1
|
Yu P, Liu B, Dong C, Chang Y. Induced Pluripotent Stem Cells-Based Regenerative Therapies in Treating Human Aging-Related Functional Decline and Diseases. Cells 2025; 14:619. [PMID: 40277944 DOI: 10.3390/cells14080619] [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: 03/11/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025] Open
Abstract
A significant increase in life expectancy worldwide has resulted in a growing aging population, accompanied by a rise in aging-related diseases that pose substantial societal, economic, and medical challenges. This trend has prompted extensive efforts within many scientific and medical communities to develop and enhance therapies aimed at delaying aging processes, mitigating aging-related functional decline, and addressing aging-associated diseases to extend health span. Research in aging biology has focused on unraveling various biochemical and genetic pathways contributing to aging-related changes, including genomic instability, telomere shortening, and cellular senescence. The advent of induced pluripotent stem cells (iPSCs), derived through reprogramming human somatic cells, has revolutionized disease modeling and understanding in humans by addressing the limitations of conventional animal models and primary human cells. iPSCs offer significant advantages over other pluripotent stem cells, such as embryonic stem cells, as they can be obtained without the need for embryo destruction and are not restricted by the availability of healthy donors or patients. These attributes position iPSC technology as a promising avenue for modeling and deciphering mechanisms that underlie aging and associated diseases, as well as for studying drug effects. Moreover, iPSCs exhibit remarkable versatility in differentiating into diverse cell types, making them a promising tool for personalized regenerative therapies aimed at replacing aged or damaged cells with healthy, functional equivalents. This review explores the breadth of research in iPSC-based regenerative therapies and their potential applications in addressing a spectrum of aging-related conditions.
Collapse
Affiliation(s)
- Peijie Yu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Bin Liu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Cheng Dong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| | - Yun Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hunghom, Hong Kong 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
2
|
Skamagki M, Zhang C, Hacisuleyman E, Galleti G, Wu C, Vinagolu RK, Cha H, Ata D, Kim J, Weiskittel T, Diop M, Aung T, Del Latto M, Kim AS, Li Z, Miele M, Zhao R, Tang LH, Hendrickson RC, Romesser PB, Smith JJ, Giannakakou P, Darnell RB, Bott MJ, Li H, Kim K. Aging-dependent dysregulation of EXOSC2 is maintained in cancer as a dependency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647279. [PMID: 40236131 PMCID: PMC11996493 DOI: 10.1101/2025.04.04.647279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Reprogramming of aged donor tissue cells into induced pluripotent stem cells (A-iPSC) preserved the epigenetic memory of aged-donor tissue, defined as genomic instability and poor tissue differentiation in our previous study. The unbalanced expression of RNA exosome subunits affects the RNA degradation complex function and is associated with geriatric diseases including premature aging and cancer progression. We hypothesized that the age-dependent progressive subtle dysregulation of EXOSC2 (exosome component 2) causes the aging traits (abnormal cell cycle and poor tissue differentiation). We used embryonic stem cells as a tool to study EXOSC2 function as the aging trait epigenetic memory determined in A-iPSC because these aging traits could not be studied in senesced aged cells or immortalized cancer cells. We found that the regulatory subunit of PP2A phosphatase, PPP2R5E, is a key target of EXOSC2 and this regulation is preserved in stem cells and cancer.
Collapse
|
3
|
Tsai HH, Hsiao FC, Yu AL, Juang JH, Yu J, Chu PH. Empagliflozin Reduces High Glucose-Induced Cardiomyopathy in hiPSC-Derived Cardiomyocytes : Glucose-induced Lipotoxicity in hiPSC-Derived Cardiomyocytes. Stem Cell Rev Rep 2025; 21:849-858. [PMID: 39841369 DOI: 10.1007/s12015-024-10839-8] [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] [Accepted: 12/26/2024] [Indexed: 01/23/2025]
Abstract
Human-induced pluripotent stem cell (hiPSC) technology has been applied in pathogenesis studies, drug screening, tissue engineering, and stem cell therapy, and patient-specific hiPSC-derived cardiomyocytes (hiPSC-CMs) have shown promise in disease modeling, including diabetic cardiomyopathy. High glucose (HG) treatment induces lipotoxicity in hiPSC-CMs, as evidenced by changes in cell size, beating rate, calcium handling, and lipid accumulation. Empagliflozin, an SGLT2 inhibitor, effectively mitigates the hypertrophic changes, abnormal calcium handling, and contractility impairment induced by HG. Glucose concentration influences SGLT2 expression in cardiomyocytes, highlighting its potential role in diabetic cardiomyopathy. These findings support the potential utility of hiPSC-CMs in studying diabetic cardiomyopathy and the efficacy of empagliflozin in ameliorating HG-induced cardiomyocyte dysfunction. Such research may advance developments in precision medicine and therapeutic interventions for patients with diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiu-Hui Tsai
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Fu-Chih Hsiao
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
- Department of Pediatrics, University of California in San Diego, San Diego, CA, USA
| | - Jyuhn-Huarng Juang
- Division of Endocrinology and Metabolism, Department of Internal Medicine and Center for Tissue Engineering, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Pao-Hsien Chu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
4
|
Mathew‐Schmitt S, Oerter S, Reitenbach E, Gätzner S, Höchner A, Jahnke H, Piontek J, Neuhaus W, Brachner A, Metzger M, Appelt‐Menzel A. Generation of Advanced Blood-Brain Barrier Spheroids Using Human-Induced Pluripotent Stem Cell-Derived Brain Capillary Endothelial-Like Cells. Adv Biol (Weinh) 2025; 9:e2400442. [PMID: 39912766 PMCID: PMC12001013 DOI: 10.1002/adbi.202400442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/12/2024] [Indexed: 02/07/2025]
Abstract
Extensively studied blood-brain barrier (BBB) in-vitro models are established on 2D cell culture inserts. However, they do not accurately represent 3D in-vivo microenvironments due to lack of direct neurovascular unit cellular contacts. Here, the establishment and characterization of a self-assembled 3D BBB spheroid model using human-induced pluripotent stem cell (hiPSC)-derived brain capillary endothelial-like cells (iBCECs) in combination with primary human astrocytes (ACs) and pericytes (PCs) are reported. This investigation compares 3D spheroids with 2D mono-cultured iBCECs derived from two different hiPSC lines and two differentiation strategies. It is observed that spheroid properties vary depending on the differentiation strategy or type of hiPSC line applied for model generation. However, spheroids demonstrate in-vivo like tight junction ultrastructure and, in comparison to 2D models, higher transcript expression of BBB specific genes. Furthermore, they possess characteristic barrier integrity, barrier functionality, and protein expression. It is inferred that hiPSC-derived BBB spheroids hold a strong potential as a reliable future BBB in-vitro test system.
Collapse
Affiliation(s)
- Sanjana Mathew‐Schmitt
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
| | - Sabrina Oerter
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Evelin Reitenbach
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Sabine Gätzner
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
| | - Alevtina Höchner
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Heinz‐Georg Jahnke
- Biotechnological‐Biomedical Center (BBZ)University of Leipzig04103LeipzigGermany
| | - Jörg Piontek
- Clinical Physiology/Nutritional MedicineDepartment of GastroenterologyRheumatology and Infectious DiseasesCharité–Universitätsmedizin Berlin12203BerlinGermany
| | - Winfried Neuhaus
- AIT Austrian Institute of Technology GmbHCentre Health and Bioresources, Competence Unit Molecular DiagnosticsVienna1210Austria
- Department of MedicineFaculty Dentistry and MedicinePrivate Danube UniversityKrems3500Austria
| | - Andreas Brachner
- AIT Austrian Institute of Technology GmbHCentre Health and Bioresources, Competence Unit Molecular DiagnosticsVienna1210Austria
| | - Marco Metzger
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| | - Antje Appelt‐Menzel
- Chair Tissue Engineering and Regenerative Medicine (TERM)University Hospital Würzburg97070WürzburgGermany
- Fraunhofer Institute for Silicate Research ISCTranslational Centre Regenerative Therapies (TLC‐RT)97070WürzburgGermany
| |
Collapse
|
5
|
Musthafa T, Nizami SK, Mishra A, Hasan G, Gopurappilly R. Altered Mitochondrial Bioenergetics and Calcium Kinetics in Young-Onset PLA2G6 Parkinson's Disease iPSCs. J Neurochem 2025; 169:e70059. [PMID: 40189860 PMCID: PMC11973445 DOI: 10.1111/jnc.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Parkinson's disease (PD) has emerged as a multisystem disorder affecting multiple cellular and organellar systems in addition to the dopaminergic neurons. Disease-specific induced pluripotent stem cells (iPSCs) model early developmental changes and cellular perturbations that are otherwise inaccessible from clinical settings. Here, we report the early changes in patient-derived iPSCs carrying a homozygous recessive mutation, R741Q, in the PLA2G6 gene. A gene-edited R747W iPSC line mirrored these phenotypes, thus validating our initial findings. Bioenergetic dysfunction and hyperpolarization of mitochondrial membrane potentials were hallmarks of the PD iPSCs. Further, a concomitant increase in glycolytic activity indicated a possible compensation for mitochondrial respiration. Elevated basal reactive oxygen species (ROS) and decreased catalase expression were also observed in the disease iPSCs. No change in autophagy was detected. These inceptive changes could be potential targets for early intervention of prodromal PD in the absence of disease-modifying therapies. However, additional investigations are crucial to delineate the cause-effect relationships of these observations.
Collapse
Affiliation(s)
- Thasneem Musthafa
- National Centre for Biological SciencesTata Institute of Fundamental ResearchBangaloreIndia
| | - Syed Kavish Nizami
- National Centre for Biological SciencesTata Institute of Fundamental ResearchBangaloreIndia
| | - Ankita Mishra
- NKure Therapeutics Pvt LtdCentre for Cellular and Molecular PlatformsBangaloreIndia
| | - Gaiti Hasan
- National Centre for Biological SciencesTata Institute of Fundamental ResearchBangaloreIndia
- Centre for High Impact Neuroscience and Translational ApplicationsKolkataIndia
| | - Renjitha Gopurappilly
- National Centre for Biological SciencesTata Institute of Fundamental ResearchBangaloreIndia
- NKure Therapeutics Pvt LtdCentre for Cellular and Molecular PlatformsBangaloreIndia
| |
Collapse
|
6
|
Azhdari M, Hausen AZ, Aghdami N, Baghaban-Eslaminejad M. Efficient and Reproducible Differentiation Protocol for Pluripotent Stem Cells into Functional Endothelial Cells: Unveiling the Path to Vascular Regeneration. Arch Med Res 2025; 56:103142. [PMID: 39837100 DOI: 10.1016/j.arcmed.2024.103142] [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: 05/30/2024] [Revised: 11/01/2024] [Accepted: 11/21/2024] [Indexed: 01/23/2025]
Abstract
INTRODUCTION Endothelial cells (ECs) play a crucial role in many treatments for cardiovascular diseases, such as blood vessel repair, tissue engineering, and drug delivery. The process of differentiating these cells is complex and involves various sources and numerous molecular and cellular events. Differentiating pluripotent stem cells (PSCs) into endothelial cells are one of the most effective sources for creating ECs in the lab and offers great potential for regenerative medicine. However, different cell types can appear during differentiation process. OBJECTIVES This study presents a reliable and reproducible protocol for efficiently differentiating human pluripotent stem cells (hPSCs) into mature endothelial cells with high purity (>98%). METHODS FLK1+ cells were isolated from hPSCs using fluorescence-activated cell sorting (FACS). Then isolated FLK1+ cells differentiated into high-purity endothelial cells (ECs) by adding endothelial growth factors (VEGF, FGF, and EGM-2 medium). The differentiated ECs were extensively characterized by evaluating key endothelial markers and assessing their functional abilities, such as tube formation and response to angiogenic signals. Finally, the ECs were further purified using a second FACS step with a CD31 antibody. RESULTS The differentiated hPSC-derived endothelial cells (hPSC-ECs) expressed high levels of PECAM-1 (CD31), VE-cadherin (CD144), and von Willebrand factor (vWF), with more than 98% of the cells showing these markers. Additionally, the hPSC-ECs formed tubular structures and effectively took up acetylated fluorescently-labeled low-density lipoprotein (DiI-ac-LDL), demonstrating their functionality as endothelial cells. CONCLUSION Our study clarifies the molecular mechanisms involved in the differentiation of hPSCs into endothelial cells, emphasizing key signaling pathways important for determining endothelial cell fate. These findings provide a framework for the scalable production of transplantable endothelial cells, representing a significant advancement in personalized therapies and tissue engineering for regenerative medicine.
Collapse
Affiliation(s)
- Manizheh Azhdari
- Department of Pathology, School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, Academic Center for Education, Culture and Research, Tehran, Iran.
| | - Axel Zur Hausen
- Department of Pathology, School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| | - Mohamadreza Baghaban-Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran, Iran
| |
Collapse
|
7
|
Tashkandi AJ, Gorman A, McGoldrick Mathers E, Carney G, Yacoub A, Setyaningsih WAW, Kuburas R, Margariti A. Metabolic and Mitochondrial Dysregulations in Diabetic Cardiac Complications. Int J Mol Sci 2025; 26:3016. [PMID: 40243689 PMCID: PMC11988959 DOI: 10.3390/ijms26073016] [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: 02/01/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The growing prevalence of diabetes highlights the urgent need to study diabetic cardiovascular complications, specifically diabetic cardiomyopathy, which is a diabetes-induced myocardial dysfunction independent of hypertension or coronary artery disease. This review examines the role of mitochondrial dysfunction in promoting diabetic cardiac dysfunction and highlights metabolic mechanisms such as hyperglycaemia-induced oxidative stress. Chronic hyperglycaemia and insulin resistance can activate harmful pathways, including advanced glycation end-products (AGEs), protein kinase C (PKC) and hexosamine signalling, uncontrolled reactive oxygen species (ROS) production and mishandling of Ca2+ transient. These processes lead to cardiomyocyte apoptosis, fibrosis and contractile dysfunction. Moreover, endoplasmic reticulum (ER) stress and dysregulated RNA-binding proteins (RBPs) and extracellular vesicles (EVs) contribute to tissue damage, which drives cardiac function towards heart failure (HF). Advanced patient-derived induced pluripotent stem cell (iPSC) cardiac organoids (iPS-COs) are transformative tools for modelling diabetic cardiomyopathy and capturing human disease's genetic, epigenetic and metabolic hallmarks. iPS-COs may facilitate the precise examination of molecular pathways and therapeutic interventions. Future research directions encourage the integration of advanced models with mechanistic techniques to promote novel therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Refik Kuburas
- Wellcome Wolfson Institute of Experimental Medicine, Queens University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.J.T.); (A.G.); (E.M.M.); (G.C.); (A.Y.); (W.A.W.S.)
| | - Andriana Margariti
- Wellcome Wolfson Institute of Experimental Medicine, Queens University Belfast, Belfast BT9 7BL, Northern Ireland, UK; (A.J.T.); (A.G.); (E.M.M.); (G.C.); (A.Y.); (W.A.W.S.)
| |
Collapse
|
8
|
Fong H, Mendel M, Jascur J, Najmi L, Kim K, Lew G, Garimalla S, Schock S, Hu J, Villegas AG, Conway A, Fontenot JD, Zompi S. A serum- and feeder-free system to generate CD4 and regulatory T cells from human iPSCs. Stem Cells 2025; 43:sxaf001. [PMID: 39878584 DOI: 10.1093/stmcls/sxaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/02/2025] [Indexed: 01/31/2025]
Abstract
iPSCs can serve as a renewable source of a consistent edited cell product, overcoming limitations of primary cells. While feeder-free generation of clinical grade iPSC-derived CD8 T cells has been achieved, differentiation of iPSC-derived CD4sp and regulatory T cells requires mouse stromal cells in an artificial thymic organoid. Here we report a serum- and feeder-free differentiation process suitable for large-scale production. Using an optimized concentration of PMA/Ionomycin, we generated iPSC-CD4sp T cells at high efficiency and converted them to Tregs using TGFβ and ATRA. Using genetic engineering, we demonstrated high, non-viral, targeted integration of an HLA-A2 CAR in iPSCs. iPSC-Tregs ± HLA-A2-targeted CAR phenotypically, transcriptionally and functionally resemble primary Tregs and suppress T-cell proliferation in vitro. Our work is the first to demonstrate an iPSC-based platform amenable to manufacturing CD4 T cells to complement iPSC-CD8 oncology products and functional iPSC-Tregs to deliver Treg cell therapies at scale.
Collapse
Affiliation(s)
- Helen Fong
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Proximity Therapeutics, San Francisco, CA 94107, United States
| | | | - John Jascur
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Proximity Therapeutics, San Francisco, CA 94107, United States
| | - Laeya Najmi
- Sangamo Therapeutics, Richmond, CA 94804, United States
- BioMarin, Novato, CA 94949, United States
| | - Ken Kim
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Garrett Lew
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Swetha Garimalla
- Sangamo Therapeutics, Richmond, CA 94804, United States
- OmniAb, Emeryville, CA 94608, United States
| | | | - Jing Hu
- Sangamo Therapeutics, Richmond, CA 94804, United States
| | - Andres Gordillo Villegas
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Kodiak Sciences, Palo Alto, CA 94304, United States
| | - Anthony Conway
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Replay, San Diego, CA 92121, United States
| | - Jason D Fontenot
- Sangamo Therapeutics, Richmond, CA 94804, United States
- Stylus Medicine, Cambridge, MA 02139, United States
| | - Simona Zompi
- Sangamo Therapeutics, Richmond, CA 94804, United States
- CARGO Therapeutics, San Carlos, CA 94070, United States
| |
Collapse
|
9
|
Diane A, Mu-U-Min RBA, Al-Siddiqi HH. Epigenetic memory as crucial contributing factor in directing the differentiation of human iPSC into pancreatic β-cells in vitro. Cell Tissue Res 2025; 399:267-276. [PMID: 39883142 PMCID: PMC11870940 DOI: 10.1007/s00441-025-03952-8] [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: 10/16/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
Impaired insulin secretion contributes to the pathogenesis of type 1 diabetes mellitus through autoimmune destruction of pancreatic β-cells and the pathogenesis of severe forms of type 2 diabetes mellitus through β-cell dedifferentiation and other mechanisms. Replenishment of malfunctioning β-cells via islet transplantation has the potential to induce long-term glycemic control in the body. However, this treatment option cannot widely be implemented in clinical due to healthy islet donor shortage. Emerging β-cell replacement with human-induced pluripotent stem cell (iPSC) provides high remedial therapy hopes. Thus, tremendous progress has been made in developing β-cell differentiation protocols in vitro; however, most of the differentiated iPSC-derived β-cells showed immature phenotypes associated with low efficiency depending on the iPSC lines used, creating a crucial barrier for their clinical implementation. Multiple mechanisms including differences in genetic, cell cycle patterns, and mitochondrial dysfunction underlie the defective differentiation propensity of iPSC into insulin-producing β-cells. Accumulating evidence recently indicated that, following the reprogramming, epigenetic memory inherited from parental cells substantially affects the differentiation capacity of many iPSC lines. Therefore, differences in epigenetic signature are likely to be essential contributing factors influencing the propensity of iPSC differentiation. In this review, we will document the impact of the epigenome on the reprogramming efficacy and differentiation potential of iPSCs and how targeting the epigenetic residual memory could be an additional strategy to improve the differentiation efficiency of existing protocols to generate fully functional hPSC-derived pancreatic β-cells for diabetes therapy and drug screening.
Collapse
Affiliation(s)
- Abdoulaye Diane
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation (QF), Hamad Bin Khalifa University (HBKU), Doha, Qatar.
| | - Razik Bin Abdul Mu-U-Min
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation (QF), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Heba Hussain Al-Siddiqi
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Qatar Foundation (QF), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
10
|
Mirizio G, Sampson S, Iwafuchi M. Interplay between pioneer transcription factors and epigenetic modifiers in cell reprogramming. Regen Ther 2025; 28:246-252. [PMID: 39834592 PMCID: PMC11745816 DOI: 10.1016/j.reth.2024.12.014] [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: 10/31/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025] Open
Abstract
The generation of induced pluripotent stem cells (iPSCs) from differentiated somatic cells by Yamanaka factors, including pioneer transcription factors (TFs), has greatly reshaped our traditional understanding of cell plasticity and demonstrated the remarkable potential of pioneer TFs. In addition to iPSC reprogramming, pioneer TFs are pivotal in direct reprogramming or transdifferentiation where somatic cells are converted into different cell types without passing through a pluripotent state. Pioneer TFs initiate a reprogramming process through chromatin opening, thereby establishing competence for new gene regulatory programs. The action of pioneer TFs is both influenced by and exerts influence on epigenetic regulation. Despite significant advances, many direct reprogramming processes remain inefficient, which limits their reliability for clinical applications. In this review, we discuss the molecular mechanisms underlying pioneer TF-driven reprogramming, with a focus on their interactions with epigenetic modifiers, including Polycomb repressive complexes (PRCs), nucleosome remodeling and deacetylase (NuRD) complexes, and the DNA methylation machinery. A deeper understanding of the dynamic interplay between pioneer TFs and epigenetic modifiers will be essential for advancing reprogramming technologies and unlocking their full clinical potential.
Collapse
Affiliation(s)
- Gerardo Mirizio
- Division of Developmental Biology, Center for Stem Cell & Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, OH, 45229, USA
| | - Samuel Sampson
- Division of Developmental Biology, Center for Stem Cell & Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, OH, 45229, USA
| | - Makiko Iwafuchi
- Division of Developmental Biology, Center for Stem Cell & Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, OH, 45229, USA
| |
Collapse
|
11
|
Rouzbahani M, Ghanaati H. Intra-Arterial Stem Cell Injection for Treating Various Diseases: A New Frontier in Interventional Radiology. Cardiovasc Intervent Radiol 2025; 48:288-296. [PMID: 39789253 DOI: 10.1007/s00270-024-03947-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025]
Abstract
This article provides radiologists with insights into stem cells' functions, sources, and potentially successful clinical treatments via intravascular injection in organs such as the liver, kidney, pancreas, musculoskeletal system, and for ischemic conditions affecting the brain, heart and limbs. Understanding stem cells' significance in interventional radiology and its limitations enables tailored interventions for diverse conditions, ensuring efficient medical care and optimal treatment selection.
Collapse
Affiliation(s)
- Maedeh Rouzbahani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Ghanaati
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Gantier M, Ménoret S, Fourrier A, Delbos F, Nguyen TH, Anegon I. Human pluripotent stem cell-derived hepatic progenitors exhibit a partially hypoimmunogenic phenotype and actively inhibit immune responses. Front Immunol 2025; 16:1507317. [PMID: 40070824 PMCID: PMC11893836 DOI: 10.3389/fimmu.2025.1507317] [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: 10/07/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction GStemHep cells are human cryopreserved hepatic progenitors derived from pluripotent of stem cells (GStem cells) using a cGMP-compliant protocol. They were highly effective in rescuing mice from acute liver failure. Methods The objective of this study was to analyze the immunogenicity and immunoregulatory properties of GStemHep cells. Results As compared to GStem cells, GStemHep cells showed complete loss of HLA-I (ABC) and they lacked of expression of HLA-II, HLA-G, HLA-E and PD-L1. GStemHep cells also showed increased expression of CD47, maintained high expression of indoleamine 2,3-dioxygenase (IDO) and heme oxygenase-1 (HO-1) and reduced expression of CD200. In comparison with GStem cells, GStemHep cultured in inflammatory conditions increased the expression of PD-L1, CD200, HO-1, HLA-E, CD47 and HLA-I (ABC) as well as maintained expression of IDO and were negative for HLA-II and HLA-G. GStemHep culture in basal or inflammatory conditions has a low or absent immunogenic activity on T cells, associated to a suppressive effect on proliferation partially mediated by IDO. We observed phagocytosis of GStemHep by macrophages that was partially inhibited by CD47 expression. NK cells were activated by resting GStemHep cells. Upon culture in inflammatory conditions that induced expression of HLA-I molecules in GStemHep cells NK cell activation was reduced. Thus, GStemHep cells are partially hypoimmune cells due to the expression of several immune checkpoint inhibitors and the absence of HLA-I molecules. In inflammatory conditions, the expression of several of these molecules was increased but also of HLA-I that could be immunogenic for T cells but it was inhibitory for NK cells. Discussion GStemHep cells show a favorable immunological profile for their use as allogeneic off-the shelf treatment of liver diseases with loss of hepatocyte function.
Collapse
Affiliation(s)
| | - Séverine Ménoret
- Nantes Université, Inserm, CNRS, SFR Santé, Inserm UMS 016 CNRS UMS 3556, Nantes, France
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | | | | | | | - Ignacio Anegon
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| |
Collapse
|
13
|
Guerra-Resendez RS, Lydon SL, Ma AJ, Bedford GC, Reed DR, Kim S, Terán ER, Nishiguchi T, Escobar M, DiNardo AR, Hilton IB. Characterization of Rationally Designed CRISPR/Cas9-Based DNA Methyltransferases with Distinct Methyltransferase and Gene Silencing Activities in Human Cell Lines and Primary Human T Cells. ACS Synth Biol 2025; 14:384-397. [PMID: 39898483 PMCID: PMC11854388 DOI: 10.1021/acssynbio.4c00569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 02/04/2025]
Abstract
Nuclease-deactivated Cas (dCas) proteins can be used to recruit epigenetic effectors, and this class of epigenetic editing technologies has revolutionized the ability to synthetically control the mammalian epigenome and transcriptome. DNA methylation is one of the most important and well-characterized epigenetic modifications in mammals, and while many different forms of dCas-based DNA methyltransferases (dCas-DNMTs) have been developed for programmable DNA methylation, these tools are frequently poorly tolerated and/or lowly expressed in mammalian cell types. Further, the use of dCas-DNMTs has largely been restricted to cell lines, which limits mechanistic insights in karyotypically normal contexts and hampers translational utility in the longer term. Here, we extend previous insights into the rational design of the catalytic core of the mammalian DNMT3A methyltransferase and test three dCas9-DNMT3A/3L variants across different human cell lines and in primary donor-derived human T cells. We find that mutations within the catalytic core of DNMT3A stabilize the expression of dCas9-DNMT3A/3L fusion proteins in Jurkat T cells without sacrificing DNA methylation or gene-silencing performance. We also show that these rationally engineered mutations in DNMT3A alter DNA methylation profiles at loci targeted with dCas9-DNMT3A/3L in cell lines and donor-derived human T cells. Finally, we leverage the transcriptionally repressive effects of dCas9-DNMT3A/3L variants to functionally link the expression of a key immunomodulatory transcription factor to cytokine secretion in donor-derived T cells. Overall, our work expands the synthetic biology toolkit for epigenetic editing and provides a roadmap for the use of engineered dCas-based DNMTs in primary mammalian cell types.
Collapse
Affiliation(s)
| | | | - Alex J. Ma
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Guy C. Bedford
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Daniel R. Reed
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Sunghwan Kim
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Erik R. Terán
- Department
of BioSciences, Rice University, Houston, Texas 77005, United States
| | - Tomoki Nishiguchi
- Global
Tuberculosis Program, Texas Children’s Hospital, Immigrant
and Global Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Mario Escobar
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Andrew R. DiNardo
- Global
Tuberculosis Program, Texas Children’s Hospital, Immigrant
and Global Health, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Isaac B. Hilton
- Systems,
Synthetic, and Physical Biology Program, Rice University, Houston, Texas 77005, United States
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department
of BioSciences, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
14
|
Kato Y, Inaba T, Shinke K, Hiramatsu N, Horie T, Sakamoto T, Hata Y, Sugihara E, Takimoto T, Nagai N, Ishigaki Y, Kojima H, Nagano O, Yamamoto N, Saya H. Comprehensive Search for Genes Involved in Thalidomide Teratogenicity Using Early Differentiation Models of Human Induced Pluripotent Stem Cells: Potential Applications in Reproductive and Developmental Toxicity Testing. Cells 2025; 14:215. [PMID: 39937006 PMCID: PMC11817626 DOI: 10.3390/cells14030215] [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: 01/06/2025] [Revised: 01/26/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Developmental toxicity testing is essential to identify substances that may harm embryonic development. This study aimed to establish a protocol for evaluating developmental toxicity using human induced pluripotent stem cells (iPSCs) by analyzing cellular activity and gene expression changes. Two ICH S5(R3) positive substances, valproic acid (VPA), which is a substance previously detected as positive by other test methods, and thalidomide (Thalido), were examined during early trichoderm differentiation without fetal bovine serum. RNA-seq analysis identified seven candidate genes, including TP63, associated with altered expression following exposure to VPA or Thalido. These genes were implicated in pathways related to tissue development, cell growth, and molecular interactions. While the assay effectively detected VPA and Thalido, its limitations include testing only soluble substances and focusing on early differentiation stages. Nevertheless, the protocol demonstrates potential for the classification and evaluation of emerging modality drugs based on physical properties such as solubility, polarity, and pH. Integration with AI analysis may enhance its capacity to uncover genetic variations and evaluate previously uncharacterized substances. This study provides a foundation for alternative developmental toxicity testing methods, with further refinements in the culture method expected to improve accuracy and applicability in regulatory toxicology.
Collapse
Affiliation(s)
- Yu Kato
- Oncology Innovation Center, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (Y.K.); (E.S.); (T.T.); (O.N.); (H.S.)
- Center for Society-Academia Collaboration, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| | - Takeshi Inaba
- Graduate School of Health Sciences, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (T.I.); (K.S.)
| | - Koudai Shinke
- Graduate School of Health Sciences, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (T.I.); (K.S.)
| | - Noriko Hiramatsu
- Center for Society-Academia Collaboration, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
- Clinical Laboratory, Fujita Health University Hospital, Toyoake 470-1192, Aichi, Japan
| | - Tetsuhiro Horie
- Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan; (T.H.); (T.S.); (Y.I.)
- Department of Pharmacy, Kanazawa Medical University Hospital, Uchinada 920-0293, Ishikawa, Japan
| | - Takuya Sakamoto
- Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan; (T.H.); (T.S.); (Y.I.)
- Department of Pharmacy, Kanazawa Medical University Hospital, Uchinada 920-0293, Ishikawa, Japan
| | - Yuko Hata
- Open Facility Center, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| | - Eiji Sugihara
- Oncology Innovation Center, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (Y.K.); (E.S.); (T.T.); (O.N.); (H.S.)
- Open Facility Center, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
| | - Tetsuya Takimoto
- Oncology Innovation Center, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (Y.K.); (E.S.); (T.T.); (O.N.); (H.S.)
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, Higashiosaka 577-8502, Osaka, Japan;
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Ishikawa, Japan; (T.H.); (T.S.); (Y.I.)
| | - Hajime Kojima
- Department of Pharmaceutical Engineering, Faculty of Engineering, Sanyo-Onoda City University, Sanyo-Onoda 756-0884, Yamaguchi, Japan;
- National Institute of Health Sciences (NIHS), Kawasaki 210-9501, Kanagawa, Japan
| | - Osamu Nagano
- Oncology Innovation Center, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (Y.K.); (E.S.); (T.T.); (O.N.); (H.S.)
| | - Naoki Yamamoto
- Center for Society-Academia Collaboration, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan;
- Graduate School of Health Sciences, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (T.I.); (K.S.)
- International Center for Cell and Gene Therapy, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Hideyuki Saya
- Oncology Innovation Center, Research Promotion Headquarters, Fujita Health University, Toyoake 470-1192, Aichi, Japan; (Y.K.); (E.S.); (T.T.); (O.N.); (H.S.)
| |
Collapse
|
15
|
Simpson HL, Smits E, Moerkens R, Wijmenga C, Mooiweer J, Jonkers IH, Withoff S. Human organoids and organ-on-chips in coeliac disease research. Trends Mol Med 2025; 31:117-137. [PMID: 39448329 DOI: 10.1016/j.molmed.2024.10.003] [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] [Received: 07/12/2024] [Revised: 09/17/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Coeliac disease (CeD) is an immune-mediated disorder characterised by gluten-triggered inflammation and damage in the small intestine, with lifelong gluten-free diet (GFD) as the only treatment. It is a multifactorial disease, involving genetic and environmental susceptibility factors, and its complexity and lack of comprehensive human model systems have hindered understanding of its pathogenesis and development of new treatments. Therefore, it is crucial to establish systems that recapitulate patient genetic background and the interactions between the small intestinal epithelial barrier, immune cells, and environment that contribute to CeD. In this review, we discuss disease complexity, recent advances in stem cell biology, organoids, tissue co-cultures, and organ-on-chip (OoC) systems that facilitate the development of comprehensive human model systems, and model applications in preclinical studies of potential treatments.
Collapse
Affiliation(s)
- Hanna L Simpson
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Eline Smits
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Renée Moerkens
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Joram Mooiweer
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Iris H Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
16
|
Peng D, Li M, Yu Z, Yan T, Yao M, Li S, Liu Z, Li L, Qiu H. Synergy between pluripotent stem cell-derived macrophages and self-renewing macrophages: Envisioning a promising avenue for the modelling and cell therapy of infectious diseases. Cell Prolif 2025; 58:e13770. [PMID: 39537185 PMCID: PMC11839195 DOI: 10.1111/cpr.13770] [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: 06/28/2024] [Revised: 09/30/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
As crucial phagocytes of the innate immune system, macrophages (Mϕs) protect mammalian hosts, maintain tissue homeostasis and influence disease pathogenesis. Nonetheless, Mϕs are susceptible to various pathogens, including bacteria, viruses and parasites, which cause various infectious diseases, necessitating a deeper understanding of pathogen-Mϕ interactions and therapeutic insights. Pluripotent stem cells (PSCs) have been efficiently differentiated into PSC-derived Mϕs (PSCdMϕs) resembling primary Mϕs, advancing the modelling and cell therapy of infectious diseases. However, the mass production of PSCdMϕs, which lack proliferative capacity, relies on large-scale expansions of PSCs, thereby increasing both costs and culture cycles. Notably, Mϕs deficient in the MafB/c-Maf genes have been reported to re-enter the cell cycle with the stimulation of specific growth factor cocktails, turning into self-renewing Mϕs (SRMϕs). This review summarizes the applications of PSCdMϕs in the modelling and cell therapy of infectious diseases and strategies for establishing SRMϕs. Most importantly, we innovatively propose that PSCs can serve as a gene editing platform to creating PSC-derived SRMϕs (termed PSRMϕs), addressing the resistance of Mϕs against genetic manipulation. We discuss the challenges and possible solutions in creating PSRMϕs. In conclusion, this review provides novel insights into the development of physiologically relevant and expandable Mϕ models, highlighting the enormous potential of PSRMϕs as a promising avenue for the modelling and cell therapy of infectious diseases.
Collapse
Affiliation(s)
- Dingkun Peng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Meilin Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Zhuoran Yu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life ScienceNortheast Agricultural UniversityHarbinChina
| | - Tingsheng Yan
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life ScienceNortheast Agricultural UniversityHarbinChina
| | - Meng Yao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Zhonghua Liu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life ScienceNortheast Agricultural UniversityHarbinChina
| | - Lian‐Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| | - Hua‐Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research InstituteChinese Academy of Agricultural SciencesHarbinChina
| |
Collapse
|
17
|
Chukwuemeka CG, Ndubueze CW, Kolawole AV, Joseph JN, Oladipo IH, Ofoezie EF, Annor-Yeboah SA, Bello ARE, Ganiyu SO. In vitro erythropoiesis: the emerging potential of induced pluripotent stem cells (iPSCs). BLOOD SCIENCE 2025; 7:e00215. [PMID: 39726795 PMCID: PMC11671056 DOI: 10.1097/bs9.0000000000000215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Due to global blood shortages and restricted donor blood storage, the focus has switched to the in vitro synthesis of red blood cells (RBCs) from induced pluripotent stem cells (iPSCs) as a potential solution. Many processes are required to synthesize RBCs from iPSCs, including the production of iPSCs from human or animal cells, differentiation of iPSCs into hematopoietic stem cells, culturing, and maturation of the hematopoietic stem cells (HSC) to make functional erythrocytes. Previous investigations on the in vitro production of erythrocytes have shown conflicting results. Some studies have demonstrated substantial yields of functional erythrocytes, whereas others have observed low yields of enucleated cells. Before large-scale in vitro RBC production can be achieved, several challenges which have limited its application in the clinic must be overcome. These issues include optimizing differentiation techniques to manufacture vast amounts of functional RBCs, upscaling the manufacturing process, cost-effectiveness, and assuring the production of RBCs with good manufacturing practices (GMP) before they can be used for therapeutic purposes.
Collapse
Affiliation(s)
| | - Chizaram W. Ndubueze
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | - Adeola V. Kolawole
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | - Joshua N. Joseph
- College of Science, University of Massey, Tennent Drive, Massey University, Palmerston North 4410, New Zealand
- Resilient Agriculture, AgResearch Limited, Grasslands Research Centre Tennent Drive, Fitzherbert Palmerston North 4410, New Zealand
| | - Ifeoluwa H. Oladipo
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | - Ezichi F. Ofoezie
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | | | - Abdur-Rahman Eneye Bello
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
- Department of Biochemistry, Confluence University of Science and Technology, Osara, Kogi State, Nigeria
| | - Sodiq O. Ganiyu
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| |
Collapse
|
18
|
Jagadale S, Damle M, Joshi MG. Bone Tissue Engineering: From Biomaterials to Clinical Trials. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:73-115. [PMID: 39881051 DOI: 10.1007/5584_2024_841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Bone tissue engineering is a promising field that aims to rebuild the bone tissue using biomaterials, cells, and signaling molecules. Materials like natural and synthetic polymers, inorganic materials, and composite materials are used to create scaffolds that mimic the hierarchical microstructure of bone. Stem cells, particularly mesenchymal stem cells (MSCs), play a crucial role in bone tissue engineering by promoting tissue regeneration and modulating the immune response. Growth factors like bone morphogenetic proteins (BMPs), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) are utilized to accelerate bone regeneration. Clinical applications include treating nonunion and mal-union fractures, osteonecrosis, orthopedic surgery, dental applications, and spinal cord injuries. Recent advances in the field include nanotechnology, 3D printing, bioprinting techniques, gene editing technologies, and microfluidic devices for drug testing. However, challenges remain, such as standardization of protocols, large-scale biomaterial production, personalized medicine approaches, cost-effectiveness, and regulatory issues. Current clinical trials are investigating the safety and efficacy of various bone tissue engineering approaches, with the potential to modernize patient care by providing more adequate treatments for bone defects and injuries.
Collapse
Affiliation(s)
- Swapnali Jagadale
- Department of Stem Cells & Regenerative Medicine, Centre for Interdisciplinary Research, D Y Patil Education Society (Deemed to be University), Kolhapur, India
| | - Mrunal Damle
- Department of Stem Cells & Regenerative Medicine, Centre for Interdisciplinary Research, D Y Patil Education Society (Deemed to be University), Kolhapur, India
| | - Meghnad G Joshi
- Department of Stem Cells & Regenerative Medicine, Centre for Interdisciplinary Research, D Y Patil Education Society (Deemed to be University), Kolhapur, India.
- Stem Plus Biotech, Sangli, India.
| |
Collapse
|
19
|
Hameed SS, Sharma TP. Generation of Retinal Ganglion Cells from Reprogrammed Keratocytes of Non-Glaucoma and Glaucoma Donors. Curr Protoc 2025; 5:e70091. [PMID: 39781605 PMCID: PMC11713219 DOI: 10.1002/cpz1.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Human induced pluripotent stem cell (hiPSC)-based disease modeling can be successfully recapitulated to mimic disease characteristics across various human pathologies. Glaucoma, a progressive optic neuropathy, primarily affects the retinal ganglion cells (RGCs). While multiple groups have successfully generated RGCs from non-diseased hiPSCs, producing RGCs from glaucomatous human samples holds significant promise for understanding disease pathology by revealing patient-specific disease signatures. Given that keratocytes originate from the neural crest and previous reports suggest that ocular fibroblasts from glaucomatous donors carry pathogenic signatures, it is highly plausible that these signatures imprinted within the keratocytes will also be present in the derived RGCs. Thus, we aimed to generate RGCs from both glaucomatous and non-glaucomatous donor keratocytes and validate disease-specific signatures in 3D retinal organoids and in isolated RGCs. Our protocol describes the generation of iPSCs from keratocytes of both glaucomatous and non-glaucomatous donors, followed by their differentiation into retinal organoids. Subsequent isolation and culturing of RGCs were performed. Disease signatures in the RGCs were validated in both 3D retinal organoids (ROs) and 2D RGC cultures, and glaucomatous RGCs in 3D and 2D cultures demonstrated increased cleaved CASP3 and significant RGC loss, indicating disease imprints in the hiPSC-derived RGCs. This model offers a venue and high throughput platform for studying glaucomatous disease pathology and holds significant potential for drug discovery using RGCs derived from human donors. © 2025 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Culturing of keratocytes from human cadaveric donors Basic Protocol 2: Reprogramming donor keratocytes into iPSCs Basic Protocol 3: Evaluation of chromosomal loss during reprogramming in iPSCs by karyotyping Basic Protocol 4: Generation of 3D ROs Basic Protocol 5: Dissociation and culturing of RGCs from 3D ROs Support Protocol 1: Immunostaining for phenotypic characterization of cells Support Protocol 2: Sectioning of 3D ROs and immunostaining Support Protocol 3: Western blotting for cleaved CASP3 and THY1.
Collapse
Affiliation(s)
- Shahna S. Hameed
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndiana
| | - Tasneem P. Sharma
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndiana
| |
Collapse
|
20
|
Mishra P, Biesiada I, Gupta P, Ghavami S, Markowski J, Łos MJ. Unraveling the Complexity and Advancements of Transdifferentiation Technologies in the Biomedical Field and Their Potential Clinical Relevance. Arch Immunol Ther Exp (Warsz) 2025; 73:aite-2025-0001. [PMID: 39637369 DOI: 10.2478/aite-2025-0001] [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: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Chronic diseases such as cancer, autoimmunity, and organ failure currently depend on conventional pharmaceutical treatment, which may cause detrimental side effects in the long term. In this regard, cell-based therapy has emerged as a suitable alternative for treating these chronic diseases. Transdifferentiation technologies have evolved as a suitable therapeutic alternative that converts one differentiated somatic cell into another phenotype by using transcription factors (TFs), small molecules, or small, single-stranded, non-coding RNA molecules (miRNA). The transdifferentiation techniques rely on simple, fast, standardized, and versatile protocols with minimal chance of tumorigenicity and genotoxicity. However, there are still challenges and limitations that need to be addressed to enhance their clinical translation percentage in the near future. Taking this into account, we have delineated the features and strategies used in the transdifferentiation techniques. Then, we delved into different intermediate states that were attained during transdifferentiation. Advancements in transdifferentiation techniques in the field of tissue engineering, autoimmunity, and cancer therapy were dissected. Furthermore, limitations, challenges, and future perspectives are outlined in this review to provide a whole new picture of the transdifferentiation techniques. Advancements in molecular biology, interdisciplinary research, bioinformatics, and artificial intelligence will push the frontiers of this technology further to establish new avenues for biomedical research.
Collapse
Affiliation(s)
- Purusottam Mishra
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Izabella Biesiada
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Payal Gupta
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, India
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, Zabrze, Poland
| | - Jarosław Markowski
- Department of Laryngology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, Gliwice, Poland
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
21
|
Bell CC, Faulkner GJ, Gilan O. Chromatin-based memory as a self-stabilizing influence on cell identity. Genome Biol 2024; 25:320. [PMID: 39736786 DOI: 10.1186/s13059-024-03461-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/16/2024] [Indexed: 01/01/2025] Open
Abstract
Cell types are traditionally thought to be specified and stabilized by gene regulatory networks. Here, we explore how chromatin memory contributes to the specification and stabilization of cell states. Through pervasive, local, feedback loops, chromatin memory enables cell states that were initially unstable to become stable. Deeper appreciation of this self-stabilizing role for chromatin broadens our perspective of Waddington's epigenetic landscape from a static surface with islands of stability shaped by evolution, to a plasticine surface molded by experience. With implications for the evolution of cell types, stabilization of resistant states in cancer, and the widespread plasticity of complex life.
Collapse
Affiliation(s)
- Charles C Bell
- Mater Research Institute, University of Queensland, TRI Building, Woolloongabba, QLD, 4102, Australia.
| | - Geoffrey J Faulkner
- Mater Research Institute, University of Queensland, TRI Building, Woolloongabba, QLD, 4102, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD, 4169, Australia
| | - Omer Gilan
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, 3004, Australia
| |
Collapse
|
22
|
Sgromo C, Cucci A, Venturin G, Follenzi A, Olgasi C. Bridging the Gap: Endothelial Dysfunction and the Role of iPSC-Derived Endothelial Cells in Disease Modeling. Int J Mol Sci 2024; 25:13275. [PMID: 39769040 PMCID: PMC11678083 DOI: 10.3390/ijms252413275] [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: 11/18/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Endothelial cells (ECs) are crucial for vascular health, regulating blood flow, nutrient exchange, and modulating immune responses and inflammation. The impairment of these processes causes the endothelial dysfunction (ED) characterized by oxidative stress, inflammation, vascular permeability, and extracellular matrix remodeling. While primary ECs have been widely used to study ED in vitro, their limitations-such as short lifespan and donor variability-pose challenges. In this context, induced iECs derived from induced pluripotent stem cells offer an innovative solution, providing an unlimited source of ECs to explore disease-specific features of ED. Recent advancements in 3D models and microfluidic systems have enhanced the physiological relevance of iEC-based models by better mimicking the vascular microenvironment. These innovations bridge the gap between understanding ED mechanisms and drug developing and screening to prevent or treat ED. This review highlights the current state of iEC technology as a model to study ED in vascular and non-vascular disorders, including diabetes, cardiovascular, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Chiara Sgromo
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (C.S.); (A.C.); (G.V.)
| | - Alessia Cucci
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (C.S.); (A.C.); (G.V.)
| | - Giorgia Venturin
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (C.S.); (A.C.); (G.V.)
| | - Antonia Follenzi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy; (C.S.); (A.C.); (G.V.)
| | - Cristina Olgasi
- Department of Translational Medicine, School of Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
| |
Collapse
|
23
|
Roslan FF, Yu Y, Wang M, Mohd Yusof NAN, Ooi GC, Then KL, Then KY, Cheong SK, Ab Patar MNA, Tan JJ. Reprogramming of Expanded Cord Blood-Derived CD34 + Cells from Umbilical Cord-Mesenchymal Stromal Cell Co-Culture to Generate Human-Induced Pluripotent Stem Cells. Cell Reprogram 2024; 26:164-176. [PMID: 39602209 DOI: 10.1089/cell.2024.0073] [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: 11/29/2024] Open
Abstract
Cord blood (CB) is widely stored as a source of hematopoietic stem cells for potential future use, though its application for autologous purposes remains limited. Repurposing CB into human-induced pluripotent stem cells (hiPSCs) can broaden its utility beyond hematological conditions. This study investigated the effects of umbilical cord-mesenchymal stromal cell (UC-MSC) co-culture on CB CD34+ cells and the characteristics of the resulting hiPSCs. CD34+ cells were isolated, expanded in UC-MSC co-culture for 3 days, and reprogrammed into hiPSCs using episomal vectors. Results showed that UC-MSC co-culture significantly increased CD34+ cell numbers (p < 0.0001, n = 6), with a reduced population doubling time of 25.1 ± 2.1 hours compared with the control (p < 0.0004, n = 6). The yield of CD34+ cells was substantially higher in the UC-MSC co-culture group. The hiPSCs exhibited comparable reprogramming efficiency, pluripotency marker expression, trilineage differentiation potential, and genomic stability to CD34+ cells expanded under standard culture conditions. These findings suggest that CD34+ cells from CB, expanded in UC-MSC co-culture, can be reprogrammed into functional hiPSCs without compromising cell quality or genetic stability.
Collapse
Affiliation(s)
- Fatin Fazrina Roslan
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Yuexin Yu
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan Zhengzhou, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Henan Xinxiang, China
| | - Mengmeng Wang
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Henan, China
- The International-Joint Lab for Non-invasive Neural Modulation/Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China
| | - Nurul Ain Nasim Mohd Yusof
- Pluripotent Stem Cell Laboratory, Hematology Unit, Cancer Research Centre, Institute for Medical Research (IMR), Block C, National Institute of Health (NIH) Malaysia, Setia Alam, Malaysia
| | - Ghee Chien Ooi
- Cryocord Sdn Bhd, Bio-X Centre, Persiaran Cyber Point Selatan Cyberjaya, Cyberjaya, Malaysia
| | - Khong Lek Then
- Cryocord Sdn Bhd, Bio-X Centre, Persiaran Cyber Point Selatan Cyberjaya, Cyberjaya, Malaysia
| | - Kong Yong Then
- Cryocord Sdn Bhd, Bio-X Centre, Persiaran Cyber Point Selatan Cyberjaya, Cyberjaya, Malaysia
| | - Soon-Keng Cheong
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman (UTAR), Kajang, Malaysia
| | | | - Jun Jie Tan
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
24
|
Begovic M, Schneider L, Zhou X, Hamdani N, Akin I, El-Battrawy I. The Role of Human-Induced Pluripotent Stem Cells in Studying Cardiac Channelopathies. Int J Mol Sci 2024; 25:12034. [PMID: 39596103 PMCID: PMC11593457 DOI: 10.3390/ijms252212034] [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: 08/23/2024] [Revised: 10/22/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiac channelopathies are inherited diseases that increase the risk of sudden cardiac death. While different genes have been associated with inherited channelopathies, there are still subtypes, e.g., catecholaminergic polymorphic ventricular tachycardia and Brugada syndrome, where the genetic cause remains unknown. Various models, including animal models, heterologous expression systems, and the human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSCs-CMs) model, have been used to study the pathophysiological mechanisms of channelopathies. Recently, researchers have focused on using hiPSCs-CMs to understand the genotype-phenotype correlation and screen drugs. By combining innovative techniques such as Clustered Regularly Interspaced Short Palindromic Repeats/Clustered Regularly Interspaced Short Palindromic Repeats associated protein 9 (CRISPR/Cas9)-mediated genome editing, and three-dimensional (3D) engineered heart tissues, we can gain new insights into the pathophysiological mechanisms of channelopathies. This approach holds promise for improving personalized drug treatment. This review highlights the role of hiPSCs-CMs in understanding the pathomechanism of Brugada syndrome and catecholaminergic polymorphic ventricular tachycardia and how these models can be utilized for drug screening.
Collapse
Affiliation(s)
- Merima Begovic
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Luca Schneider
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Xiaobo Zhou
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Nazha Hamdani
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Physiology, Cardiovascular Research Institute, University Maastricht, 6229HX Maastricht, The Netherlands
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, 1089 Budapest, Hungary
- Department of Cardiology and Rhythmology, St. Josef Hospital, Ruhr University, 44791 Bochum, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Ibrahim El-Battrawy
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Cardiology and Rhythmology, St. Josef Hospital, Ruhr University, 44791 Bochum, Germany
| |
Collapse
|
25
|
Morihara H, Yokoe S, Wakabayashi S, Takai S. TMEM182 inhibits myocardial differentiation of human iPS cells by maintaining the activated state of Wnt/β-catenin signaling through an increase in ILK expression. FASEB Bioadv 2024; 6:565-579. [PMID: 39512841 PMCID: PMC11539028 DOI: 10.1096/fba.2024-00086] [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: 06/06/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024] Open
Abstract
Transmembrane protein 182 (TMEM182) is notably abundant in muscle and adipose tissue, but its role in the heart remains unknown. This study examined the contribution of TMEM182 in the differentiation of human induced pluripotent stem cells (hiPSCs) into cardiomyocytes. For this, we generated hiPSCs overexpressing TMEM182 in a doxycycline-inducible manner and induced their differentiation into cardiomyocytes. On Day 12 of differentiation, expression of the cardiomyocyte markers, TNNT2 and MYH6, was significantly decreased in TMEM182-overexpressing cells. Additionally, we found that phosphorylation of GSK-3β (Ser9) and β-catenin (Ser552) was increased during TMEM182 overexpression, suggesting activation of Wnt/β-catenin signaling. We further focused on integrin-linked kinase (ILK) as the mechanism by which TMEM182 activates Wnt/β-catenin signaling. Evaluation showed that ILK expression was increased in cells overexpressing TMEM182. These results suggest that TMEM182 maintains Wnt/β-catenin signaling in an activated state after mesoderm formation by increasing ILK expression, thereby suppressing hiPSCs differentiation into cardiomyocytes.
Collapse
Affiliation(s)
- Hirofumi Morihara
- Department of Pharmacology, Faculty of MedicineOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Shunichi Yokoe
- Department of Pharmacology, Faculty of MedicineOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| | - Shigeo Wakabayashi
- Department of Pharmacology, Faculty of MedicineOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
- Department of Nursing, Faculty of Health SciencesOsaka Aoyama UniversityMinohJapan
| | - Shinji Takai
- Department of Pharmacology, Faculty of MedicineOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
- Department of Innovative Medicine, Graduate School of MedicineOsaka Medical and Pharmaceutical UniversityTakatsukiJapan
| |
Collapse
|
26
|
Gao H, Pathan S, Dixon BREA, Pugazenthi A, Mathison M, Mohamed TMA, Rosengart TK, Yang J. Sall4 and Gata4 induce cardiac fibroblast transition towards a partially multipotent state with cardiogenic potential. Sci Rep 2024; 14:24182. [PMID: 39406776 PMCID: PMC11480346 DOI: 10.1038/s41598-024-73975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/23/2024] [Indexed: 10/19/2024] Open
Abstract
Cardiac cellular fate transition holds remarkable promise for the treatment of ischemic heart disease. We report that overexpressing two transcription factors, Sall4 and Gata4, which play distinct and overlapping roles in both pluripotent stem cell reprogramming and embryonic heart development, induces a fraction of stem-like cells in rodent cardiac fibroblasts that exhibit unlimited ex vivo expandability with clonogenicity. Transcriptomic and phenotypic analyses reveal that around 32 ± 6.4% of the expanding cells express Nkx2.5, while 13 ± 3.6% express Oct4. Activated signaling pathways like PI3K/Akt, Hippo, Wnt, and multiple epigenetic modification enzymes are also detected. Under suitable conditions, these cells demonstrate a high susceptibility to differentiating into cardiomyocyte, endothelial cell, and extracardiac neuron-like cells. The presence of partially pluripotent-like cells is characterized by alkaline phosphatase staining, germ layer marker expression, and tumor formation in injected mice (n = 5). Additionally, significant stem-like fate transitions and cardiogenic abilities are induced in human cardiac fibroblasts, but not in rat or human skin fibroblasts. Molecularly, we identify that SALL4 and GATA4 physically interact and synergistically stimulate the promoters of pluripotency genes but repress fibrogenic gene, which correlates with a primitive transition process. Together, this study uncovers a new cardiac regenerative mechanism that could potentially advance therapeutic endeavors and tissue engineering.
Collapse
Affiliation(s)
- Hong Gao
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Saliha Pathan
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Beverly R E A Dixon
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Aarthi Pugazenthi
- Department of Pathology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Tamer M A Mohamed
- Department of Surgery, Texas Heart Institute, 6519 Fannin Street, Houston, TX, 77030, USA
| | - Todd K Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Jianchang Yang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
Hajmousa G, de Almeida RC, Bloks N, Ruiz AR, Bouma M, Slieker R, Kuipers TB, Nelissen RGHH, Ito K, Freund C, Ramos YFM, Meulenbelt I. The role of DNA methylation in chondrogenesis of human iPSCs as a stable marker of cartilage quality. Clin Epigenetics 2024; 16:141. [PMID: 39407288 PMCID: PMC11481477 DOI: 10.1186/s13148-024-01759-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Lack of insight into factors that determine purity and quality of human iPSC (hiPSC)-derived neo-cartilage precludes applications of this powerful technology toward regenerative solutions in the clinical setting. Here, we set out to generate methylome-wide landscapes of hiPSC-derived neo-cartilages from different tissues-of-origin and integrated transcriptome-wide data to identify dissimilarities in set points of methylation with associated transcription and the respective pathways in which these genes act. METHODS We applied in vitro chondrogenesis using hiPSCs generated from two different tissue sources: skin fibroblasts and articular cartilage. Upon differentiation toward chondrocytes, these are referred to as hFiCs and hCiC, respectively. Genome-wide DNA methylation and RNA sequencing datasets were generated of the hiPSC-derived neo-cartilages, and the epigenetically regulated transcriptome was compared to that of neo-cartilage deposited by human primary articular cartilage (hPAC). RESULTS Methylome-wide landscapes of neo-cartilages of hiPSCs reprogrammed from two different somatic tissues were 85% similar to that of hPACs. By integration of transcriptome-wide data, differences in transcriptionally active CpGs between hCiC relative to hPAC were prioritized. Among the CpG-gene pairs lower expressed in hCiCs relative to hPACs, we identified genes such as MGP, GDF5, and CHAD enriched in closely related pathways and involved in cartilage development that likely mark phenotypic differences in chondrocyte states. Vice versa, among the CpG-gene pairs higher expressed, we identified genes such as KIF1A or NKX2-2 enriched in neurogenic pathways and likely reflecting off target differentiation. CONCLUSIONS We did not find significant variation between the neo-cartilages derived from hiPSCs of different tissue sources, suggesting that application of a robust differentiation protocol such as we applied here is more important as compared to the epigenetic memory of the cells of origin. Results of our study could be further exploited to improve quality, purity, and maturity of hiPSC-derived neo-cartilage matrix, ultimately to realize introduction of sustainable, hiPSC-derived neo-cartilage implantation into clinical practice.
Collapse
Affiliation(s)
- Ghazaleh Hajmousa
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Niek Bloks
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Alejandro Rodríguez Ruiz
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Marga Bouma
- Department of Anatomy and Embryology and Human iPSC Hotel, 2333 ZA, Leiden, The Netherlands
| | - Roderick Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas B Kuipers
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rob G H H Nelissen
- Department of Orthopedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Christian Freund
- Department of Anatomy and Embryology and Human iPSC Hotel, 2333 ZA, Leiden, The Netherlands
| | - Yolande F M Ramos
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Post-zone S-05-P, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
28
|
Cao D, Garai S, DiFrisco J, Veenvliet JV. The logic of monsters: development and morphological diversity in stem-cell-based embryo models. Interface Focus 2024; 14:20240023. [PMID: 39464644 PMCID: PMC11503023 DOI: 10.1098/rsfs.2024.0023] [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: 07/18/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/29/2024] Open
Abstract
Organoids and stem-cell-based embryo models (SEMs) are imperfect organ or embryo representations that explore a much larger space of possible forms, or morphospace, compared to their in vivo counterparts. Here, we discuss SEM biology in light of seminal work by Pere Alberch, a leading figure in early evo-devo, interpreting SEMs as developmental 'monstrosities' in the Alberchian sense. Alberch suggested that ordered patterns in aberrant development-i.e. 'the logic of monsters'-reveal developmental constraints on possible morphologies. In the same vein, we detail how SEMs have begun to shed light on structural features of normal development, such as developmental variability, the relative importance of internal versus external constraints, boundary conditions and design principles governing robustness and canalization. We argue that SEMs represent a powerful experimental tool to explore and expand developmental morphospace and propose that the 'monstrosity' of SEMs can be leveraged to uncover the 'hidden' rules and developmental constraints that robustly shape and pattern the embryo.
Collapse
Affiliation(s)
- Dominica Cao
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT06520, USA
| | - Sumit Garai
- Theoretical Biology Lab, The Francis Crick Institute, LondonNW1 1AT, UK
- Division of Biosciences, Medical Sciences Building, University College London, Gower Street, LondonWC1E 6BT, UK
| | - James DiFrisco
- Theoretical Biology Lab, The Francis Crick Institute, LondonNW1 1AT, UK
| | - Jesse V. Veenvliet
- Stembryogenesis Lab, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden01307, Germany
- Center for Systems Biology Dresden, Dresden01307, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden01307, Germany
| |
Collapse
|
29
|
Yagi M, Horng JE, Hochedlinger K. Manipulating cell fate through reprogramming: approaches and applications. Development 2024; 151:dev203090. [PMID: 39348466 PMCID: PMC11463964 DOI: 10.1242/dev.203090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/11/2024] [Indexed: 10/02/2024]
Abstract
Cellular plasticity progressively declines with development and differentiation, yet these processes can be experimentally reversed by reprogramming somatic cells to induced pluripotent stem cells (iPSCs) using defined transcription factors. Advances in reprogramming technology over the past 15 years have enabled researchers to study diseases with patient-specific iPSCs, gain fundamental insights into how cell identity is maintained, recapitulate early stages of embryogenesis using various embryo models, and reverse aspects of aging in cultured cells and animals. Here, we review and compare currently available reprogramming approaches, including transcription factor-based methods and small molecule-based approaches, to derive pluripotent cells characteristic of early embryos. Additionally, we discuss our current understanding of mechanisms that resist reprogramming and their role in cell identity maintenance. Finally, we review recent efforts to rejuvenate cells and tissues with reprogramming factors, as well as the application of iPSCs in deriving novel embryo models to study pre-implantation development.
Collapse
Affiliation(s)
- Masaki Yagi
- Department of Molecular Biology, Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joy E. Horng
- Department of Molecular Biology, Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
30
|
Lin C, Liu S, Huang M, Zhang Y, Hu X. Induction of human stem cells into ameloblasts by reaggregation strategy. Stem Cell Res Ther 2024; 15:332. [PMID: 39334282 PMCID: PMC11437913 DOI: 10.1186/s13287-024-03948-1] [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: 05/10/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Human epithelium-derived stem cells and induced pluripotent stem cells (hiPSCs) possess the capability to support tooth formation and differentiate into functional enamel-secreting ameloblasts, making them promising epithelial-component substitutes for future human tooth regeneration. However, current tissue recombination approaches are not only technically challenging, requiring precise induction procedures and sophisticated microsurgery, but also exhibit low success rates in achieving tooth formation and ameloblastic differentiation. METHODS Suspended human keratinocyte stem cells (hKSCs) or cells from three hiPSC lines were directly mixed with dissociated embryonic mouse dental mesenchymal cells (mDMCs) that possess odontogenic potential in different proportions and reaggregated them to construct bioengineered tooth germs. The success rates of tooth formation and ameloblastic differentiation were confirmed after subrenal culture. The sorting capability, sequential development, and ameloblastic differentiation of stem cells were examined via GFP tracing, RT-PCR, and histological analysis, respectively. RESULTS Our reaggregation approach achieved an impressive success rate of more than 90% in tooth formation and 100% in ameloblastic differentiation when the chimeric tooth germs contained 1%~10% hKSCs or 5% hiPSCs. In addition, we observed that hiPSCs, upon exposure to mDMCs, initially transformed into epidermal cells, as indicated by KRT14 and CD29 expression, before progressing into dental epithelial cells, as indicated by SP6 and SHH expression. We also found that epithelial-derived hiPSCs, when reaggregated with mDMCs, were more favorable for tooth formation than their mesenchymal-derived counterparts. CONCLUSIONS This study establishes a simplified yet highly effective cell-cell reaggregation strategy for inducing stem cells to support tooth formation and differentiate into functional ameloblasts, paving the way for novel approaches for the development of stem cell-based tooth organoids and bioengineered tooth germs in vitro.
Collapse
Affiliation(s)
- Chensheng Lin
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, Fujian, P.R. China.
| | - Shiyu Liu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, Fujian, P.R. China
| | - Minjun Huang
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, Fujian, P.R. China
| | - Yanding Zhang
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, Fujian, P.R. China
| | - Xuefeng Hu
- Fujian Key Laboratory of Developmental and Neural Biology & Southern Center for Biomedical Research, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, Fujian, P.R. China.
| |
Collapse
|
31
|
Ranjan R, Ma B, Gleason RJ, Liao Y, Bi Y, Davis BEM, Yang G, Clark M, Mahajan V, Condon M, Broderick NA, Chen X. Modulating DNA Polα Enhances Cell Reprogramming Across Species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613993. [PMID: 39345551 PMCID: PMC11429986 DOI: 10.1101/2024.09.19.613993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
As a fundamental biological process, DNA replication ensures the accurate copying of genetic information. However, the impact of this process on cellular plasticity in multicellular organisms remains elusive. Here, we find that reducing the level or activity of a replication component, DNA Polymerase α (Polα), facilitates cell reprogramming in diverse stem cell systems across species. In Drosophila male and female germline stem cell lineages, reducing Polα levels using heterozygotes significantly enhances fertility of both sexes, promoting reproductivity during aging without compromising their longevity. Consistently, in C. elegans the pola heterozygous hermaphrodites exhibit increased fertility without a reduction in lifespan, suggesting that this phenomenon is conserved. Moreover, in male germline and female intestinal stem cell lineages of Drosophila, polα heterozygotes exhibit increased resistance to tissue damage caused by genetic ablation or pathogen infection, leading to enhanced regeneration and improved survival during post-injury recovery, respectively. Additionally, fine tuning of an inhibitor to modulate Polα activity significantly enhances the efficiency of reprogramming human embryonic fibroblasts into induced pluripotent cells. Together, these findings unveil novel roles of a DNA replication component in regulating cellular reprogramming potential, and thus hold promise for promoting tissue health, facilitating post-injury rehabilitation, and enhancing healthspan.
Collapse
Affiliation(s)
- Rajesh Ranjan
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Binbin Ma
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Ryan J. Gleason
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yijun Liao
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yingshan Bi
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brendon E. M. Davis
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Guanghui Yang
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Maggie Clark
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vikrant Mahajan
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Madison Condon
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| |
Collapse
|
32
|
Potapova T, Kostos P, McKinney S, Borchers M, Haug J, Guarracino A, Solar S, Gogol M, Monfort Anez G, de Lima LG, Wang Y, Hall K, Hoffman S, Garrison E, Phillippy AM, Gerton JL. Epigenetic control and inheritance of rDNA arrays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612795. [PMID: 39372739 PMCID: PMC11451732 DOI: 10.1101/2024.09.13.612795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Ribosomal RNA (rRNA) genes exist in multiple copies arranged in tandem arrays known as ribosomal DNA (rDNA). The total number of gene copies is variable, and the mechanisms buffering this copy number variation remain unresolved. We surveyed the number, distribution, and activity of rDNA arrays at the level of individual chromosomes across multiple human and primate genomes. Each individual possessed a unique fingerprint of copy number distribution and activity of rDNA arrays. In some cases, entire rDNA arrays were transcriptionally silent. Silent rDNA arrays showed reduced association with the nucleolus and decreased interchromosomal interactions, indicating that the nucleolar organizer function of rDNA depends on transcriptional activity. Methyl-sequencing of flow-sorted chromosomes, combined with long read sequencing, showed epigenetic modification of rDNA promoter and coding region by DNA methylation. Silent arrays were in a closed chromatin state, as indicated by the accessibility profiles derived from Fiber-seq. Removing DNA methylation restored the transcriptional activity of silent arrays. Array activity status remained stable through the iPS cell re-programming. Family trio analysis demonstrated that the inactive rDNA haplotype can be traced to one of the parental genomes, suggesting that the epigenetic state of rDNA arrays may be heritable. We propose that the dosage of rRNA genes is epigenetically regulated by DNA methylation, and these methylation patterns specify nucleolar organizer function and can propagate transgenerationally.
Collapse
Affiliation(s)
- Tamara Potapova
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Paxton Kostos
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Sean McKinney
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Jeff Haug
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Andrea Guarracino
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Steven Solar
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Madelaine Gogol
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | | | - Yan Wang
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Kate Hall
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Erik Garrison
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Adam M. Phillippy
- Genome Informatics Section, Center for Genomics and Data Science Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer L. Gerton
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
33
|
Benazzato C, Lojudice F, Pöehlchen F, Leite PEC, Manucci AC, Van der Linden V, Jungmann P, Sogayar MC, Bruni-Cardoso A, Russo FB, Beltrão-Braga P. Zika virus vertical transmission induces neuroinflammation and synapse impairment in brain cells derived from children born with Congenital Zika Syndrome. Sci Rep 2024; 14:18002. [PMID: 39097642 PMCID: PMC11297915 DOI: 10.1038/s41598-024-65392-8] [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: 12/01/2023] [Accepted: 06/18/2024] [Indexed: 08/05/2024] Open
Abstract
Zika virus (ZIKV) infection was first reported in 2015 in Brazil as causing microcephaly and other developmental abnormalities in newborns, leading to the identification of Congenital Zika Syndrome (CZS). Viral infections have been considered an environmental risk factor for neurodevelopmental disorders outcome, such as Autism Spectrum Disorder (ASD). Moreover, not only the infection per se, but maternal immune system activation during pregnancy, has been linked to fetal neurodevelopmental disorders. To understand the impact of ZIKV vertical infection on brain development, we derived induced pluripotent stem cells (iPSC) from Brazilian children born with CZS, some of the patients also being diagnosed with ASD. Comparing iPSC-derived neurons from CZS with a control group, we found lower levels of pre- and postsynaptic proteins and reduced functional synapses by puncta co-localization. Furthermore, neurons and astrocytes derived from the CZS group showed decreased glutamate levels. Additionally, the CZS group exhibited elevated levels of cytokine production, one of which being IL-6, already associated with the ASD phenotype. These preliminary findings suggest that ZIKV vertical infection may cause long-lasting disruptions in brain development during fetal stages, even in the absence of the virus after birth. These disruptions could contribute to neurodevelopmental disorders manifestations such as ASD. Our study contributes with novel knowledge of the CZS outcomes and paves the way for clinical validation and the development of potential interventions to mitigate the impact of ZIKV vertical infection on neurodevelopment.
Collapse
Affiliation(s)
- Cecilia Benazzato
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil
| | - Fernando Lojudice
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo-SP, 01246-903, Brazil
| | - Felizia Pöehlchen
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil
- Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Paulo Emílio Corrêa Leite
- Clinical Research Unit of the Antonio Pedro Hospital, Federal Fluminense University, Rio de Janeiro, 24220-900, Brazil
| | - Antonio Carlos Manucci
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | | | - Patricia Jungmann
- Pathology Department, University of Pernambuco, Recife, 50670-901, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo-SP, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Alexandre Bruni-Cardoso
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - Fabiele B Russo
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil.
| | - Patricia Beltrão-Braga
- Microbiology Department, Institute of Biomedical Sciences (ICB-II), University of São Paulo, Av. Prof Lineu Prestes, 1374, 2Nd Floor, Room 235, São Paulo, SP, 05508-000, Brazil.
- Institute Pasteur of São Paulo, Av. Prof. Lucio Martins Rodrigues 370, A-Building, 4Th Floor, São Paulo-SP, 05508-020, Brazil.
| |
Collapse
|
34
|
Tereshchenko Y, Petkov SG, Behr R. The Efficiency of In Vitro Differentiation of Primate iPSCs into Cardiomyocytes Depending on Their Cell Seeding Density and Cell Line Specificity. Int J Mol Sci 2024; 25:8449. [PMID: 39126016 PMCID: PMC11312487 DOI: 10.3390/ijms25158449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
A thorough characterization of induced pluripotent stem cells (iPSCs) used with in vitro models or therapeutics is essential. Even iPSCs derived from a single donor can exhibit variability within and between cell lines, which can lead to heterogeneity in results and hinder the promising future of cell replacement therapies. In this study, the cell seeding density of human and rhesus monkey iPSCs was tested to maximize the cell line-specific yield of the generated cardiomyocytes. We found that, despite using the same iPSC generation and differentiation protocols, the cell seeding density for the cell line-specific best differentiation efficiency could differ by a factor of four for the four cell lines used here. In addition, the cell lines showed differences in the range of cell seeding densities that they could tolerate without the severe loss of differentiation efficiency. Overall, our data show that the cell seeding density is a critical parameter for the differentiation inefficiency of primate iPSCs to cardiomyocytes and that iPSCs generated with the same episomal approach still exhibit considerable heterogeneity. Therefore, individual characterization of iPSC lines is required, and functional comparability with in vivo processes must be ensured to warrant the translatability of in vitro research with iPSCs.
Collapse
Affiliation(s)
- Yuliia Tereshchenko
- Research Platform Stem Cell Biology and Regeneration, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; (Y.T.); (S.G.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Lower Saxony, 37077 Göttingen, Germany
| | - Stoyan G. Petkov
- Research Platform Stem Cell Biology and Regeneration, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; (Y.T.); (S.G.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Lower Saxony, 37077 Göttingen, Germany
| | - Rüdiger Behr
- Research Platform Stem Cell Biology and Regeneration, German Primate Center–Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; (Y.T.); (S.G.P.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Lower Saxony, 37077 Göttingen, Germany
| |
Collapse
|
35
|
Clancy CE, Santana LF. Advances in induced pluripotent stem cell-derived cardiac myocytes: technological breakthroughs, key discoveries and new applications. J Physiol 2024; 602:3871-3892. [PMID: 39032073 PMCID: PMC11326976 DOI: 10.1113/jp282562] [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: 02/21/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024] Open
Abstract
A transformation is underway in precision and patient-specific medicine. Rapid progress has been enabled by multiple new technologies including induced pluripotent stem cell-derived cardiac myocytes (iPSC-CMs). Here, we delve into these advancements and their future promise, focusing on the efficiency of reprogramming techniques, the fidelity of differentiation into the cardiac lineage, the functional characterization of the resulting cardiac myocytes, and the many applications of in silico models to understand general and patient-specific mechanisms controlling excitation-contraction coupling in health and disease. Furthermore, we explore the current and potential applications of iPSC-CMs in both research and clinical settings, underscoring the far-reaching implications of this rapidly evolving field.
Collapse
Affiliation(s)
- Colleen E Clancy
- Department of Physiology & Membrane Biology, School of Medicine, University of California Davis, Davis, CA, USA
- Center for Precision Medicine and Data Sciences, University of California Davis, School of Medicine, Sacramento, CA, USA
| | - L Fernando Santana
- Department of Physiology & Membrane Biology, School of Medicine, University of California Davis, Davis, CA, USA
- Center for Precision Medicine and Data Sciences, University of California Davis, School of Medicine, Sacramento, CA, USA
| |
Collapse
|
36
|
Han J, Son H, Jung D, Kim KY, Jin C, Hwang H, Kang SS, Mitalipov S, An HJ, Lee Y, Kang E. Comparison of Natural Killer Cells Differentiated from Various Pluripotent Stem Cells. Int J Mol Sci 2024; 25:8209. [PMID: 39125777 PMCID: PMC11311719 DOI: 10.3390/ijms25158209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/03/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Allogeneic natural killer (NK) cell therapy has been effective in treating cancer. Many studies have tested NK cell therapy using human pluripotent stem cells (hPSCs). However, the impacts of the origin of PSC-NK cells on competence are unclear. In this study, several types of hPSCs, including human-induced PSCs (hiPSCs) generated from CD34+, CD3-CD56+, and CD56- cells in umbilical cord blood (UCB), three lines of human embryonic stem cells (hESCs, ES-1. ES-2 and ES-3) and MHC I knockout (B2M-KO)-ESCs were used to differentiate into NK cells and their capacities were analyzed. All PSC types could differentiate into NK cells. Among the iPSC-derived NK cells (iPSC-NKs) and ESC-derived NK cells (ES-NKs), 34+ iPSCs and ES-3 had a higher growth rate and cytotoxicity, respectively, ES-3 also showed better efficacy than 34+ iPSCs. B2M-KO was similar to the wild type. These results suggest that the screening for differentiation of PSCs into NK cells prior to selecting the PSC lines for the development of NK cell immunotherapy is an essential process for universal allotransplantation, including the chimeric antigen receptor (CAR).
Collapse
Affiliation(s)
- Jongsuk Han
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Hyeongbin Son
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Daun Jung
- Department of Pathology, CHA Bundang Medical Center, CHA University, Sungnam-si 13496, Gyeonggi-do, Republic of Korea; (D.J.); (K.-Y.K.); (H.-J.A.)
| | - Ki-Yeon Kim
- Department of Pathology, CHA Bundang Medical Center, CHA University, Sungnam-si 13496, Gyeonggi-do, Republic of Korea; (D.J.); (K.-Y.K.); (H.-J.A.)
| | - Chaeyeon Jin
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Hyeonwook Hwang
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Soon-Suk Kang
- Cell Therapy 3 Center, CHA Advanced Research Institute, CHA Bundang Medical Center, Sungnam-si 13488, Gyeonggi-do, Republic of Korea;
| | - Shoukhrat Mitalipov
- Center for Embryonic Cell and Gene Therapy, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Hee-Jung An
- Department of Pathology, CHA Bundang Medical Center, CHA University, Sungnam-si 13496, Gyeonggi-do, Republic of Korea; (D.J.); (K.-Y.K.); (H.-J.A.)
| | - Yeonmi Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
- Cell Therapy 3 Center, CHA Advanced Research Institute, CHA Bundang Medical Center, Sungnam-si 13488, Gyeonggi-do, Republic of Korea;
| | - Eunju Kang
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
- Cell Therapy 3 Center, CHA Advanced Research Institute, CHA Bundang Medical Center, Sungnam-si 13488, Gyeonggi-do, Republic of Korea;
- Department of Biochemistry, School of Medicine, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| |
Collapse
|
37
|
Nakamura Y, Kobayashi H, Fukuda N, Tanaka S, Murata Y, Hatanaka Y, Haketa A, Tsunemi A, Chen L, Abe M. Induced pluripotent stem cells derived renal tubular cells from a patient with pseudohypoparathyroidism and its response to parathyroid hormone stimulation. Mol Biol Rep 2024; 51:790. [PMID: 38990390 DOI: 10.1007/s11033-024-09751-8] [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: 02/01/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION Creating induced pluripotent stem cells (iPSCs) from somatic cells of patients with genetic diseases offers a pathway to generate disease-specific iPSCs carrying genetic markers. Differentiating these iPSCs into renal tubular cells can aid in understanding the pathophysiology of rare inherited renal tubular diseases through cellular experiments. MATERIALS AND METHODS Two Japanese patients with Pseudohypoparathyroidism (PHP), a 49-year-old woman and a 71-year-old man, were studied. iPSC-derived tubular cells were established from their peripheral blood mononuclear cells (PBMCs). We examined changes in intracellular and extracellular cyclic adenosine monophosphate (cAMP) levels in these cells in response to parathyroid hormone (PTH) stimulation. RESULTS Renal tubular cells, differentiated from iPSCs of a healthy control (648A1), showed a PTH-dependent increase in both intracellular and extracellular cAMP levels. However, the renal tubular cells derived from the PHP patients' iPSCs showed inconsistent changes in cAMP levels upon PTH exposure. CONCLUSION We successfully created disease-specific iPSCs from PHP patients' PBMCs, differentiated them into tubular cells, and replicated the distinctive response of the disease to PTH in vitro. This approach could enhance our understanding of the pathophysiology of inherited renal tubular diseases and contribute to developing effective treatments.
Collapse
Affiliation(s)
- Yoshihiro Nakamura
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Hiroki Kobayashi
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan.
| | - Noboru Fukuda
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Sho Tanaka
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Yusuke Murata
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Yoshinari Hatanaka
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Akira Haketa
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Akiko Tsunemi
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Lan Chen
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Masanori Abe
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi Kami-chou, Itabashi-ku, Tokyo, 173-8610, Japan
| |
Collapse
|
38
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [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: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
39
|
Seah I, Goh D, Banerjee A, Su X. Modeling inherited retinal diseases using human induced pluripotent stem cell derived photoreceptor cells and retinal pigment epithelial cells. Front Med (Lausanne) 2024; 11:1328474. [PMID: 39011458 PMCID: PMC11246861 DOI: 10.3389/fmed.2024.1328474] [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: 10/26/2023] [Accepted: 06/18/2024] [Indexed: 07/17/2024] Open
Abstract
Since the discovery of induced pluripotent stem cell (iPSC) technology, there have been many attempts to create cellular models of inherited retinal diseases (IRDs) for investigation of pathogenic processes to facilitate target discovery and validation activities. Consistency remains key in determining the utility of these findings. Despite the importance of consistency, quality control metrics are still not widely used. In this review, a toolkit for harnessing iPSC technology to generate photoreceptor, retinal pigment epithelial cell, and organoid disease models is provided. Considerations while developing iPSC-derived IRD models such as iPSC origin, reprogramming methods, quality control metrics, control strategies, and differentiation protocols are discussed. Various iPSC IRD models are dissected and the scientific hurdles of iPSC-based disease modeling are discussed to provide an overview of current methods and future directions in this field.
Collapse
Affiliation(s)
- Ivan Seah
- Translational Retinal Research Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Debbie Goh
- Department of Ophthalmology, National University Hospital (NUH), Singapore, Singapore
| | - Animesh Banerjee
- Translational Retinal Research Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xinyi Su
- Translational Retinal Research Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Ophthalmology, National University Hospital (NUH), Singapore, Singapore
- Singapore Eye Research Institute (SERI), Singapore, Singapore
| |
Collapse
|
40
|
Rehman A, Fatima I, Noor F, Qasim M, Wang P, Jia J, Alshabrmi FM, Liao M. Role of small molecules as drug candidates for reprogramming somatic cells into induced pluripotent stem cells: A comprehensive review. Comput Biol Med 2024; 177:108661. [PMID: 38810477 DOI: 10.1016/j.compbiomed.2024.108661] [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: 03/18/2024] [Revised: 04/08/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
With the use of specific genetic factors and recent developments in cellular reprogramming, it is now possible to generate lineage-committed cells or induced pluripotent stem cells (iPSCs) from readily available and common somatic cell types. However, there are still significant doubts regarding the safety and effectiveness of the current genetic methods for reprogramming cells, as well as the conventional culture methods for maintaining stem cells. Small molecules that target specific epigenetic processes, signaling pathways, and other cellular processes can be used as a complementary approach to manipulate cell fate to achieve a desired objective. It has been discovered that a growing number of small molecules can support lineage differentiation, maintain stem cell self-renewal potential, and facilitate reprogramming by either increasing the efficiency of reprogramming or acting as a genetic reprogramming factor substitute. However, ongoing challenges include improving reprogramming efficiency, ensuring the safety of small molecules, and addressing issues with incomplete epigenetic resetting. Small molecule iPSCs have significant clinical applications in regenerative medicine and personalized therapies. This review emphasizes the versatility and potential safety benefits of small molecules in overcoming challenges associated with the iPSCs reprogramming process.
Collapse
Affiliation(s)
- Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Israr Fatima
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Fatima Noor
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan; Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University of Faisalabad, 38000, Pakistan
| | - Peng Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China
| | - Jinrui Jia
- Laboratory of Animal Fat Deposition and Muscle Development, Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, PR China
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Mingzhi Liao
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, PR China.
| |
Collapse
|
41
|
Taherian M, Bayati P, Mojtabavi N. Stem cell-based therapy for fibrotic diseases: mechanisms and pathways. Stem Cell Res Ther 2024; 15:170. [PMID: 38886859 PMCID: PMC11184790 DOI: 10.1186/s13287-024-03782-5] [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: 01/29/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Fibrosis is a pathological process, that could result in permanent scarring and impairment of the physiological function of the affected organ; this condition which is categorized under the term organ failure could affect various organs in different situations. The involvement of the major organs, such as the lungs, liver, kidney, heart, and skin, is associated with a high rate of morbidity and mortality across the world. Fibrotic disorders encompass a broad range of complications and could be traced to various illnesses and impairments; these could range from simple skin scars with beauty issues to severe rheumatologic or inflammatory disorders such as systemic sclerosis as well as idiopathic pulmonary fibrosis. Besides, the overactivation of immune responses during any inflammatory condition causing tissue damage could contribute to the pathogenic fibrotic events accompanying the healing response; for instance, the inflammation resulting from tissue engraftment could cause the formation of fibrotic scars in the grafted tissue, even in cases where the immune system deals with hard to clear infections, fibrotic scars could follow and cause severe adverse effects. A good example of such a complication is post-Covid19 lung fibrosis which could impair the life of the affected individuals with extensive lung involvement. However, effective therapies that halt or slow down the progression of fibrosis are missing in the current clinical settings. Considering the immunomodulatory and regenerative potential of distinct stem cell types, their application as an anti-fibrotic agent, capable of attenuating tissue fibrosis has been investigated by many researchers. Although the majority of the studies addressing the anti-fibrotic effects of stem cells indicated their potent capabilities, the underlying mechanisms, and pathways by which these cells could impact fibrotic processes remain poorly understood. Here, we first, review the properties of various stem cell types utilized so far as anti-fibrotic treatments and discuss the challenges and limitations associated with their applications in clinical settings; then, we will summarize the general and organ-specific mechanisms and pathways contributing to tissue fibrosis; finally, we will describe the mechanisms and pathways considered to be employed by distinct stem cell types for exerting anti-fibrotic events.
Collapse
Affiliation(s)
- Marjan Taherian
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Paria Bayati
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Nazanin Mojtabavi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
42
|
Jeyagaran A, Urbanczyk M, Layland SL, Weise F, Schenke-Layland K. Forward programming of hiPSCs towards beta-like cells using Ngn3, Pdx1, and MafA. Sci Rep 2024; 14:13608. [PMID: 38871849 PMCID: PMC11176171 DOI: 10.1038/s41598-024-64346-4] [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: 11/24/2023] [Accepted: 06/07/2024] [Indexed: 06/15/2024] Open
Abstract
Transplantation of stem cell-derived β-cells is a promising therapeutic advancement in the treatment of type 1 diabetes mellitus. A current limitation of this approach is the long differentiation timeline that generates a heterogeneous population of pancreatic endocrine cells. To address this limitation, an inducible lentiviral overexpression system of mature β-cell markers was introduced into human induced-pluripotent stem cells (hiPSCs). Following the selection of the successfully transduced hiPSCs, the cells were treated with doxycycline in the pancreatic progenitor induction medium to support their transition toward the pancreatic lineage. Cells cultured with doxycycline presented the markers of interest, NGN3, PDX1, and MAFA, after five days of culture, and glucose-stimulated insulin secretion assays demonstrated that the cells were glucose-responsive in a monolayer culture. When cultured as a spheroid, the markers of interest and insulin secretion in a static glucose-stimulated insulin secretion assay were maintained; however, insulin secretion upon consecutive glucose challenges was limited. Comparison to human fetal and adult donor tissues identified that although the hiPSC-derived spheroids present similar markers to adult insulin-producing cells, they are functionally representative of fetal development. Together, these results suggest that with optimization of the temporal expression of these markers, forward programming of hiPSCs towards insulin-producing cells could be a possible alternative for islet transplantation.
Collapse
Affiliation(s)
- Abiramy Jeyagaran
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Max Urbanczyk
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
| | - Shannon L Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076, Tübingen, Germany
- Department of Women's Health, Eberhard Karls University, 72076, Tübingen, Germany
| | - Frank Weise
- NMI Natural and Medical Sciences Institute at the University Tübingen, 72770, Reutlingen, Germany
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine, Eberhard Karls University Tübingen, 72076, Tübingen, Germany.
- NMI Natural and Medical Sciences Institute at the University Tübingen, 72770, Reutlingen, Germany.
| |
Collapse
|
43
|
Eintracht J, Owen N, Harding P, Moosajee M. Disruption of common ocular developmental pathways in patient-derived optic vesicle models of microphthalmia. Stem Cell Reports 2024; 19:839-858. [PMID: 38821055 PMCID: PMC11390689 DOI: 10.1016/j.stemcr.2024.05.001] [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: 09/13/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/02/2024] Open
Abstract
Genetic perturbations influencing early eye development can result in microphthalmia, anophthalmia, and coloboma (MAC). Over 100 genes are associated with MAC, but little is known about common disease mechanisms. In this study, we generated induced pluripotent stem cell (iPSC)-derived optic vesicles (OVs) from two unrelated microphthalmia patients and healthy controls. At day 20, 35, and 50, microphthalmia patient OV diameters were significantly smaller, recapitulating the "small eye" phenotype. RNA sequencing (RNA-seq) analysis revealed upregulation of apoptosis-initiating and extracellular matrix (ECM) genes at day 20 and 35. Western blot and immunohistochemistry revealed increased expression of lumican, nidogen, and collagen type IV, suggesting ECM overproduction. Increased apoptosis was observed in microphthalmia OVs with reduced phospho-histone 3 (pH3+) cells confirming decreased cell proliferation at day 35. Pharmacological inhibition of caspase-8 activity with Z-IETD-FMK decreased apoptosis in one patient model, highlighting a potential therapeutic approach. These data reveal shared pathophysiological mechanisms contributing to a microphthalmia phenotype.
Collapse
Affiliation(s)
| | | | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; Moorfields Eye Hospital NHS Foundation Trust, London EC1V 9EL, UK; Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
44
|
Sabetkish S, Currie P, Meagher L. Recent trends in 3D bioprinting technology for skeletal muscle regeneration. Acta Biomater 2024; 181:46-66. [PMID: 38697381 DOI: 10.1016/j.actbio.2024.04.038] [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: 12/14/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
Skeletal muscle is a pro-regenerative tissue, that utilizes a tissue-resident stem cell system to effect repair upon injury. Despite the demonstrated efficiency of this system in restoring muscle mass after many acute injuries, in conditions of severe trauma such as those evident in volumetric muscle loss (VML) (>20 % by mass), this self-repair capability is unable to restore tissue architecture, requiring interventions which currently are largely surgical. As a possible alternative, the generation of artificial muscle using tissue engineering approaches may also be of importance in the treatment of VML and muscle diseases such as dystrophies. Three-dimensional (3D) bioprinting has been identified as a promising technique for regeneration of the complex architecture of skeletal muscle. This review discusses existing treatment strategies following muscle damage, recent progress in bioprinting techniques, the bioinks used for muscle regeneration, the immunogenicity of scaffold materials, and in vitro and in vivo maturation techniques for 3D bio-printed muscle constructs. The pros and cons of these bioink formulations are also highlighted. Finally, we present the current limitations and challenges in the field and critical factors to consider for bioprinting approaches to become more translationa and to produce clinically relevant engineered muscle. STATEMENT OF SIGNIFICANCE: This review discusses the physiopathology of muscle injuries and existing clinical treatment strategies for muscle damage, the types of bioprinting techniques that have been applied to bioprinting of muscle, and the bioinks commonly used for muscle regeneration. The pros and cons of these bioinks are highlighted. We present a discussion of existing gaps in the literature and critical factors to consider for the translation of bioprinting approaches and to produce clinically relevant engineered muscle. Finally, we provide insights into what we believe will be the next steps required before the realization of the application of tissue-engineered muscle in humans. We believe this manuscript is an insightful, timely, and instructive review that will guide future muscle bioprinting research from a fundamental construct creation approach, down a translational pathway to achieve the desired impact in the clinic.
Collapse
Affiliation(s)
- Shabnam Sabetkish
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Peter Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia
| | - Laurence Meagher
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
45
|
Bingnan W, Jiao T, Ghorbani A, Baghei S. Enhancing regenerative potential: A comprehensive review of stem cell transplantation for sports-related neuronal injuries, with a focus on spinal cord injuries and peripheral nervous system damage. Tissue Cell 2024; 88:102429. [PMID: 38833939 DOI: 10.1016/j.tice.2024.102429] [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: 01/24/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
Neuronal injuries, as one of the consequences of sports-related incidents, exert a profound influence on the athletes' future, potentially leading to complete immobility and impeding their athletic pursuits. In cases of severe damage inflicted upon the spinal cord (SC) and peripheral nervous systems (PNS), the regenerative process is notably compromised, rendering it essentially inefficient. Among the pivotal therapeutic approaches for the enhancement and prevention of secondary SC injuries (SCI), stem cell transplantation (SCT) stands out prominently. Stem cells, whether directly involved in replacement and reconstruction or indirectly through modification and secretion of crucial bioenvironmental factors, engage in the intricate process of tissue regeneration. Stem cells, through the secretion of neurotrophic factors (NTFs) (aiming to modulate the immune system), reduction of inflammation, axonal growth stimulation, and myelin formation, endeavor to facilitate the regeneration of damaged SC tissue. The fundamental challenges of this approach encompass the proper selection of suitable stem cell candidates for transplantation and the establishment of an appropriate microenvironment conducive to SC repair. In this article, an attempt has been made to explore sports-related injuries, particularly SCI, to comprehensively review innovative methods for treating SCI, and to address the existing challenges. Additionally, some of the stem cells used in neural injuries and the process of their utilization have been discussed.
Collapse
Affiliation(s)
- Wang Bingnan
- Department of P.E, Central South University, Changsha 410083, China
| | - Tong Jiao
- The High School Attached to Hunan Normal University Bocai Experimental Middle School,Changsha 410208, China.
| | - A Ghorbani
- Biotechnology Department, Islamic Azad University, Isfahan, Iran
| | - Sh Baghei
- Biotechnology Department, Islamic Azad University, Isfahan, Iran.
| |
Collapse
|
46
|
Nakamura Y, Niho S, Shimizu Y. Cell-Based Therapy for Fibrosing Interstitial Lung Diseases, Current Status, and Potential Applications of iPSC-Derived Cells. Cells 2024; 13:893. [PMID: 38891026 PMCID: PMC11172081 DOI: 10.3390/cells13110893] [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: 04/14/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Fibrosing interstitial lung diseases (FILDs), e.g., due to idiopathic pulmonary fibrosis (IPF), are chronic progressive diseases with a poor prognosis. The management of these diseases is challenging and focuses mainly on the suppression of progression with anti-fibrotic drugs. Therefore, novel FILD treatments are needed. In recent years, cell-based therapy with various stem cells has been investigated for FILD, and the use of mesenchymal stem cells (MSCs) has been widely reported and clinical studies are also ongoing. Induced pluripotent stem cells (iPSCs) have also been reported to have an anti-fibrotic effect in FILD; however, these have not been as well studied as MSCs in terms of the mechanisms and side effects. While MSCs show a potent anti-fibrotic effect, the possibility of quality differences between donors and a stable supply in the case of donor shortage or reduced proliferative capacity after cell passaging needs to be considered. The application of iPSC-derived cells has the potential to overcome these problems and may lead to consistent quality of the cell product and stable product supply. This review provides an overview of iPSCs and FILD, followed by the current status of cell-based therapy for FILD, and then discusses the possibilities and perspectives of FILD therapy with iPSC-derived cells.
Collapse
Affiliation(s)
- Yusuke Nakamura
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
- Center of Regenerative Medicine, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
| | - Seiji Niho
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
| | - Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University School of Medicine, Mibu 321-0293, Japan; (Y.N.); (S.N.)
- Center of Regenerative Medicine, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
- Respiratory Endoscopy Center, Dokkyo Medical University Hospital, Mibu 321-0293, Japan
| |
Collapse
|
47
|
Tsitkov S, Valentine K, Kozareva V, Donde A, Frank A, Lei S, E Van Eyk J, Finkbeiner S, Rothstein JD, Thompson LM, Sareen D, Svendsen CN, Fraenkel E. Disease related changes in ATAC-seq of iPSC-derived motor neuron lines from ALS patients and controls. Nat Commun 2024; 15:3606. [PMID: 38697975 PMCID: PMC11066062 DOI: 10.1038/s41467-024-47758-8] [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: 01/05/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS), like many other neurodegenerative diseases, is highly heritable, but with only a small fraction of cases explained by monogenic disease alleles. To better understand sporadic ALS, we report epigenomic profiles, as measured by ATAC-seq, of motor neuron cultures derived from a diverse group of 380 ALS patients and 80 healthy controls. We find that chromatin accessibility is heavily influenced by sex, the iPSC cell type of origin, ancestry, and the inherent variance arising from sequencing. Once these covariates are corrected for, we are able to identify ALS-specific signals in the data. Additionally, we find that the ATAC-seq data is able to predict ALS disease progression rates with similar accuracy to methods based on biomarkers and clinical status. These results suggest that iPSC-derived motor neurons recapitulate important disease-relevant epigenomic changes.
Collapse
Affiliation(s)
- Stanislav Tsitkov
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kelsey Valentine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Velina Kozareva
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aneesh Donde
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Aaron Frank
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Steve Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA, USA
- Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, CA, USA
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey D Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA, USA
| | - Dhruv Sareen
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- The Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
48
|
Luanpitpong S, Tangkiettrakul K, Kang X, Srisook P, Poohadsuan J, Samart P, Klaihmon P, Janan M, Lorthongpanich C, Laowtammathron C, Issaragrisil S. OGT and OGA gene-edited human induced pluripotent stem cells for dissecting the functional roles of O-GlcNAcylation in hematopoiesis. Front Cell Dev Biol 2024; 12:1361943. [PMID: 38752196 PMCID: PMC11094211 DOI: 10.3389/fcell.2024.1361943] [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: 12/27/2023] [Accepted: 03/14/2024] [Indexed: 05/18/2024] Open
Abstract
Hematopoiesis continues throughout life to produce all types of blood cells from hematopoietic stem cells (HSCs). Metabolic state is a known regulator of HSC self-renewal and differentiation, but whether and how metabolic sensor O-GlcNAcylation, which can be modulated via an inhibition of its cycling enzymes O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT), contributes to hematopoiesis remains largely unknown. Herein, isogenic, single-cell clones of OGA-depleted (OGAi) and OGT-depleted (OGTi) human induced pluripotent stem cells (hiPSCs) were successfully generated from the master hiPSC line MUSIi012-A, which were reprogrammed from CD34+ hematopoietic stem/progenitor cells (HSPCs) containing epigenetic memory. The established OGAi and OGTi hiPSCs exhibiting an increase or decrease in cellular O-GlcNAcylation concomitant with their loss of OGA and OGT, respectively, appeared normal in phenotype and karyotype, and retained pluripotency, although they may favor differentiation toward certain germ lineages. Upon hematopoietic differentiation through mesoderm induction and endothelial-to-hematopoietic transition, we found that OGA inhibition accelerates hiPSC commitment toward HSPCs and that disruption of O-GlcNAc homeostasis affects their commitment toward erythroid lineage. The differentiated HSPCs from all groups were capable of giving rise to all hematopoietic progenitors, thus confirming their functional characteristics. Altogether, the established single-cell clones of OGTi and OGAi hiPSCs represent a valuable platform for further dissecting the roles of O-GlcNAcylation in blood cell development at various stages and lineages of blood cells. The incomplete knockout of OGA and OGT in these hiPSCs makes them susceptible to additional manipulation, i.e., by small molecules, allowing the molecular dynamics studies of O-GlcNAcylation.
Collapse
Affiliation(s)
- Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kantpitchar Tangkiettrakul
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Xing Kang
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pimonwan Srisook
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jirarat Poohadsuan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parinya Samart
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Phatchanat Klaihmon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
49
|
Cerneckis J, Cai H, Shi Y. Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications. Signal Transduct Target Ther 2024; 9:112. [PMID: 38670977 PMCID: PMC11053163 DOI: 10.1038/s41392-024-01809-0] [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: 07/28/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024] Open
Abstract
The induced pluripotent stem cell (iPSC) technology has transformed in vitro research and holds great promise to advance regenerative medicine. iPSCs have the capacity for an almost unlimited expansion, are amenable to genetic engineering, and can be differentiated into most somatic cell types. iPSCs have been widely applied to model human development and diseases, perform drug screening, and develop cell therapies. In this review, we outline key developments in the iPSC field and highlight the immense versatility of the iPSC technology for in vitro modeling and therapeutic applications. We begin by discussing the pivotal discoveries that revealed the potential of a somatic cell nucleus for reprogramming and led to successful generation of iPSCs. We consider the molecular mechanisms and dynamics of somatic cell reprogramming as well as the numerous methods available to induce pluripotency. Subsequently, we discuss various iPSC-based cellular models, from mono-cultures of a single cell type to complex three-dimensional organoids, and how these models can be applied to elucidate the mechanisms of human development and diseases. We use examples of neurological disorders, coronavirus disease 2019 (COVID-19), and cancer to highlight the diversity of disease-specific phenotypes that can be modeled using iPSC-derived cells. We also consider how iPSC-derived cellular models can be used in high-throughput drug screening and drug toxicity studies. Finally, we discuss the process of developing autologous and allogeneic iPSC-based cell therapies and their potential to alleviate human diseases.
Collapse
Affiliation(s)
- Jonas Cerneckis
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Hongxia Cai
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
50
|
Ozkan A, Padmanabhan HK, Shipman SL, Azim E, Kumar P, Sadegh C, Basak AN, Macklis JD. Directed differentiation of functional corticospinal-like neurons from endogenous SOX6+/NG2+ cortical progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.21.590488. [PMID: 38712174 PMCID: PMC11071355 DOI: 10.1101/2024.04.21.590488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Corticospinal neurons (CSN) centrally degenerate in amyotrophic lateral sclerosis (ALS), along with spinal motor neurons, and loss of voluntary motor function in spinal cord injury (SCI) results from damage to CSN axons. For functional regeneration of specifically affected neuronal circuitry in vivo , or for optimally informative disease modeling and/or therapeutic screening in vitro , it is important to reproduce the type or subtype of neurons involved. No such appropriate in vitro models exist with which to investigate CSN selective vulnerability and degeneration in ALS, or to investigate routes to regeneration of CSN circuitry for ALS or SCI, critically limiting the relevance of much research. Here, we identify that the HMG-domain transcription factor Sox6 is expressed by a subset of NG2+ endogenous cortical progenitors in postnatal and adult cortex, and that Sox6 suppresses a latent neurogenic program by repressing inappropriate proneural Neurog2 expression by progenitors. We FACS-purify these genetically accessible progenitors from postnatal mouse cortex and establish a pure culture system to investigate their potential for directed differentiation into CSN. We then employ a multi-component construct with complementary and differentiation-sharpening transcriptional controls (activating Neurog2, Fezf2 , while antagonizing Olig2 with VP16:Olig2 ). We generate corticospinal-like neurons from SOX6+/NG2+ cortical progenitors, and find that these neurons differentiate with remarkable fidelity compared with corticospinal neurons in vivo . They possess appropriate morphological, molecular, transcriptomic, and electrophysiological characteristics, without characteristics of the alternate intracortical or other neuronal subtypes. We identify that these critical specifics of differentiation are not reproduced by commonly employed Neurog2 -driven differentiation. Neurons induced by Neurog2 instead exhibit aberrant multi-axon morphology and express molecular hallmarks of alternate cortical projection subtypes, often in mixed form. Together, this developmentally-based directed differentiation from genetically accessible cortical progenitors sets a precedent and foundation for in vitro mechanistic and therapeutic disease modeling, and toward regenerative neuronal repopulation and circuit repair.
Collapse
|