1
|
Kattaru S, Echambadi Loganathan S, Kodavala S, Chodimella CS, Potukuchi VGKS. Platelet-Derived Growth Factor Promotes Glomerular Mesangial Cells Differentiation of Human Bone Marrow Hematopoietic Stem Cells - An In Vitro Study. J Cell Biochem 2025; 126:e70012. [PMID: 40065657 DOI: 10.1002/jcb.70012] [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: 11/17/2024] [Revised: 01/24/2025] [Accepted: 02/20/2025] [Indexed: 05/13/2025]
Abstract
Glomerular filtration function and homeostasis are largely due to the cross-talk between podocytes, endothelial cells, and mesangial cells (MCs). Any disturbance in this association causes glomerular diseases (GD). Cell-based therapies are the best option in the treatment of GD. It is contemplated that hematopoietic stem cells (HSCs) are best suited to regenerate these cells; earlier, we have shown the differentiation of HSCs into podocytes. In this study, MCs formation was initiated with retinoic acid (RA), BMP-7, and Activin A, resulting in comma-shaped intermediate mesoderm (IM) cells prominently expressing Osr1. Followed by inducing with EGF, FGF, and BMP-7, which resulted in elongated metanephric mesenchyme (MM) cells conspicuously expressing Pax2, Wt1, Foxd1, and Eya1. Finally, MM cells were induced with platelet-derived growth factor to form polygonal-shaped MCs expressing α-smooth muscle actin, desmin, CD44, and PDGFRβ. The growing MCs showed positivity to periodic acid Schiff's, and ANAE staining with a prominent expression of the Itga8 elucidating phagocytic property of MCs. These MCs showed conspicuous expression of CD133, notch-2, and telomerase, determining the quiescence nature with a 31.2% proliferation rate revealed through Ki-67 staining. The functionality of MCs was assessed by growing MCs in 5.5 and 25 mM glucose concentrations, and noticeable expression of angiotensinogen, angiotensin-I and II, angiotensin-converting enzyme, collagen-4, fibronectin, and TGFβ1 was observed in 25 mM concentration, while lowered expression of these genes was observed in 5.5 mM concentration explaining the role of MCs in regulating the filtration pressure. All these findings demonstrate that HSCs can rejuvenate the insulted glomerulus.
Collapse
Affiliation(s)
- Surekha Kattaru
- Stem Cell Laboratory, Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | | | - Sireesha Kodavala
- Stem Cell Laboratory, Department of Biotechnology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Chandra Sekhar Chodimella
- Department of Hematology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | | |
Collapse
|
2
|
Deng J, Tan Y, Xu Z, Wang H. Advances in hematopoietic stem cells ex vivo expansion associated with bone marrow niche. Ann Hematol 2024; 103:5035-5057. [PMID: 38684510 DOI: 10.1007/s00277-024-05773-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Hematopoietic stem cells (HSCs) are an ideal source for the treatment of many hematological diseases and malignancies, as well as diseases of other systems, because of their two important features, self-renewal and multipotential differentiation, which have the ability to rebuild the blood system and immune system of the body. However, so far, the insufficient number of available HSCs, whether from bone marrow (BM), mobilized peripheral blood or umbilical cord blood, is still the main restricting factor for the clinical application. Therefore, strategies to expand HSCs numbers and maintain HSCs functions through ex vivo culture are urgently required. In this review, we outline the basic biology characteristics of HSCs, and focus on the regulatory factors in BM niche affecting the functions of HSCs. Then, we introduce several representative strategies used for HSCs from these three sources ex vivo expansion associated with BM niche. These findings have deepened our understanding of the mechanisms by which HSCs balance self-renewal and differentiation and provided a theoretical basis for the efficient clinical HSCs expansion.
Collapse
Affiliation(s)
- Ju Deng
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Tan
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhifang Xu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- The Key Laboratory of Molecular Diagnosis and Treatment of Hematological Disease of Shanxi Province, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
3
|
Elahimanesh M, Shokri N, Shabani R, Rahimi M, Najafi M. Exploring the potential of predicted miRNAs on the genes involved in the expansion of hematopoietic stem cells. Sci Rep 2024; 14:15551. [PMID: 38969714 PMCID: PMC11226654 DOI: 10.1038/s41598-024-66614-9] [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: 02/11/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024] Open
Abstract
A major challenge in therapeutic approaches applying hematopoietic stem cells (HSCs) is the cell quantity. The primary objective of this study was to predict the miRNAs and anti-miRNAs using bioinformatics tools and investigate their effects on the expression levels of key genes predicted in the improvement of proliferation, and the inhibition of differentiation in HSCs isolated from Human umbilical cord blood (HUCB). A network including genes related to the differentiation and proliferation stages of HSCs was constructed by enriching data of text (PubMed) and StemChecker server with KEGG signaling pathways, and was improved using GEO datasets. Bioinformatics tools predicted a profile from miRNAs containing miR-20a-5p, miR-423-5p, and chimeric anti-miRNA constructed from 5'-miR-340/3'-miR-524 for the high-score genes (RB1, SMAD4, STAT1, CALML4, GNG13, and CDKN1A/CDKN1B genes) in the network. The miRNAs and anti-miRNA were transferred into HSCs using polyethylenimine (PEI). The gene expression levels were estimated using the RT-qPCR technique in the PEI + (miRNA/anti-miRNA)-contained cell groups (n = 6). Furthermore, CD markers (90, 16, and 45) were evaluated using flow cytometry. Strong relationships were found between the high-score genes, miRNAs, and chimeric anti-miRNA. The RB1, SMAD4, and STAT1 gene expression levels were decreased by miR-20a-5p (P < 0.05). Additionally, the anti-miRNA increased the gene expression level of GNG13 (P < 0.05), whereas the miR-423-5p decreased the CDKN1A gene expression level (P < 0.01). The cellular count also increased significantly (P < 0.05) but the CD45 differentiation marker did not change in the cell groups. The study revealed the predicted miRNA/anti-miRNA profile expands HSCs isolated from HUCB. While miR-20a-5p suppressed the RB1, SMAD4, and STAT1 genes involved in cellular differentiation, the anti-miRNA promoted the GNG13 gene related to the proliferation process. Notably, the mixed miRNA/anti-miRNA group exhibited the highest cellular expansion. This approach could hold promise for enhancing the cell quantity in HSC therapy.
Collapse
Affiliation(s)
- Mohammad Elahimanesh
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Shokri
- Clinical Biochemistry Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Anatomy Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Rahimi
- Shahid Akbarabadi Clinical Research Development Unit (ShACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
- Clinical Biochemistry Department, Faculty of Medical Sciences, Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Jiang L, Ye Y, Han Y, Wang Q, Lu H, Li J, Qian W, Zeng X, Zhang Z, Zhao Y, Shi J, Luo Y, Qiu Y, Sun J, Sheng J, Huang H, Qian P. Microplastics dampen the self-renewal of hematopoietic stem cells by disrupting the gut microbiota-hypoxanthine-Wnt axis. Cell Discov 2024; 10:35. [PMID: 38548771 PMCID: PMC10978833 DOI: 10.1038/s41421-024-00665-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 03/01/2024] [Indexed: 04/01/2024] Open
Abstract
Microplastics (MPs) are contaminants ubiquitously found in the global biosphere that enter the body through inhalation or ingestion, posing significant risks to human health. Recent studies emerge that MPs are present in the bone marrow and damage the hematopoietic system. However, it remains largely elusive about the specific mechanisms by which MPs affect hematopoietic stem cells (HSCs) and their clinical relevance in HSC transplantation (HSCT). Here, we established a long-term MPs intake mouse model and found that MPs caused severe damage to the hematopoietic system. Oral gavage administration of MPs or fecal transplantation of microbiota from MPs-treated mice markedly undermined the self-renewal and reconstitution capacities of HSCs. Mechanistically, MPs did not directly kill HSCs but disrupted gut structure and permeability, which eventually ameliorated the abundance of Rikenellaceae and hypoxanthine in the intestine and inactivated the HPRT-Wnt signaling in bone marrow HSCs. Furthermore, administration of Rikenellaceae or hypoxanthine in mice as well as treatment of WNT10A in the culture system substantially rescued the MPs-induced HSC defects. Finally, we validated in a cohort of human patients receiving allogenic HSCT from healthy donors, and revealed that the survival time of patients was negatively correlated with levels of MPs, while positively with the abundance of Rikenellaceae, and hypoxanthine in the HSC donors' feces and blood. Overall, our study unleashes the detrimental roles and mechanisms of MPs in HSCs, which provides potential strategies to prevent hematopoietic damage from MPs and serves as a fundamental critique for selecting suitable donors for HSCT in clinical practice.
Collapse
Affiliation(s)
- Lingli Jiang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Yishan Ye
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yingli Han
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Qiwei Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Huan Lu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Jinxin Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Wenchang Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Xin Zeng
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Zhaoru Zhang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
| | - Yanmin Zhao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jimin Shi
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yi Luo
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yunfei Qiu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun Sun
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinghao Sheng
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China
| | - He Huang
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China.
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
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
|
6
|
Karima G, Kim HD. Unlocking the regenerative key: Targeting stem cell factors for bone renewal. J Tissue Eng 2024; 15:20417314241287491. [PMID: 39479284 PMCID: PMC11523181 DOI: 10.1177/20417314241287491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024] Open
Abstract
Stem cell factors (SCFs) are pivotal factors existing in both soluble and membrane-bound forms, expressed by endothelial cells (ECs) and fibroblasts throughout the body. These factors enhance cell growth, viability, and migration in multipotent cell lineages. The preferential expression of SCF by arteriolar ECs indicates that arterioles create a unique microenvironment tailored to hematopoietic stem cells (HSCs). Insufficiency of SCF within bone marrow (BM)-derived adipose tissue results in decreased their overall cellularity, affecting HSCs and their immediate progenitors critical for generating diverse blood cells and maintaining the hematopoietic microenvironment. SCF deficiency disrupts BM function, impacting the production and differentiation of HSCs. Additionally, deleting SCF from adipocytes reduces lipogenesis, highlighting the crucial role of SCF/c-kit signaling in controlling lipid accumulation. This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of recent advancements, challenges, and future directions for leveraging SCF as a key agent in regenerative medicine.
Collapse
Affiliation(s)
- Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Hwan D. Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
7
|
Halász H, Szatmári Z, Kovács K, Koppán M, Papp S, Szabó-Meleg E, Szatmári D. Changes of Ex Vivo Cervical Epithelial Cells Due to Electroporation with JMY. Int J Mol Sci 2023; 24:16863. [PMID: 38069185 PMCID: PMC10706833 DOI: 10.3390/ijms242316863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
The ionic environment within the nucleoplasm might diverge from the conditions found in the cytoplasm, potentially playing a role in the cellular stress response. As a result, it is conceivable that interactions of nuclear actin and actin-binding proteins (ABPs) with apoptosis factors may differ in the nucleoplasm and cytoplasm. The primary intracellular stress response is Ca2+ influx. The junctional mediating and regulating Y protein (JMY) is an actin-binding protein and has the capability to interact with the apoptosis factor p53 in a Ca2+-dependent manner, forming complexes that play a regulatory role in cytoskeletal remodelling and motility. JMY's presence is observed in both the cytoplasm and nucleoplasm. Here, we show that ex vivo ectocervical squamous cells subjected to electroporation with JMY protein exhibited varying morphological alterations. Specifically, the highly differentiated superficial and intermediate cells displayed reduced nuclear size. In inflamed samples, nuclear enlargement and simultaneous cytoplasmic reduction were observable and showed signs of apoptotic processes. In contrast, the less differentiated parabasal and metaplastic cells showed increased cytoplasmic activity and the formation of membrane protrusions. Surprisingly, in severe inflammation, vaginosis or ASC-US (Atypical Squamous Cells of Undetermined Significance), JMY appears to influence only the nuclear and perinuclear irregularities of differentiated cells, and cytoplasmic abnormalities still existed after the electroporation. Our observations can provide an appropriate basis for the exploration of the relationship between cytopathologically relevant morphological changes of epithelial cells and the function of ABPs. This is particularly important since ABPs are considered potential diagnostic and therapeutic biomarkers for both cancers and chronic inflammation.
Collapse
Affiliation(s)
- Henriett Halász
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | | | - Krisztina Kovács
- Department of Pathology, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | | | - Szilárd Papp
- DaVinci Clinics, 7635 Pécs, Hungary; (M.K.); (S.P.)
| | - Edina Szabó-Meleg
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| | - Dávid Szatmári
- Department of Biophysics, Medical School, University of Pécs, 7624 Pécs, Hungary; (H.H.); (E.S.-M.)
| |
Collapse
|
8
|
Bonilla X, Lara AM, Llano-León M, López-González DA, Hernández-Mejía DG, Bustos RH, Camacho-Rodríguez B, Perdomo-Arciniegas AM. Mesenchymal Stromal Cells from Perinatal Tissues as an Alternative for Ex Vivo Expansion of Hematopoietic Progenitor and Stem Cells from Umbilical Cord Blood. Int J Mol Sci 2023; 24:15544. [PMID: 37958529 PMCID: PMC10648510 DOI: 10.3390/ijms242115544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
Umbilical cord blood (UCB) serves as a source of hematopoietic stem and progenitor cells (HSPCs) utilized in the regeneration of hematopoietic and immune systems, forming a crucial part of the treatment for various benign and malignant hematological diseases. UCB has been utilized as an alternative HSPC source to bone marrow (BM). Although the use of UCB has extended transplantation access to many individuals, it still encounters significant challenges in selecting a histocompatible UCB unit with an adequate cell dose for a substantial proportion of adults with malignant hematological diseases. Consequently, recent research has focused on developing ex vivo expansion strategies for UCB HSPCs. Our results demonstrate that co-cultures with the investigated mesenchymal stromal cells (MSCs) enable a 10- to 15-fold increase in the cellular dose of UCB HSPCs while partially regulating the proliferation capacity when compared to HSPCs expanded with early acting cytokines. Furthermore, the secretory profile of UCB-derived MSCs closely resembles that of BM-derived MSCs. Moreover, both co-cultures exhibit alterations in cytokine secretion, which could potentially impact HSPC proliferation during the expansion process. This study underscores the fact that UCB-derived MSCs possess a remarkably similar supportive capacity to BM-derived MSCs, implying their potential use as feeder layers in the ex vivo expansion process of HSPCs.
Collapse
Affiliation(s)
- Ximena Bonilla
- Pharmaceutical Biotechnology Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá 111611, Colombia; (A.M.L.); (D.A.L.-G.); (B.C.-R.)
| | - Ana Milena Lara
- Pharmaceutical Biotechnology Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá 111611, Colombia; (A.M.L.); (D.A.L.-G.); (B.C.-R.)
| | - Manuela Llano-León
- Advanced Therapies Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá 111611, Colombia; (M.L.-L.); (D.G.H.-M.)
| | - David A. López-González
- Pharmaceutical Biotechnology Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá 111611, Colombia; (A.M.L.); (D.A.L.-G.); (B.C.-R.)
| | - David G. Hernández-Mejía
- Advanced Therapies Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá 111611, Colombia; (M.L.-L.); (D.G.H.-M.)
| | - Rosa Helena Bustos
- Therapeutic Evidence Group, Clinical Pharmacology, Universidad de La Sabana and Clínica Universidad de La Sabana, Chía 140013, Colombia;
| | - Bernardo Camacho-Rodríguez
- Pharmaceutical Biotechnology Unit, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, Bogotá 111611, Colombia; (A.M.L.); (D.A.L.-G.); (B.C.-R.)
| | | |
Collapse
|
9
|
Shao L, Paik N, Sanborn M, Bandara T, Vijaykumar A, Sottoriva K, Rehman J, Nombela-Arrieta C, Pajcini K. Hematopoietic Jagged1 is a fetal liver niche factor required for functional maturation and engraftment of fetal hematopoietic stem cells. Proc Natl Acad Sci U S A 2023; 120:e2210058120. [PMID: 37155858 PMCID: PMC10193977 DOI: 10.1073/pnas.2210058120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 04/04/2023] [Indexed: 05/10/2023] Open
Abstract
Notch signaling is essential for the emergence of definitive hematopoietic stem cells (HSCs) in the embryo and their development in the fetal liver niche. However, how Notch signaling is activated and which fetal liver cell type provides the ligand for receptor activation in HSCs is unknown. Here we provide evidence that endothelial Jagged1 (Jag1) has a critical early role in fetal liver vascular development but is not required for hematopoietic function during fetal HSC expansion. We demonstrate that Jag1 is expressed in many hematopoietic cells in the fetal liver, including HSCs, and that its expression is lost in adult bone marrow HSCs. Deletion of hematopoietic Jag1 does not affect fetal liver development; however, Jag1-deficient fetal liver HSCs exhibit a significant transplantation defect. Bulk and single-cell transcriptomic analysis of HSCs during peak expansion in the fetal liver indicates that loss of hematopoietic Jag1 leads to the downregulation of critical hematopoietic factors such as GATA2, Mllt3, and HoxA7, but does not perturb Notch receptor expression. Ex vivo activation of Notch signaling in Jag1-deficient fetal HSCs partially rescues the functional defect in a transplant setting. These findings indicate a new fetal-specific niche that is based on juxtracrine hematopoietic Notch signaling and reveal Jag1 as a fetal-specific niche factor essential for HSC function.
Collapse
Affiliation(s)
- Lijian Shao
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Na Yoon Paik
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Mark A. Sanborn
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL60612
| | - Thilinie Bandara
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Anjali Vijaykumar
- Department of Medical Oncology and Hematology, University Hospital Zurich, 8091Zurich, Switzerland
| | - Kilian Sottoriva
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL60612
| | - Cesar Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital Zurich, 8091Zurich, Switzerland
| | - Kostandin V. Pajcini
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL60612
| |
Collapse
|
10
|
Dausinas Ni P, Hartman M, Slack J, Basile C, Liu S, Wan J, O'Leary HA. Novel differential calcium regulation of hematopoietic stem and progenitor cells under physiological low oxygen conditions. J Cell Physiol 2023. [PMID: 37051890 DOI: 10.1002/jcp.30942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 11/28/2022] [Accepted: 12/23/2022] [Indexed: 04/14/2023]
Abstract
Low oxygen bone marrow (BM) niches (~1%-4% low O2 ) provide critical signals for hematopoietic stem/progenitor cells (HSC/HSPCs). Our presented data are the first to investigate live, sorted HSC/HSPCs in their native low O2 conditions. Transcriptional and proteomic analysis uncovered differential Ca2+ regulation that correlated with overlapping phenotypic populations consisting of robust increases of cytosolic and mitochondrial Ca2+ , ABC transporter (ABCG2) expression and sodium/hydrogen exchanger (NHE1) expression in live, HSC/HSPCs remaining in constant low O2. We identified a novel Ca2+ high population in HSPCs predominantly detected in low O2 that displayed enhanced frequency of phenotypic LSK/LSKCD150 in low O2 replating assays compared to Ca2+ low populations. Inhibition of the Ca2+ regulator NHE1 (Cariporide) resulted in attenuation of both the low O2 induced Ca2+ high population and subsequent enhanced maintenance of phenotypic LSK and LSKCD150 during low O2 replating. These data reveal multiple levels of differential Ca2+ regulation in low O2 resulting in phenotypic, signaling, and functional consequences in HSC/HSPCs.
Collapse
Affiliation(s)
- Paige Dausinas Ni
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Melissa Hartman
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jacob Slack
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christopher Basile
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Center of Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Heather A O'Leary
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| |
Collapse
|
11
|
The differentiation of human induced pluripotent stem cells into hematopoietic stem cells on 3D bone scaffold in a dynamic culture system. Tissue Cell 2023; 82:102044. [PMID: 36905860 DOI: 10.1016/j.tice.2023.102044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Hematopoietic stem cell transplantation is used for cell-based therapy for many hematological disorders. However, difficulty in finding proper donors has limited this source of stem cells. For clinical application, the generation of these cells from induced pluripotent stem cells (iPSs) is a fascinating and endless source. One of the experimental methods to generate HSCs from iPSs is the mimicking of the hematopoietic niche. In the current study, as the first phase of differentiation, embryoid bodies were formed from iPSs. They were then cultured in different dynamic conditions in order to determine the appropriate settings for their differentiation into HSCs. The dynamic culture was composed of DBM Scaffold with or without growth factor. After ten days, the specific HSC markers (CD34, CD133, CD31 and CD45) were assessed using flow-cytometry. Our findings demonstrated that the dynamic conditions were significantly suitable than static ones. In addition, in 3D scaffold and dynamic system the expression of CXCR4, as a homing marker, was increased. These results suggest that the 3D culture bioreactor with DBM scaffold could provide a new approach for differentiation of iPSs into HSCs. Moreover, this system could provide maximum mimicry of bone marrow niche.
Collapse
|
12
|
Albayrak E, Kocabaş F. Therapeutic targeting and HSC proliferation by small molecules and biologicals. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:425-496. [PMID: 37061339 DOI: 10.1016/bs.apcsb.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Hematopoietic stem cells (HSCs) have considerably therapeutic value on autologous and allogeneic transplantation for many malignant/non-malignant hematological diseases, especially with improvement of gene therapy. However, acquirement of limited cell dose from HSC sources is the main handicap for successful transplantation. Therefore, many strategies based on the utilization of various cytokines, interaction of stromal cells, modulation of several extrinsic and intrinsic factors have been developed to promote ex vivo functional HSC expansion with high reconstitution ability until today. Besides all these strategies, small molecules become prominent with their ease of use and various advantages when they are translated to the clinic. In the last two decades, several small molecule compounds have been investigated in pre-clinical studies and, some of them were evaluated in different stages of clinical trials for their safety and efficiencies. In this chapter, we will present an overview of HSC biology, function, regulation and also, pharmacological HSC modulation with small molecules from pre-clinical and clinical perspectives.
Collapse
|
13
|
Li M, Morse B, Kassim S. Development and clinical translation considerations for the next wave of gene modified hematopoietic stem and progenitor cells therapies. Expert Opin Biol Ther 2022; 22:1177-1191. [PMID: 35833356 DOI: 10.1080/14712598.2022.2101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Consistent and reliable manufacture of gene modified hematopoietic stem and progenitor cell (HPSC) therapies will be of the utmost importance as they become more mainstream and address larger populations. Robust development campaigns will be needed to ensure that these products will be delivered to patients with the highest quality standards. AREAS COVERED Through publicly available manuscripts, press releases, and news articles - this review touches on aspects related to HSPC therapy, development, and manufacturing. EXPERT OPINION Recent advances in genome modification technology coupled with the longstanding clinical success of HSPCs warrants great optimism for the next generation of engineered HSPC-based therapies. Treatments for some diseases that have thus far been intractable now appear within reach. Reproducible manufacturing will be of critical importance in delivering these therapies but will be challenging due to the need for bespoke materials and methods in combination with the lack of off-the-shelf solutions. Continued progress in the field will manifest in the form of industrialization which currently requires attention and resources directed toward the custom reagents, a focus on closed and automated processes, and safer and more precise genome modification technologies that will enable broader, faster, and safer access to these life-changing therapies.
Collapse
Affiliation(s)
| | - Brent Morse
- Dark Horse Consulting Group, Walnut Creek, CA, USA
| | | |
Collapse
|
14
|
Mayer IM, Hoelbl-Kovacic A, Sexl V, Doma E. Isolation, Maintenance and Expansion of Adult Hematopoietic Stem/Progenitor Cells and Leukemic Stem Cells. Cancers (Basel) 2022; 14:1723. [PMID: 35406494 PMCID: PMC8996967 DOI: 10.3390/cancers14071723] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are rare, self-renewing cells that perch on top of the hematopoietic tree. The HSCs ensure the constant supply of mature blood cells in a tightly regulated process producing peripheral blood cells. Intense efforts are ongoing to optimize HSC engraftment as therapeutic strategy to treat patients suffering from hematopoietic diseases. Preclinical research paves the way by developing methods to maintain, manipulate and expand HSCs ex vivo to understand their regulation and molecular make-up. The generation of a sufficient number of transplantable HSCs is the Holy Grail for clinical therapy. Leukemia stem cells (LSCs) are characterized by their acquired stem cell characteristics and are responsible for disease initiation, progression, and relapse. We summarize efforts, that have been undertaken to increase the number of long-term (LT)-HSCs and to prevent differentiation towards committed progenitors in ex vivo culture. We provide an overview and compare methods currently available to isolate, maintain and enrich HSC subsets, progenitors and LSCs and discuss their individual advantages and drawbacks.
Collapse
Affiliation(s)
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; (I.M.M.); (A.H.-K.); (E.D.)
| | | |
Collapse
|
15
|
Mann Z, Sengar M, Verma YK, Rajalingam R, Raghav PK. Hematopoietic Stem Cell Factors: Their Functional Role in Self-Renewal and Clinical Aspects. Front Cell Dev Biol 2022; 10:664261. [PMID: 35399522 PMCID: PMC8987924 DOI: 10.3389/fcell.2022.664261] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/14/2022] [Indexed: 01/29/2023] Open
Abstract
Hematopoietic stem cells (HSCs) possess two important properties such as self-renewal and differentiation. These properties of HSCs are maintained through hematopoiesis. This process gives rise to two subpopulations, long-term and short-term HSCs, which have become a popular convention for treating various hematological disorders. The clinical application of HSCs is bone marrow transplant in patients with aplastic anemia, congenital neutropenia, sickle cell anemia, thalassemia, or replacement of damaged bone marrow in case of chemotherapy. The self-renewal attribute of HSCs ensures long-term hematopoiesis post-transplantation. However, HSCs need to be infused in large numbers to reach their target site and meet the demands since they lose their self-renewal capacity after a few passages. Therefore, a more in-depth understanding of ex vivo HSCs expansion needs to be developed to delineate ways to enhance the self-renewability of isolated HSCs. The multifaceted self-renewal process is regulated by factors, including transcription factors, miRNAs, and the bone marrow niche. A developed classical hierarchical model that outlines the hematopoiesis in a lineage-specific manner through in vivo fate mapping, barcoding, and determination of self-renewal regulatory factors are still to be explored in more detail. Thus, an in-depth study of the self-renewal property of HSCs is essentially required to be utilized for ex vivo expansion. This review primarily focuses on the Hematopoietic stem cell self-renewal pathway and evaluates the regulatory molecular factors involved in considering a targeted clinical approach in numerous malignancies and outlining gaps in the current knowledge.
Collapse
Affiliation(s)
- Zoya Mann
- Independent Researcher, New Delhi, India
| | - Manisha Sengar
- Department of Zoology, Deshbandhu College, University of Delhi, Delhi, India
| | - Yogesh Kumar Verma
- Stem Cell and Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), Delhi, India
| | - Raja Rajalingam
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Pawan Kumar Raghav
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
16
|
Jaffredo T, Balduini A, Bigas A, Bernardi R, Bonnet D, Canque B, Charbord P, Cumano A, Delwel R, Durand C, Fibbe W, Forrester L, de Franceschi L, Ghevaert C, Gjertsen B, Gottgens B, Graf T, Heidenreich O, Hermine O, Higgs D, Kleanthous M, Klump H, Kouskoff V, Krause D, Lacaud G, Celso CL, Martens JH, Méndez-Ferrer S, Menendez P, Oostendorp R, Philipsen S, Porse B, Raaijmakers M, Robin C, Stunnenberg H, Theilgaard-Mönch K, Touw I, Vainchenker W, Corrons JLV, Yvernogeau L, Schuringa JJ. The EHA Research Roadmap: Normal Hematopoiesis. Hemasphere 2021; 5:e669. [PMID: 34853826 PMCID: PMC8615310 DOI: 10.1097/hs9.0000000000000669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/02/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Thierry Jaffredo
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | | | - Anna Bigas
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- Josep Carreras Leukemia Research Institute (IJC), Barcelona, Spain
- Centro de Investigación Biomedica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Bernardi
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Bruno Canque
- INSERM U976, Universite de Paris, Ecole Pratique des Hautes Etudes/PSL Research University, Institut de Recherche Saint Louis, France
| | - Pierre Charbord
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Anna Cumano
- Unité Lymphopoïèse, Département d’Immunologie, INSERM U1223, Institut Pasteur, Cellule Pasteur, Université de Paris, France
| | - Ruud Delwel
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Charles Durand
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Willem Fibbe
- Leiden University Medical Center, The Netherlands
| | - Lesley Forrester
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Scotland
| | | | | | - Bjørn Gjertsen
- Department of Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Berthold Gottgens
- Wellcome - MRC Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, United Kingdom
| | - Thomas Graf
- Center for Genomic Regulation, Barcelona Institute for Science and Technology and Universitat Pompeu Fabra, Barcelona, Spain
| | - Olaf Heidenreich
- Prinses Máxima Centrum voor kinderoncologie, Utecht, The Netherlands
| | - Olivier Hermine
- Department of Hematology and Laboratory of Physiopathology and Treatment of Blood Disorders, Hôpital Necker, Imagine institute, University of Paris, France
| | - Douglas Higgs
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | | | - Hannes Klump
- Institute for Transfusion Medicine, University Hospital Essen, Germany
| | | | - Daniela Krause
- Goethe University Frankfurt and Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - George Lacaud
- Cancer Research UK Manchester Institute, The University of Manchester, United Kingdom
| | | | - Joost H.A. Martens
- Department of Molecular Biology, RIMLS, Radboud University, Nijmegen, The Netherlands
| | | | - Pablo Menendez
- Centro de Investigación Biomedica en Red-Oncología (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- RICORS-RETAV, Instituto de Salud Carlos III, Madrid, Spain
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| | - Robert Oostendorp
- Department of Internal Medicine III, Technical University of Munich, School of Medicine, Germany
| | - Sjaak Philipsen
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, The Netherlands
| | - Bo Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Marc Raaijmakers
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Catherine Robin
- Hubrecht Institute-KNAW and University Medical Center Utrecht, The Netherlands
- Regenerative medicine center, University Medical Center Utrecht, The Netherlands
| | - Henk Stunnenberg
- Prinses Máxima Centrum voor kinderoncologie, Utecht, The Netherlands
| | - Kim Theilgaard-Mönch
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Denmark
- Department of Hematology, Rigshospitalet/National University Hospital, University of Copenhagen, Denmark
| | - Ivo Touw
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Joan-Lluis Vives Corrons
- Red Blood Cell and Hematopoietic Disorders Research Unit, Institute for Leukaemia Research Josep Carreras, Badalona, Barcelona
| | - Laurent Yvernogeau
- Sorbonne Université, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement/UMR7622, Paris, France
| | - Jan Jacob Schuringa
- Department of Experimental Hematology, University Medical Center Groningen, The Netherlands
| |
Collapse
|
17
|
Sudo K, Yamazaki S, Wilkinson AC, Nakauchi H, Nakamura Y. Polyvinyl alcohol hydrolysis rate and molecular weight influence human and murine HSC activity ex vivo. Stem Cell Res 2021; 56:102531. [PMID: 34509158 PMCID: PMC8629160 DOI: 10.1016/j.scr.2021.102531] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/22/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022] Open
Abstract
Ex vivo expansion of hematopoietic stem cells (HSCs) is one of the most promising strategies to increase the availability of transplantable HSCs and improve bone marrow transplantation outcomes. We recently demonstrated that mouse HSCs could be efficiently expanded in polyvinyl alcohol (PVA)-containing culture medium using only recombinant stem cell factor and thrombopoietin cytokines. However, the behavior of human HSCs in these simple PVA-based media was not fully elucidated. In this study, we analyzed the compatibility of PVA of different hydrolysis rates (HR) and molecular weights (MW) to support functional human and mouse HSCs ex vivo. Human and mouse HSCs proliferated more frequently in media containing PVA with lower HR than with higher HR, but both PVA types supported HSC multilineage reconstitution potential. Importantly, human HSCs cultured in PVA-containing media engrafted not only in irradiated recipients but also in non-irradiated recipients. Our results demonstrate that human HSCs can be maintained ex vivo using PVA-based culture systems and suggest approaches for future optimization of human HSC expansion.
Collapse
Affiliation(s)
- Kazuhiro Sudo
- Cell Engineering Division, BioResource Research Center, RIKEN, Tsukuba, Japan.
| | - Satoshi Yamazaki
- Division of Stem Cell Therapy, Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Laboratory of Stem Cell Therapy, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan.
| | - Adam C Wilkinson
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, UK
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Institute for Stem Cell Biology and Regenerative Medicine and Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yukio Nakamura
- Cell Engineering Division, BioResource Research Center, RIKEN, Tsukuba, Japan
| |
Collapse
|
18
|
Yao CL, Huang P, Liu TC, Lin YK, Chen CY, Lai YT, Chin TY, Tseng TY, Hsu YC. CCL2 associated with CD38 expression during ex vivo expansion in human cord blood-derived hematopoietic stem cells. Aging (Albany NY) 2021; 13:19878-19893. [PMID: 34375303 PMCID: PMC8386547 DOI: 10.18632/aging.203398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/08/2021] [Indexed: 11/26/2022]
Abstract
To date, different experimental strategies have been developed for the ex vivo expansion of human hematopoietic stem cells (HSCs) for clinical applications. However, differences in the genomic function of expanded HSCs under different culture systems remain unclear. In this study, we compared the gene expression profiles of HSCs in ex vivo expanded serum (10% FBS, fetal bovine serum) and serum-free culture systems and analyzed the molecular functions of differentially expressed genes using microarray chips. We identified 839 differentially expressed genes between the two culture systems. These genes were enriched in the TNF -regulated inflammatory pathway in an FBS culture system. In addition, the mRNA expression of CCL2 (C-C motif chemokine ligand 2), TNF (tumor necrosis factor) and FOS (FBJ murine osteosarcoma viral oncogene homolog) was validated by RT-qPCR. Our data revealed that ex vivo expansion of HSCs using the FBS culture system induces an inflammatory response and high CD38 expression, indicating that this system might activate an inflammatory pathway and induce expression of the cancer marker CD38 during ex vivo expansion of HSCs. This study provides a transcriptional profile and new insights into the genomic functions of HSCs under different expanded cultures.
Collapse
Affiliation(s)
- Chao-Ling Yao
- Department of Chemical Engineering, National Cheng Kung University, Tainan 701, Taiwan.,Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan.,Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan
| | - Poyin Huang
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan.,Department of Neurology, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung City 807, Taiwan.,Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 807, Taiwan.,Department of Neurology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
| | - Tsai-Chi Liu
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| | - Yung-Kai Lin
- Institute of Food Safety and Risk Management, National Taiwan Ocean University, Keelung City 202, Taiwan.,Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung City 402, Taiwan
| | - Ching-Yun Chen
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| | - Yi-Ting Lai
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan
| | - Tzu Yun Chin
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| | - Tsung-Yu Tseng
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Chung-Li, Taoyuan City 320, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Chung-Li, Taoyuan City 320, Taiwan
| |
Collapse
|
19
|
Fernandes SS, Limaye LS, Kale VP. Differentiated Cells Derived from Hematopoietic Stem Cells and Their Applications in Translational Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:29-43. [PMID: 34114129 DOI: 10.1007/5584_2021_644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Hematopoietic stem cells (HSCs) and their development are one of the most widely studied model systems in mammals. In adults, HSCs are predominantly found in the bone marrow, from where they maintain homeostasis. Besides bone marrow and mobilized peripheral blood, cord blood is also being used as an alternate allogenic source of transplantable HSCs. HSCs from both autologous and allogenic sources are being applied for the treatment of various conditions like blood cancers, anemia, etc. HSCs can further differentiate to mature blood cells. Differentiation process of HSCs is being extensively studied so as to obtain a large number of pure populations of various differentiated cells in vitro so that they can be taken up for clinical trials. The ability to generate sufficient quantity of clinical-grade specialized blood cells in vitro would take the field of hematology a step ahead in translational medicine.
Collapse
Affiliation(s)
| | - Lalita S Limaye
- Stem Cell Lab, National Centre for Cell Science, Pune, India
| | - Vaijayanti P Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, India.
| |
Collapse
|
20
|
Zimran E, Papa L, Hoffman R. Ex vivo expansion of hematopoietic stem cells: Finally transitioning from the lab to the clinic. Blood Rev 2021; 50:100853. [PMID: 34112560 DOI: 10.1016/j.blre.2021.100853] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 01/17/2023]
Abstract
Hematopoietic stem cells (HSCs) have been used for therapeutic purposes for decades in the form of autologous and allogeneic transplantation and are currently emerging as an attractive target for gene therapy. A low stem cell dose is a major barrier to the application of HSC therapy in several situations, primarily umbilical cord blood transplantation and gene modification. Strategies that promote ex vivo expansion of the numbers of functional HSCs could overcome this barrier, hence have been the subject of intense and prolonged research. Several ex vivo expansion strategies have advanced to evaluation clinical trials, which are showing favorable outcomes along with convincing safety signals. Preclinical studies have recently confirmed beneficial incorporation of ex vivo expansion into HSC gene modification protocols. Collectively, ex vivo HSC expansion holds promise for significantly broadening the availability of cord blood units for transplantation, and for optimizing gene therapy protocols to enable their clinical application.
Collapse
Affiliation(s)
- Eran Zimran
- Hematology Department, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Kiryat Hadassah 1, POB 1200, Jerusalem, 911200, Israel.
| | - Luena Papa
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levi Place, Box 1079, New York, NY 10029, USA.
| | - Ronald Hoffman
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levi Place, Box 1079, New York, NY 10029, USA.
| |
Collapse
|
21
|
Sica RA, Terzioglu MK, Mahmud D, Mahmud N. Mechanistic Basis of ex Vivo Umbilical Cord Blood Stem Progenitor Cell Expansion. Stem Cell Rev Rep 2021; 16:628-638. [PMID: 32424674 DOI: 10.1007/s12015-020-09981-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Umbilical cord blood (CB) transplantation has been used successfully in humans for three decades due to its rapid availability for patients lacking a suitable allogeneic donor, less stringent HLA matching requirements, and low rates of relapse and chronic graft-versus-host disease (GVHD). However, CB transplantation is associated with complications, such as delayed hematopoietic engraftment, graft failure, which increases infection and bleeding and causes longer hospital stays, and transplant-related mortality. The majority of these biological limitations are due to the unforeseeable functional potency of multipotent hematopoietic stem cells (HSCs), which reduce the predictability of successful transplantation; however, several strategies have been developed to increase the number of hematopoietic stem progenitor cells (HSPCs) infused during CB transplantation. This review primarily addresses the methods that promote ex vivo CB expansion within the context of symmetrical and asymmetrical HSC division and those that rely on epigenetic mechanisms, along with the reportedly most successful cytokine combinations. We also review recent clinical research on small molecules (StemRegenin-1, UM171, and nicotinamide) in ex vivo expanded CB and discuss yet unvalidated preclinical strategies. Expanding and transplanting CB graft enriched in HSPCs in a single CB unit is a particularly exciting prospect with the potential to improve the use and availability of CB grafts. Greater knowledge of optimal ex vivo expansion strategies, cell longevity, and graft potency will expand the scope of cellular therapies. Also the development of adequate ex vivo HSPC expansion strategies could bring expanded cord blood grafts to the forefront of transplant therapy and regenerative medicine.
Collapse
Affiliation(s)
- R Alejandro Sica
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA
| | - Meryem K Terzioglu
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA
| | - Dolores Mahmud
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA
| | - Nadim Mahmud
- Division of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine Chicago, 840 South Wood Street, Clinical Sciences Building (CSB) Rm# 826, Chicago, IL, 60612, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
22
|
Ghafouri-Fard S, Niazi V, Taheri M, Basiri A. Effect of Small Molecule on ex vivo Expansion of Cord Blood Hematopoietic Stem Cells: A Concise Review. Front Cell Dev Biol 2021; 9:649115. [PMID: 33898442 PMCID: PMC8063724 DOI: 10.3389/fcell.2021.649115] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/22/2021] [Indexed: 12/29/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are a group of cells being produced during embryogenesis to preserve the blood system. They might also be differentiated to non-hematopoietic cells, including neural, cardiac and myogenic cells. Therefore, they have vast applications in the treatment of human disorders. Considering the restricted quantities of HSCs in the umbilical cord blood, inadequate mobilization of bone marrow stem cells, and absence of ethnic dissimilarity, ex vivo expansion of these HSCs is an applicable method for obtaining adequate amounts of HSCs. Several molecules such as NR-101, zVADfmk, zLLYfmk, Nicotinamide, Resveratrol, the Copper chelator TEPA, dmPGE2, Garcinol, and serotonin have been used in combination of cytokines to expand HSCs ex vivo. The most promising results have been obtained from cocktails that influence multipotency and self-renewal features from different pathways. In the current manuscript, we provide a concise summary of the effects of diverse small molecules on expansion of cord blood HSCs.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Abstract
Blood is generated throughout life by continued proliferation and differentiation of hematopoietic progenitors, while at the top of the hierarchy, hematopoietic stem cells (HSCs) remain largely quiescent. This way HSCs avoid senescence and preserve their capacity to repopulate the hematopoietic system. But HSCs are not always quiescent, proliferating extensively in conditions such as those found in the fetal liver. Understanding the elusive mechanisms that regulate HSC fate would enable us to comprehend a crucial piece of HSC biology and pave the way for ex-vivo HSC expansion with clear clinical benefit. Here we review how metabolism, endoplasmic reticulum stress and oxidative stress condition impact HSCs decision to self-renew or differentiate and how these signals integrate into the mammalian target of rapamycin (mTOR) pathway. We argue that the bone marrow microenvironment continuously favors differentiation through the activation of the mTOR complex (mTORC)1 signaling, while the fetal liver microenvironment favors self-renewal through the inverse mechanism. In addition, we also postulate that strategies that have successfully achieved HSC expansion, directly or indirectly, lead to the inactivation of mTORC1. Finally, we propose a mechanism by which mTOR signaling, during cell division, conditions HSC fate. This mechanism has already been demonstrated in mature hematopoietic cells (T-cells), that face a similar decision after activation, either undergoing clonal expansion or differentiation.
Collapse
|
24
|
Papa L, Djedaini M, Kintali M, Schaniel C, Hoffman R. Ex Vivo Expansion of Adult Hematopoietic Stem and Progenitor Cells with Valproic Acid. Methods Mol Biol 2021; 2185:267-280. [PMID: 33165854 DOI: 10.1007/978-1-0716-0810-4_16] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Abstract
Umbilical cord blood (UCB) units provide an alternative source of human hematopoietic stem cells (HSCs) for patients who require allogeneic stem cell transplantation but lack a matched donor. However, the limited number of HSCs within each UCB unit remains a major challenge for their use in regenerative medicine and HSC transplantation in adults. Efficient expansion of human HSCs in ex vivo cultures initiated with CD34+ cells isolated from UCBs can overcome this limitation. The method described here utilizes a deacetylase inhibitor, valproic acid (VPA), to rapidly expand to a high degree the numbers of functional HSCs and committed progenitors (HPCs). The expanded HSCs are capable of establishing both short-term and long-term multilineage hematopoietic reconstitution. This highly reproducible and simple protocol can be also applied to expansion of both HSCs and HPCs from different sources including the bone marrow and peripheral blood.
Collapse
Affiliation(s)
- Luena Papa
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mansour Djedaini
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manisha Kintali
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christoph Schaniel
- Department of Pharmacological Sciences, Mount Sinai Institute for System Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Cell, Developmental and Regenerative Biology, Black Family Stem Cell Institute, Mount Sinai Institute for System Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Hoffman
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
25
|
Papa L, Djedaini M, Martin TC, Zangui M, Beaumont KG, Sebra R, Parsons R, Schaniel C, Hoffman R. Limited Mitochondrial Activity Coupled With Strong Expression of CD34, CD90 and EPCR Determines the Functional Fitness of ex vivo Expanded Human Hematopoietic Stem Cells. Front Cell Dev Biol 2020; 8:592348. [PMID: 33384995 PMCID: PMC7769876 DOI: 10.3389/fcell.2020.592348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/10/2020] [Indexed: 01/04/2023] Open
Abstract
Ex vivo expansion strategies of human hematopoietic stem cell (HSC) grafts with suboptimal stem cell dose have emerged as promising strategies for improving outcomes of HSC transplantation in patients with hematological malignancies. While exposure of HSCs to ex vivo cultures expands the number of phenotypically identifiable HSCs, it frequently alters the transcriptomic and metabolic profiles, therefore, compromising their long-term (LT) hematopoietic reconstitution capacity. Within the heterogeneous pool of expanded HSCs, the precise phenotypic, transcriptomic and metabolic profile and thus, the identity of HSCs that confer LT repopulation potential remains poorly described. Utilizing valproic acid (VPA) in ex vivo cultures of umbilical cord blood (UCB)-CD34+ cells, we demonstrate that expanded HSCs phenotypically marked by expression of the stem cell markers CD34, CD90 and EPCR (CD201) are highly enriched for LT-HSCs. Furthermore, we report that low mitochondrial membrane potential, and, hence, mitochondrial activity distinguishes LT-HSCs within the expanded pool of phenotypically defined HSCs. Remarkably, such reduced mitochondrial activity is restricted to cells with the highest expression levels of CD34, CD90 and EPCR phenotypic markers. Together, our findings reveal that high expression of CD34, CD90 and EPCR in conjunction with low mitochondrial activity is critical for identification of functional LT-HSCs generated within ex vivo expansion cultures.
Collapse
Affiliation(s)
- Luena Papa
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mansour Djedaini
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Tiphaine C Martin
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Mahtab Zangui
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kristin G Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ramon Parsons
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Christoph Schaniel
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ronald Hoffman
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
26
|
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: 42] [Impact Index Per Article: 8.4] [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
|
27
|
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: 2.2] [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
|
28
|
Marquez Romero S, Hettler F, Hausinger R, Schreck C, Landspersky T, Henkel L, Angerpointner C, Demir IE, Schiemann M, Bassermann F, Götze KS, Istvánffy R, Oostendorp RAJ. Secreted factors from mouse embryonic fibroblasts maintain repopulating function of single cultured hematopoietic stem cells. Haematologica 2020; 106:2633-2640. [PMID: 33543864 PMCID: PMC8485655 DOI: 10.3324/haematol.2020.249102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Indexed: 11/09/2022] Open
Abstract
Hematopoietic stem cell self-renewal, proliferation, and differentiation are independently regulated by intrinsic as well as extrinsic mechanisms. We previously demonstrated that murine proliferation of hematopoietic stem cells is supported in serum-free medium supplemented with two growth factors, stem cell factor and interleukin 11. The survival of hematopoietic stem cells is additionally improved by supplementing this medium with two more growth factors, neural growth factor and collagen 1 (four growth factors) or serum-free medium conditioned by the hematopoietic stem cell-supportive stromal UG26-1B6 cells1. Here, we describe a robust and versatile alternative source of conditioned medium from mouse embryonic fibroblasts. We found that this conditioned medium supports survival and phenotypical identity of hematopoietic stem cells, as well as cell cycle entry in single cell cultures of CD34- CD48- CD150+ Lineage- SCA1+ KIT+ cells supplemented with two growth factors. Strikingly, in comparison with cultures in serum-free medium with four growth factors, conditioned medium from mouse embryonic fibroblasts increases the numbers of proliferating clones and the number of Lineage- SCA1+ KIT+ cells, both with two and four growth factors. In addition, conditioned medium from mouse embryonic fibroblasts supports self-renewal in culture of cells with short- and long-term hematopoiesis-repopulating ability in vivo. These findings identify conditioned medium from mouse embryonic fibroblasts as a robust alternative serumfree source of factors to maintain self-renewal of in vivo-repopulating hematopoetic stem cells in culture.
Collapse
Affiliation(s)
- Sandra Marquez Romero
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich, Germany; Technical University of Munich, Klinikum rechts der Isar, Department of Surgery, Munich
| | - Franziska Hettler
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich
| | - Renate Hausinger
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich
| | - Christina Schreck
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich
| | - Theresa Landspersky
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich
| | - Lynette Henkel
- Technical University of Munich, Flow Cytometry Unit of the Technical University Munich, Institute for Medical Microbiology, Immunology and Hygiene (CyTUM-MIH), Munich
| | - Corinne Angerpointner
- Technical University of Munich, Flow Cytometry Unit of the Technical University Munich, Institute for Medical Microbiology, Immunology and Hygiene (CyTUM-MIH), Munich
| | - Ihsan E Demir
- Technical University of Munich, Klinikum rechts der Isar, Department of Surgery, Munich
| | - Matthias Schiemann
- Technical University of Munich, Flow Cytometry Unit of the Technical University Munich, Institute for Medical Microbiology, Immunology and Hygiene (CyTUM-MIH), Munich
| | - Florian Bassermann
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg
| | - Katharina S Götze
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg
| | - Rouzanna Istvánffy
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich, Germany; Technical University of Munich, Klinikum rechts der Isar, Department of Surgery, Munich
| | - Robert A J Oostendorp
- Technical University of Munich, Klinikum rechts der Isar, Clinic and Polyclinic for Internal Medicine III, Munich.
| |
Collapse
|
29
|
Zimran E, Papa L, Djedaini M, Patel A, Iancu-Rubin C, Hoffman R. Expansion and preservation of the functional activity of adult hematopoietic stem cells cultured ex vivo with a histone deacetylase inhibitor. Stem Cells Transl Med 2020; 9:531-542. [PMID: 31950644 PMCID: PMC7103619 DOI: 10.1002/sctm.19-0199] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 12/27/2019] [Indexed: 12/17/2022] Open
Abstract
Attempts to expand ex vivo the numbers of human hematopoietic stem cells (HSCs) without compromising their marrow repopulating capacity and their ability to establish multilineage hematopoiesis has been the subject of intense investigation. Although most such efforts have focused on cord blood HSCs, few have been applied to adult HSCs, a more clinically relevant HSC source for gene modification. To date, the strategies that have been used to expand adult HSCs have resulted in modest effects or HSCs with lineage bias and a limited ability to generate T cells in vivo. We previously reported that culturing umbilical cord blood CD34+ cells in serum‐free media supplemented with valproic acid (VPA), a histone deacetylase inhibitor, and a combination of cytokines led to the expansion of the numbers of fully functional HSCs. In the present study, we used this same approach to expand the numbers of adult human CD34+ cells isolated from mobilized peripheral blood and bone marrow. This approach resulted in a significant increase in the numbers of phenotypically defined HSCs (CD34+CD45RA‐CD90+D49f+). Cells incubated with VPA also exhibited increased aldehyde dehydrogenase activity and decreased mitochondrial membrane potential, each functional markers of HSCs. Grafts harvested from VPA‐treated cultures were able to engraft in immune‐deficient mice and, importantly, to generate cellular progeny belonging to each hematopoietic lineage in similar proportion to that observed with unmanipulated CD34+ cells. These data support the utility of VPA‐mediated ex vivo HSC expansion for gene modification of adult HSCs.
Collapse
Affiliation(s)
- Eran Zimran
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.,Hematology Department, Hadassah University Center, Jerusalem, Israel
| | - Luena Papa
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mansour Djedaini
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ami Patel
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Camelia Iancu-Rubin
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ronald Hoffman
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|