated p38α and mTORC1 potentiates stemness maintenance of HSCs from SR1‐expanded hUCB CD34+ cells via senescence inhibition. Thus, we established a new strategy to maintain the stemness of ex vivo differentiation inhibitor‐expanded human HSCs via coinhibition of multiple independent senescence initiating signal pathways. This senescence inhibition‐induced stemness maintenance of ex vivo expanded HSCs could also have an important role in other HSC expansion systems.
Collapse
Affiliation(s)
- Xiaoyi Li
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiao Ma
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Ying Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Danyue Peng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Huifang Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Suhua Chen
- Department of Gynaecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yin Xiao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Lei Li
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Fanjun Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Science, Tianjin, People's Republic of China
| | - Jiwei Chang
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, People's Republic of China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Chinese Academy of Medical Science, Tianjin, People's Republic of China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
48
|
Epigenetic Features of Human Perinatal Stem Cells Redefine Their Stemness Potential. Cells 2020; 9:cells9051304. [PMID: 32456308 PMCID: PMC7290760 DOI: 10.3390/cells9051304] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023] Open
Abstract
Human perinatal stem cells (SCs) can be isolated from fetal annexes without ethical or safety limitations. They are generally considered multipotent; nevertheless, their biological characteristics are still not fully understood. The aim of this study was to investigate the pluripotency potential of human perinatal SCs as compared to human induced pluripotent stem cells (hiPSCs). Despite the low expression of the pluripotent factors NANOG, OCT4, SOX2, and C-KIT in perinatal SC, we observed minor differences in the promoters DNA-methylation profile of these genes with respect to hiPSCs; we also demonstrated that in perinatal SCs miR-145-5p had an inverse trend in comparison to these stemness markers, suggesting that NANOG, OCT4, and SOX2 were regulated at the post-transcriptional level. The reduced expression of stemness markers was also associated with shorter telomere lengths and shift of the oxidative metabolism between hiPSCs and fetal annex-derived cells. Our findings indicate the differentiation ability of perinatal SCs might not be restricted to the mesenchymal lineage due to an epigenetic barrier, but other regulatory mechanisms such as telomere shortening or metabolic changes might impair their differentiation potential and challenge their clinical application.
Collapse
|
49
|
Duleba M, Yamamoto Y, Neupane R, Rao W, Xie J, Qi Y, Liew AA, Niroula S, Zhang Y, Mahalingam R, Wang S, Goller K, Ajani JA, Vincent M, Ho KY, Hou JK, Hyams JS, Sylvester FA, Crum CP, McKeon F, Xian W. Cloning of ground-state intestinal stem cells from endoscopic biopsy samples. Nat Protoc 2020; 15:1612-1627. [PMID: 32238950 DOI: 10.1038/s41596-020-0298-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/16/2020] [Indexed: 12/30/2022]
Abstract
'Adult' or 'somatic' stem cells harbor an intrinsic ability to regenerate tissues. Heterogeneity of such stem cells along the gastrointestinal tract yields the known segmental specificity of this organ and may contribute to the pathology of certain enteric conditions. Here we detail technology for the generation of 'libraries' of clonogenic cells from 1-mm-diamter endoscopic biopsy samples from the human gastrointestinal tract. Each of the 150-300 independent clones in a typical stem cell library can be clonally expanded to billions of cells in a few weeks while maintaining genomic stability and the ability to undergo multipotent differentiation to the specific epithelia from which the sample originated. The key to this methodology is the intrinsic immortality of normal intestinal stem cells (ISCs) and culture systems that maintain them as highly immature, ground-state ISCs marked by a single-cell clonogenicity of 70% and a corresponding 250-fold proliferative advantage over spheroid technologies. Clonal approaches such as this enhance the resolution of molecular genetics, make genome editing easier, and may be useful in regenerative medicine, unravelling heterogeneity in disease, and facilitating drug discovery.
Collapse
Affiliation(s)
- Marcin Duleba
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Yusuke Yamamoto
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Rahul Neupane
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Wei Rao
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Jingzhong Xie
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Yutao Qi
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Audrey-Ann Liew
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Suchan Niroula
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Yanting Zhang
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Rajasekaran Mahalingam
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Shan Wang
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Kristina Goller
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Matthew Vincent
- Tract Pharmaceuticals, Inc., Marlborough, Massachusetts, USA
| | - Khek Yu Ho
- Departments of Medicine and Pathology, National University of Singapore, Singapore, Singapore
| | - Jason K Hou
- Division of Gastroenterology, Baylor College of Medicine, Houston, Texas, USA
| | - Jeffrey S Hyams
- Division of Digestive Diseases, Hepatology, and Nutrition, Connecticut Children's Medical Center, Hartford, Connecticut, USA
| | - Francisco A Sylvester
- Division of Gastroenterology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Christopher P Crum
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Frank McKeon
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA.
| | - Wa Xian
- Stem Cell Center, Department of Biology and Biochemistry, University of Houston, Houston, Texas, USA.
| |
Collapse
|
50
|
Elahi S, Holling GA, Stablewski AB, Olejniczak SH. Improved hematopoietic differentiation of mouse embryonic stem cells through manipulation of the RNA binding protein ARS2. Stem Cell Res 2020; 43:101710. [PMID: 31986485 PMCID: PMC7406152 DOI: 10.1016/j.scr.2020.101710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 10/25/2022] Open
Abstract
The RNA binding protein ARS2 is highly expressed in hematopoietic progenitor populations and is required for adult hematopoiesis. Recent molecular studies found that ARS2 coordinates interactions between nascent RNA polymerase II transcripts and downstream RNA processing machineries, yet how such interactions influence hematopoiesis remains largely unknown. Techniques to differentiate embryonic stem cells (ESC) to hematopoietic progenitor cells (HPC) and mature blood cells have increased molecular understanding of hematopoiesis. Taking such an in vitro approach to examine the influence of ARS2 on hematopoiesis, we found that ARS2 suppresses expression of some HSC signature genes and differentiation of ESC to a HPC population (CSMD-HPC) identified by markers expressed on bone marrow resident hematopoietic stem cells. In line with ARS2's ability to promote proliferation of cultured cells, ARS2 knockout ESC showed limited expansion and yielded less CSMD-HPC than wild-type ESC. In contrast, transient ARS2 knockdown led to doubling the number of CSMD-HPC generated per ESC without affecting further differentiation into mature T-cells. Overall, data indicate that ARS2 negatively regulates early hematopoietic differentiation of ESC, in stark contrast to its supportive role in adult hematopoiesis. Consequently, manipulation of ARS2 expression and/or function has potential utility in hematopoietic cell engineering and regenerative medicine.
Collapse
Affiliation(s)
- Seerat Elahi
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - G Aaron Holling
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Aimee B Stablewski
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Scott H Olejniczak
- Department of Pathology and Anatomical Sciences, State University of New York at Buffalo, Buffalo, NY, United States; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States.
| |
Collapse
|