1
|
Wang J, Zhao M, Fu D, Wang M, Han C, Lv Z, Wang L, Liu J. Human neural stem cell-derived extracellular vesicles protect against ischemic stroke by activating the PI3K/AKT/mTOR pathway. Neural Regen Res 2025; 20:3245-3258. [PMID: 39248158 PMCID: PMC11881723 DOI: 10.4103/nrr.nrr-d-23-01144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/11/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00028/figure1/v/2024-12-20T164640Z/r/image-tiff Human neural stem cell-derived extracellular vesicles exhibit analogous functions to their parental cells, and can thus be used as substitutes for stem cells in stem cell therapy, thereby mitigating the risks of stem cell therapy and advancing the frontiers of stem cell-derived treatments. This lays a foundation for the development of potentially potent new treatment modalities for ischemic stroke. However, the precise mechanisms underlying the efficacy and safety of human neural stem cell-derived extracellular vesicles remain unclear, presenting challenges for clinical translation. To promote the translation of therapy based on human neural stem cell-derived extracellular vesicles from the bench to the bedside, we conducted a comprehensive preclinical study to evaluate the efficacy and safety of human neural stem cell-derived extracellular vesicles in the treatment of ischemic stroke. We found that administration of human neural stem cell-derived extracellular vesicles to an ischemic stroke rat model reduced the volume of cerebral infarction and promoted functional recovery by alleviating neuronal apoptosis. The human neural stem cell-derived extracellular vesicles reduced neuronal apoptosis by enhancing phosphorylation of phosphoinositide 3-kinase, mammalian target of rapamycin, and protein kinase B, and these effects were reversed by treatment with a phosphoinositide 3-kinase inhibitor. These findings suggest that human neural stem cell-derived extracellular vesicles play a neuroprotective role in ischemic stroke through activation of phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway. Finally, we showed that human neural stem cell-derived extracellular vesicles have a good in vivo safety profile. Therefore, human neural stem cell-derived extracellular vesicles are a promising potential agent for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Mengke Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Dong Fu
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning Province, China
| | - Meina Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Chao Han
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Zhongyue Lv
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning Province, China
| |
Collapse
|
2
|
Wiley L, Cheek M, LaFar E, Ma X, Sekowski J, Tanguturi N, Iltis A. The Ethics of Human Embryo Editing via CRISPR-Cas9 Technology: A Systematic Review of Ethical Arguments, Reasons, and Concerns. HEC Forum 2025; 37:267-303. [PMID: 39302534 PMCID: PMC12014773 DOI: 10.1007/s10730-024-09538-1] [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] [Accepted: 08/29/2024] [Indexed: 09/22/2024]
Abstract
The possibility of editing the genomes of human embryos has generated significant discussion and interest as a matter of science and ethics. While it holds significant promise to prevent or treat disease, research on and potential clinical applications of human embryo editing also raise ethical, regulatory, and safety concerns. This systematic review included 223 publications to identify the ethical arguments, reasons, and concerns that have been offered for and against the editing of human embryos using CRISPR-Cas9 technology. We identified six major themes: risk/harm; potential benefit; oversight; informed consent; justice, equity, and other social considerations; and eugenics. We explore these themes and provide an overview and analysis of the critical points in the current literature.
Collapse
Affiliation(s)
- Lindsay Wiley
- Wake Forest University Center for Bioethics, Health and Society, Winston-Salem, USA
| | - Mattison Cheek
- Wake Forest University Center for Bioethics, Health and Society, Winston-Salem, USA
| | - Emily LaFar
- Wake Forest University Center for Bioethics, Health and Society, Winston-Salem, USA
| | - Xiaolu Ma
- Department of Communication Studies, University of Minnesota, Minneapolis, USA
| | - Justin Sekowski
- Wake Forest University Center for Bioethics, Health and Society, Winston-Salem, USA
| | - Nikki Tanguturi
- Wake Forest University Center for Bioethics, Health and Society, Winston-Salem, USA
| | - Ana Iltis
- Department of Philosophy, Wake Forest University Center for Bioethics, Health and Society, Winston-Salem, USA.
| |
Collapse
|
3
|
Azzi J, Wehbi Z, Kobeissy PH, Kerek R. Teaching the science of life: A multidisciplinary educational approach to reproductive technology debates through the lens of developmental biology. Dev Biol 2025; 522:31-39. [PMID: 40073955 DOI: 10.1016/j.ydbio.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/16/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
In vitro fertilization, cryopreservation and pre-implantation genetic testing are transformative reproductive technologies offering hope for individuals facing fertility challenges. Scientifically, understanding the science of developmental biology is essential for comprehending the mechanisms and implications of these technologies. In embryological sciences, biological perspectives identify life as a series of developmental stages ranging from conception to viability, each potentially representing a different 'beginning' of life. However, the concept of ensoulment, rooted in religious and cultural beliefs, introduces a speculative dimension ultimately influencing how legal systems worldwide define and protect human life in the context of reproductive decision-making. Legally, high-profile cases such as 'Sofia Vergara v. Nick Loeb' or 'Quintavalle v. Human Fertilization and Embryology Authority' raise questions about both parental and fetal reproductive rights and consent. This paper highlights the importance of a multidisciplinary approach in developmental biology education for responsible and equitable practices of reproductive technologies. It underscores the importance of incorporating these considerations into the classroom teaching as case study discussions aligned with the DEI approach, to better equip students for the controversies they may encounter in their roles of developmental biologists.
Collapse
Affiliation(s)
- Jovany Azzi
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Byblos, Lebanon
| | - Zeinab Wehbi
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Philippe Hussein Kobeissy
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon.
| | - Racha Kerek
- Department of Biological Sciences, School of Arts and Sciences, Lebanese American University (LAU), Byblos, Lebanon.
| |
Collapse
|
4
|
Beckett M, Franklin S, Rugg-Gunn PJ. Identifying enabling strategies for effective public dialogue in human embryo research. Stem Cell Reports 2025; 20:102498. [PMID: 40315857 DOI: 10.1016/j.stemcr.2025.102498] [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: 01/11/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 05/04/2025] Open
Abstract
Public dialogue is crucial for understanding societal views on human embryo research, and the complexity and sensitivity of this topic require special considerations of how such dialogues are facilitated. Here, we identify enablers of effective dialogue, which can improve the design and delivery of engagement exercises related to embryo research.
Collapse
Affiliation(s)
- Matilda Beckett
- Epigenetics Programme, Babraham Institute, CB22 3AT Cambridge, UK; Department of Sociology, University of Cambridge, CB2 1SB Cambridge, UK
| | - Sarah Franklin
- Department of Sociology, University of Cambridge, CB2 1SB Cambridge, UK
| | - Peter J Rugg-Gunn
- Epigenetics Programme, Babraham Institute, CB22 3AT Cambridge, UK; Loke Centre for Trophoblast Research, University of Cambridge, CB2 3EG Cambridge, UK; Cambridge Stem Cell Institute, University of Cambridge, CB2 0AW Cambridge, UK.
| |
Collapse
|
5
|
Gharibi B, Inge OCK, Rodriguez-Hernandez I, Driscoll PC, Dubois C, Jiang M, Howell M, Skehel JM, Macrae JI, Santos SDM. Post-gastrulation amnioids as a stem cell-derived model of human extra-embryonic development. Cell 2025:S0092-8674(25)00458-1. [PMID: 40378847 DOI: 10.1016/j.cell.2025.04.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 02/25/2025] [Accepted: 04/16/2025] [Indexed: 05/19/2025]
Abstract
The amnion, an extra-embryonic tissue in mammalian embryos, is thought to provide crucial signaling, structural, and nutritional support during pregnancy. Despite its pivotal importance, studying human amnion formation and function has been hampered by the lack of accurate in vitro models. Here, we present an embryonic stem cell-derived 3D model of the post-gastrulation amnion, post-gastrulation amnioids (PGAs), that faithfully recapitulates extra-embryonic development up to 4 weeks post-fertilization, closely mimicking the functional traits of the human amniotic sac. PGAs self-organize, forming the amnion and the yolk sac, and are surrounded by the extra-embryonic mesoderm. Using PGAs, we show that GATA3 is required and sufficient for amniogenesis and that an autoregulatory feedback loop governs amnion formation, whereby extra-embryonic signals promote amnion specification. The reproducibility and scalability of the PGA system, with its precise cellular, structural, and functional integrity, opens avenues for investigating embryo-amnion interactions beyond gastrulation and offers an ideal platform for large-scale pharmacological and clinical studies.
Collapse
Affiliation(s)
- Borzo Gharibi
- Quantitative Stem Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| | - Oliver C K Inge
- Quantitative Stem Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Paul C Driscoll
- Metabolomics, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Ming Jiang
- High-throughput Screening, The Francis Crick Institute, London NW1 1AT, UK
| | - Michael Howell
- High-throughput Screening, The Francis Crick Institute, London NW1 1AT, UK
| | - J Mark Skehel
- Proteomics, The Francis Crick Institute, London NW1 1AT, UK
| | - James I Macrae
- Metabolomics, The Francis Crick Institute, London NW1 1AT, UK
| | - Silvia D M Santos
- Quantitative Stem Cell Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
6
|
Shen L, Li C, Li Y, Guan X, Zou W, Liu J. Imaging technology in tracking the intravital fate of transplanted stem cells. Pharmacol Res 2025; 216:107752. [PMID: 40306602 DOI: 10.1016/j.phrs.2025.107752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/15/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Stem cell therapy emerges as a promising alternative strategy for diseases that currently lack effective treatment options. Investigating the pharmacokinetic properties of stem cells, such as their survival, migration, differentiation, and engraftment dynamics, offers valuable insights for elucidating therapeutic mechanisms, refining treatment protocols, and ultimately enhancing therapeutic efficacy. Moreover, the pharmaceutical research of stem cell products is an essential prerequisite for regulatory approval. This contribution focus on the development of advanced imaging technologies for noninvasive monitoring the intravital fate of implanted stem cells, as well as the advantages and challenges of each imaging approach. Through comprehensive analysis of stem cell metabolic pathway, we identify critical barriers to clinical translation of stem cell therapy. In the end, we discuss future perspectives and opportunities in stem cell tracking and functional assessment.
Collapse
Affiliation(s)
- Liming Shen
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Chengze Li
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yulian Li
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xin Guan
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Wei Zou
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116023, China
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116023, China.
| |
Collapse
|
7
|
Hua L, Peng Y, Yan L, Yuan P, Qiao J. Moving toward totipotency: the molecular and cellular features of totipotent and naive pluripotent stem cells. Hum Reprod Update 2025:dmaf006. [PMID: 40299455 DOI: 10.1093/humupd/dmaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 01/06/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Dissecting the key molecular mechanism of embryonic development provides novel insights into embryogenesis and potential intervention strategies for clinical practices. However, the ability to study the molecular mechanisms of early embryo development in humans, such as zygotic genome activation and lineage segregation, is meaningfully constrained by methodological limitations and ethical concerns. Totipotent stem cells have an extended developmental potential to differentiate into embryonic and extraembryonic tissues, providing a suitable model for studying early embryo development. Recently, a series of ground-breaking results on stem cells have identified totipotent-like cells or induced pluripotent stem cells into totipotent-like cells. OBJECTIVE AND RATIONALE This review followed the PRISMA guidelines, surveys the current works of literature on totipotent, naive, and formative pluripotent stem cells, introduces the molecular and biological characteristics of those stem cells, and gives advice for future research. SEARCH METHODS The search method employed the terms 'totipotent' OR 'naive pluripotent stem cell' OR 'formative pluripotent stem cell' for unfiltered search on PubMed, Web of Science, and Cochrane Library. Papers included were those with information on totipotent stem cells, naive pluripotent stem cells, or formative pluripotent stem cells until June 2024 and were published in the English language. Articles that have no relevance to stem cells, or totipotent, naive pluripotent, or formative pluripotent cells were excluded. OUTCOMES There were 152 records included in this review. These publications were divided into four groups according to the species of the cells included in the studies: 67 human stem cell studies, 70 mouse stem cell studies, 9 porcine stem cell studies, and 6 cynomolgus stem cell studies. Naive pluripotent stem cell models have been established in other species such as porcine and cynomolgus. Human and mouse totipotent stem cells, e.g. human 8-cell-like cells, human totipotent blastomere-like cells, and mouse 2-cell-like cells, have been successfully established and exhibit high developmental potency for both embryonic and extraembryonic contributions. However, the observed discrepancies between these cells and real embryos in terms of epigenetics and transcription suggest that further research is warranted. Our results systematically reviewed the established methods, molecular characteristics, and developmental potency of different naive, formative pluripotent, and totipotent stem cells. Furthermore, we provide a parallel comparison between animal and human models, and offer recommendations for future applications to advance early embryo research and assisted reproduction technologies. WIDER IMPLICATIONS Totipotent cell models provide a valuable resource to understand the underlying mechanisms of embryo development and forge new paths toward future treatment of infertility and regenerative medicine. However, current in vitro cell models exhibit epigenetic and transcriptional differences from in vivo embryos, and many cell models are unstable across passages, thus imperfectly recapitulating embryonic development. In this regard, standardizing and expanding current research on totipotent stem cell models are essential to enhance our capability to resemble and decipher embryogenesis.
Collapse
Affiliation(s)
- Lingyue Hua
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yuyang Peng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Liying Yan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Peng Yuan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics, Beijing, China
| |
Collapse
|
8
|
Wang W, Song AR, Liu HW, Li YK. Enhancing the clinical translation of stem cell models by focusing on standardization and international regulatory cooperation. World J Stem Cells 2025; 17:102788. [PMID: 40308886 PMCID: PMC12038456 DOI: 10.4252/wjsc.v17.i4.102788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/12/2025] [Accepted: 03/10/2025] [Indexed: 04/23/2025] Open
Abstract
The article by Granjeiro et al provided a thorough review of the role of stem cell models in the development of advanced therapy medicinal products. It emphasized the potential of stem cell models to refine preclinical studies and align with regulatory requirements for clinical applications. This article introduced a new perspective on enhancing the transition of stem cell research into clinical practice, focusing on the importance of international regulatory harmonization and the need for standardization in stem cell-based therapies.
Collapse
Affiliation(s)
- Wei Wang
- Department of Traditional Chinese Medicine, Youyang People's Hospital, Chongqing 409800, China
| | - An-Ran Song
- Department of Traditional Chinese Medicine, Youyang People's Hospital, Chongqing 409800, China
| | - Hong-Wen Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Yi-Kai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China.
| |
Collapse
|
9
|
Akita D, Tsukimura N, Kazama T, Takahashi R, Taniguchi Y, Inoue J, Suzuki A, Tanabe N, Seki K, Arai Y, Asano M, Sato S, Hagiwara Y, Kano K, Honda M, Matsumoto T. Regeneration of Two-Walled Infrabony Periodontal Defects in Swine After Buccal Fat Pad-Derived Dedifferentiated Fat Cell Autologous Transplantation. Biomolecules 2025; 15:604. [PMID: 40305349 PMCID: PMC12024700 DOI: 10.3390/biom15040604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
Mature adipocyte-derived dedifferentiated fat (DFAT) cells show proliferative capabilities and multipotency. Given that the buccal fat pad (BFP) serves as a readily available resource for DFAT cell isolation, BFP-derived DFAT (BFP-DFAT) cells are a promising candidate in orofacial tissue engineering. In this research, we assessed the regenerative capacity of the periodontium through autologous BFP-DFAT cell transplantation in adult swine (micro-minipigs; MMPs). The BFP-DFAT cells were transplanted into inflammation-inducing two-walled infrabony periodontal defects located on the mesial of the second mandibular premolar (n = 6). Twelve weeks post-transplantation, a remarkable attachment gain was noted in the DFAT group, based on probing depths and clinical attachment levels. Histological and immunohistochemical analyses indicated new continuous cellular cementum and alveolar bone formation within the created infrabony defect. Well-organized periodontal ligament-like fibers were embedded between newly formed cementum and the alveolar bone. Histometric analysis demonstrated that the DFAT group had a 2.2-fold increase in new alveolar bone length and a 2.2-fold enhancement in vascularization than those in the control group. Except for minor inflammation in the lungs, no teratomas were detected in the recipient MMPs. BFP-DFAT cells significantly enhanced periodontal tissue regeneration, thus representing an optimal source for tissue engineering applications in dentistry.
Collapse
Affiliation(s)
- Daisuke Akita
- Department of Partial Denture Prosthodontics, Nihon University School of Dentistry, Tokyo 101-8310, Japan;
| | - Naoki Tsukimura
- Division of General Dentistry, The Nippon Dental University School of Life Dentistry, Tokyo 102-8158, Japan;
| | - Tomohiko Kazama
- Division of Cell Regeneration and Transplantation, Department of Functional Morphology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (T.K.); (T.M.)
| | - Rie Takahashi
- Section of Laboratory Animals, Nihon University School of Medicine, Tokyo 173-8601, Japan; (R.T.); (Y.T.)
| | - Yoshiki Taniguchi
- Section of Laboratory Animals, Nihon University School of Medicine, Tokyo 173-8601, Japan; (R.T.); (Y.T.)
| | - Jin Inoue
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry, Tokyo 101-8310, Japan; (J.I.); (A.S.); (N.T.)
| | - Ayana Suzuki
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry, Tokyo 101-8310, Japan; (J.I.); (A.S.); (N.T.)
| | - Nodoka Tanabe
- Division of Applied Oral Sciences, Nihon University Graduate School of Dentistry, Tokyo 101-8310, Japan; (J.I.); (A.S.); (N.T.)
| | - Keisuke Seki
- Department of Comprehensive Dentistry and Clinical Education, Nihon University School of Dentistry, Tokyo 101-8310, Japan;
| | - Yoshinori Arai
- Department of Oral and Maxillofacial Radiology, Nihon University School of Dentistry, Tokyo 101-8310, Japan;
| | - Masatake Asano
- Department of Pathology, Nihon University School of Dentistry, Tokyo 101-8310, Japan;
| | - Shuichi Sato
- Department of Periodontology, Nihon University School of Dentistry, Tokyo 101-8310, Japan;
| | - Yoshiyuki Hagiwara
- Department of Partial Denture Prosthodontics, Nihon University School of Dentistry, Tokyo 101-8310, Japan;
| | - Koichiro Kano
- Laboratory of Cell and Tissue Biology, College of Bioresource Science, Nihon University, Fujisawa 252-0880, Japan;
| | - Masaki Honda
- Department of Oral Anatomy, Aichi Gakuin University School of Dentistry, Nagoya 464-8650, Japan;
| | - Taro Matsumoto
- Division of Cell Regeneration and Transplantation, Department of Functional Morphology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (T.K.); (T.M.)
| |
Collapse
|
10
|
Hosseini N, Kazeminejad E, Oladnabi M, Khosravi A. Isolation and characterization of a new SHED cell line as a standard source for stem cell research and clinical translation. Tissue Cell 2025; 93:102649. [PMID: 39637488 DOI: 10.1016/j.tice.2024.102649] [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] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND AND AIMS Stem cells from human exfoliated deciduous teeth (SHED) are multi-potent mesenchymal stem/stromal cells (MSCs) and are inspected a favorable, non-invasive source beneficial to stem cell-mediated regeneration of damaged tissues. Our aim was to establish and characterize a non-immortalized SHED cell line as an accessible resource and novel platform for stem cell research and tissue regeneration studies. METHODS A Healthy exfoliated deciduous molar was extracted from a 12-year-old girl and shipped to an animal cell culture laboratory. Outgrowing primary cells from explanted small pulp tissues were monitored daily and characterized after passage 3 both morphologically and functionally. The SHED cell line was characterized by calculation of doubling time, cytogenetic analyses, STR analysis, adherence to cell culture flasks under standard cell culture media, and immunophenotypic analysis of specific MSC markers (CD90+, CD73+, CD34- and CD45-) using flow cytometry method. Differentiation potential to osteoblast, adipocyte, and chondrocyte was evaluated under standard differentiation media Expression of OCT-4 and NANOG genes was also assessed using RT-PCR method. RESULTS After the third day, SHED cells were visible. SHED cells were subcultured when they reached 90 % confluence after approximately 17 days. The doubling time of SHED cells was forty seven hours. SHED immunophenotyping showed the high expression level of CD90 (99.2 %) and CD73 (45.9 %), and approximately no expression of CD34 (0.079 %) and CD45 (0.19 %). The human origin, female gender and chromosomal normality of SHED cells was confirmed by cytogenetic analysis. The STR matching analysis showed that SHED cells are well-identified and authentic. No genetic instability and cross-contamination were observed in SHED cells. CONCLUSIONS This study provides a new SHED cell line with a normal karyotype and all the characteristics of MSCs, which can be used as a favorable model cell line in biomedical research and a promising source for clinical translation.
Collapse
Affiliation(s)
- Niloufar Hosseini
- Department of Medical Biotechnology, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ezatolah Kazeminejad
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Dental Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Morteza Oladnabi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Gorgan Congenital Malformations Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Ayyoob Khosravi
- Stem Cell Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Department of Molecular Medicine, Faculty of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran.
| |
Collapse
|
11
|
Gu B, Li M, Li D, Huang K. CRISPR-Cas9 Targeting PCSK9: A Promising Therapeutic Approach for Atherosclerosis. J Cardiovasc Transl Res 2025; 18:424-441. [PMID: 39804565 DOI: 10.1007/s12265-024-10587-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 12/28/2024] [Indexed: 05/01/2025]
Abstract
CRISPR-Cas9 gene editing technology, as an innovative biomedical tool, holds significant potential in the prevention and treatment of atherosclerosis. By precisely editing key genes such as PCSK9, CRISPR-Cas9 offers the possibility of long-term regulation of low-density lipoprotein cholesterol (LDL-C), which may reduce the risk of cardiovascular diseases. Early clinical studies of gene editing therapies like VERVE-101 have yielded encouraging results, highlighting both the feasibility and potential efficacy of this technology. However, clinical applications still face challenges such as off-target effects, immunogenicity, and long-term safety. Future research should focus on enhancing the specificity and efficiency of gene editing, optimizing delivery systems, and improving personalized treatment strategies. Additionally, the establishment of ethical and legal regulatory frameworks will be critical for the safe adoption of this technology. With the continued advancement of gene editing technology, CRISPR-Cas9 may become an important tool for treating atherosclerosis and other complex diseases.
Collapse
Affiliation(s)
- Bin Gu
- Department of Cardiology, Affiliated Hospital of Southwest Medical University, No.1 Section 1, Xiang Lin Road, Longmatan District, Luzhou, Sichuan, 646000, China
| | - Min Li
- Department of Cardiology, Neijiang Dongxing District People's Hospital, Neijiang, Sichuan, 641300, China
| | - Dan Li
- Department of Cardiology, Neijiang Dongxing District People's Hospital, Neijiang, Sichuan, 641300, China
| | - Kaisen Huang
- Department of Cardiology, Affiliated Hospital of Southwest Medical University, No.1 Section 1, Xiang Lin Road, Longmatan District, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
12
|
Wang YC, Cheng JB, Feng ML. Requirements for standardizing the assessment of mesenchymal stem cell therapy and its effects on osteoarthritis. World J Orthop 2025; 16:104451. [PMID: 40124721 PMCID: PMC11924026 DOI: 10.5312/wjo.v16.i3.104451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 03/12/2025] Open
Abstract
Publications of Soufan et al and Kristjánsson et al in the World Journal of Orthopedics on mesenchymal stem cell (MSC) therapy for osteoarthritis (OA) represent a significant exploration of regenerative medicine's potential in OA treatment. In their research, it is highlighted that MSCs can alleviate OA symptoms and even regenerate cartilage, potentially reversing the disease. They also compared the efficacy of three MSC subtypes, emphasizing the therapeutic advantages of adipose-derived MSCs. MSC injections, a novel and less invasive alternative to traditional treatments such as chondrocyte transplantation or arthroplasty, have a low cost, low risks, and favorable outcomes, presenting a promising approach for OA patients. Additionally, we stressed that the efficacy evaluation criteria, heterogeneity, safety, and other factors must be carefully considered to further advance the clinical translation of MSC therapy for OA.
Collapse
Affiliation(s)
- Yu-Chen Wang
- Department of Orthopaedic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jing-Bo Cheng
- Department of Orthopaedic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Ming-Li Feng
- Department of Orthopaedic Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
13
|
Davidson SM, Andreadou I, Antoniades C, Bartunek J, Basso C, Brundel BJJM, Byrne RA, Chiva-Blanch G, da Costa Martins P, Evans PC, Girão H, Giricz Z, Gollmann-Tepeköylü C, Guzik T, Gyöngyösi M, Hübner N, Joner M, Kleinbongard P, Krieg T, Liehn E, Madonna R, Maguy A, Paillard M, Pesce M, Petersen SE, Schiattarella GG, Sluijter JPG, Steffens S, Streckfuss-Bömeke K, Thielmann M, Tucker A, Van Linthout S, Wijns W, Wojta J, Wu JC, Perrino C. Opportunities and challenges for the use of human samples in translational cardiovascular research: a scientific statement of the ESC Working Group on Cellular Biology of the Heart, the ESC Working Group on Cardiovascular Surgery, the ESC Council on Basic Cardiovascular Science, the ESC Scientists of Tomorrow, the European Association of Percutaneous Cardiovascular Interventions of the ESC, and the Heart Failure Association of the ESC. Cardiovasc Res 2025:cvaf023. [PMID: 40084813 DOI: 10.1093/cvr/cvaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 03/16/2025] Open
Abstract
Animal models offer invaluable insights into disease mechanisms but cannot entirely mimic the variability and heterogeneity of human populations, nor the increasing prevalence of multi-morbidity. Consequently, employing human samples-such as whole blood or fractions, valvular and vascular tissues, myocardium, pericardium, or human-derived cells-is essential for enhancing the translational relevance of cardiovascular research. For instance, myocardial tissue slices, which preserve crucial structural and functional characteristics of the human heart, can be used in vitro to examine drug responses. Human blood serves as a rich source of biomarkers, including extracellular vesicles, various types of RNA (miRNA, lncRNA, and circRNAs), circulating inflammatory cells, and endothelial colony-forming cells, facilitating detailed studies of cardiovascular diseases. Primary cardiomyocytes and vascular cells isolated from human tissues are invaluable for mechanistic investigations in vitro. In cases where these are unavailable, human induced pluripotent stem cells serve as effective substitutes, albeit with specific limitations. However, the use of human samples presents challenges such as ethical approvals, tissue procurement and storage, variability in patient genetics and treatment regimens, and the selection of appropriate control samples. Biobanks are central to the efficient use of these scarce and valuable resources. This scientific statement discusses opportunities to implement the use of human samples for cardiovascular research within specific clinical contexts, offers a practical framework for acquiring and utilizing different human materials, and presents examples of human sample applications for specific cardiovascular diseases, providing a valuable resource for clinicians, translational and basic scientists engaged in cardiovascular research.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Ioanna Andreadou
- School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Antoniades
- RDM Division of Cardiovascular Medicine, Acute Multidisciplinary Imaging and Interventional Centre, University of Oxford, Headley Way, Headington, Oxford OX3 9DU, UK
| | - Jozef Bartunek
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, Cardiovascular Pathology, University of Padua, Padua, Italy
| | - Bianca J J M Brundel
- Physiology, Amsterdam UMC Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
| | - Robert A Byrne
- Cardiovascular Research Institute Dublin, Mater Private Network, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gemma Chiva-Blanch
- Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Network Research Centre on Obesity and Nutrition Physiopathology, Instituto de Salud Carlos III, Madrid, Spain
| | - Paula da Costa Martins
- Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paul C Evans
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Henrique Girão
- Center for Innovative Biomedicine and Biotechnology, Clinical Academic Centre of Coimbra, Faculty of Medicine, University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Coimbra, Portugal
| | - Zoltan Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Can Gollmann-Tepeköylü
- Department for Cardiac Surgery, Cardiac Regeneration Research, Medical University of Innsbruck, Anichstraße 35 A, 6020 Innsbruck, Austria
| | - Tomasz Guzik
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Norbert Hübner
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
- Charite-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Michael Joner
- Department of Cardiology, German Heart Center Munich, Technical University of Munich, Lazarettstrasse 36, 80636 Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Petra Kleinbongard
- Faculty of Medicine University of Duisburg-Essen, Institute of Pathophysiology, Duisburg-Essen, Germany
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Elisa Liehn
- Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rosalinda Madonna
- Cardiology Division, Department of Pathology, University of Pisa, Pisa, Italy
| | - Ange Maguy
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Melanie Paillard
- Laboratoire CarMeN-IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, Univ-Lyon, 69500 Bron, France
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Department of Aerospace and Mechanical Engineering, Politecnico di Torino, Italy
- Department of Cell Biology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
- Health Data Research UK, London, UK
- Alan Turing Institute, London, UK
| | - Gabriele G Schiattarella
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
- Deutsches Herzzentrum der Charité (DHZC), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen, Germany and German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Matthias Thielmann
- West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Art Tucker
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité, BIH Center for Regenerative Therapies, Universitätmedizin Berlin, Berlin, Germany
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
| | - William Wijns
- The Lambe Institute for Translational Research and Curam, University of Galway, Galway, Ireland
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Core Facilities, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
14
|
Xie H, An C, Bai B, Luo J, Sun N, Ci B, Jin L, Mo P, Lu Y, Zhong K, Yu Y, Tan T, Li R, Fan Y. Modeling early gastrulation in human blastoids with DNA methylation patterns of natural blastocysts. Cell Stem Cell 2025; 32:409-425.e8. [PMID: 39814012 DOI: 10.1016/j.stem.2024.12.010] [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: 12/21/2023] [Revised: 08/27/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Blastoids are a promising model for studying early human embryogenesis, but current models have limitations in post-implantation development and lack comprehensive epigenetic assessments, especially regarding genomic imprinting. These issues can lead to failures in accurately modeling early embryonic development. In this study, we developed a high-fidelity blastoid model using 4 chemicals + leukemia inhibitory factor (LIF) (4CL) naive human pluripotent stem cells (hPSCs) (4CL blastoids). 4CL blastoids closely resemble human blastocysts in morphology and transcriptional profiles, exhibiting similar DNA methylation and gene imprinting patterns. By extending the 3D culture to 14 days, these blastoids mimic early gastrulation, demonstrating the specification and migration of cells. They also show the transcriptional signature of hemogenic angioblast (HAB) cells at Carnegie stage 6 (CS6). This model bridges pre- and post-implantation stages, offering valuable insights into early tissue formation and human development.
Collapse
Affiliation(s)
- Han Xie
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Chenrui An
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Bing Bai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Jiajia Luo
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Nianqin Sun
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Baiquan Ci
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Long Jin
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Peiting Mo
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yawen Lu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Ke Zhong
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing 100191, China.
| | - Tao Tan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, Yunnan, China.
| | - Rong Li
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.
| | - Yong Fan
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
15
|
Garcia‐Aponte OF, Kahlenberg S, Kouroupis D, Egger D, Kasper C. Effects of Hydrogels on Mesenchymal Stem/Stromal Cells Paracrine Activity and Extracellular Vesicles Production. J Extracell Vesicles 2025; 14:e70057. [PMID: 40091440 PMCID: PMC11911545 DOI: 10.1002/jev2.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are a valuable source of paracrine factors, as they have a remarkable secretory capacity, and there is a sizeable knowledge base to develop industrial and clinical production protocols. Promising cell-free approaches for tissue regeneration and immunomodulation are driving research towards secretome applications, among which extracellular vesicles (EVs) are steadily gaining attention. However, the manufacturing and application of EVs is limited by insufficient yields, knowledge gaps, and low standardization. Facing these limitations, hydrogels represent a versatile three-dimensional (3D) culture platform that can incorporate extracellular matrix (ECM) components to mimic the natural stem cell environment in vitro; via these niche-mimicking properties, hydrogels can regulate MSCs' morphology, adhesion, proliferation, differentiation and secretion capacities. However, the impact of the hydrogel's architectural, biochemical and biomechanical properties on the production of EVs remains poorly understood, as the field is still in its infancy and the interdependency of culture parameters compromises the comparability of the studies. Therefore, this review summarizes and discusses the reported effects of hydrogel encapsulation and culture on the secretion of MSC-EVs. Considering the effects of cell-material interactions on the overall paracrine activity of MSCs, we identify persistent challenges from low standardization and process control, and outline future paths of research, such as the synergic use of hydrogels and bioreactors to enhance MSC-EV generation.
Collapse
Affiliation(s)
- Oscar Fabian Garcia‐Aponte
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Simon Kahlenberg
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
- Diabetes Research Institute & Cell Transplant Center, Miller School of MedicineUniversity of MiamiMiamiFloridaUSA
| | - Dominik Egger
- Institute of Cell Biology and BiophysicsLeibniz University HannoverHannoverGermany
| | - Cornelia Kasper
- Department of Biotechnology and Food Science, Institute of Cell and Tissue Culture TechnologiesUniversity of Natural Resources and Life SciencesViennaAustria
| |
Collapse
|
16
|
Pan Y, Li L, Cao N, Liao J, Chen H, Zhang M. Advanced nano delivery system for stem cell therapy for Alzheimer's disease. Biomaterials 2025; 314:122852. [PMID: 39357149 DOI: 10.1016/j.biomaterials.2024.122852] [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: 06/20/2024] [Revised: 09/10/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
Alzheimer's Disease (AD) represents one of the most significant neurodegenerative challenges of our time, with its increasing prevalence and the lack of curative treatments underscoring an urgent need for innovative therapeutic strategies. Stem cells (SCs) therapy emerges as a promising frontier, offering potential mechanisms for neuroregeneration, neuroprotection, and disease modification in AD. This article provides a comprehensive overview of the current landscape and future directions of stem cell therapy in AD treatment, addressing key aspects such as stem cell migration, differentiation, paracrine effects, and mitochondrial translocation. Despite the promising therapeutic mechanisms of SCs, translating these findings into clinical applications faces substantial hurdles, including production scalability, quality control, ethical concerns, immunogenicity, and regulatory challenges. Furthermore, we delve into emerging trends in stem cell modification and application, highlighting the roles of genetic engineering, biomaterials, and advanced delivery systems. Potential solutions to overcome translational barriers are discussed, emphasizing the importance of interdisciplinary collaboration, regulatory harmonization, and adaptive clinical trial designs. The article concludes with reflections on the future of stem cell therapy in AD, balancing optimism with a pragmatic recognition of the challenges ahead. As we navigate these complexities, the ultimate goal remains to translate stem cell research into safe, effective, and accessible treatments for AD, heralding a new era in the fight against this devastating disease.
Collapse
Affiliation(s)
- Yilong Pan
- Department of Cardiology, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| | - Long Li
- Department of Neurosurgery, First Hospital of China Medical University, Liaoning, 110001, China.
| | - Ning Cao
- Army Medical University, Chongqing, 400000, China
| | - Jun Liao
- Institute of Systems Biomedicine, Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Huiyue Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Liaoning, 110001, China.
| | - Meng Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Liaoning, 110004, China.
| |
Collapse
|
17
|
Kim MS, Yoon S, Choi J, Kim YJ, Lee G. Stem Cell-Based Approaches in Parkinson's Disease Research. Int J Stem Cells 2025; 18:21-36. [PMID: 38449089 PMCID: PMC11867902 DOI: 10.15283/ijsc23169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative condition characterized by the loss of midbrain dopaminergic neurons, leading to motor symptoms. While current treatments provide limited relief, they don't alter disease progression. Stem cell technology, involving patient-specific stem cell-derived neurons, offers a promising avenue for research and personalized regenerative therapies. This article reviews the potential of stem cell-based research in PD, summarizing ongoing efforts, their limitations, and introducing innovative research models. The integration of stem cell technology and advanced models promises to enhance our understanding and treatment strategies for PD.
Collapse
Affiliation(s)
- Min Seong Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Subeen Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Jiwoo Choi
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Yong Jun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Korea
- KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, Korea
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Solomon Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Bolideei M, Barzigar R, Gahrouei RB, Mohebbi E, Haider KH, Paul S, Paul MK, Mehran MJ. Applications of Gene Editing and Nanotechnology in Stem Cell-Based Therapies for Human Diseases. Stem Cell Rev Rep 2025:10.1007/s12015-025-10857-0. [PMID: 40014250 DOI: 10.1007/s12015-025-10857-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Stem cell research is a dynamic and fast-advancing discipline with great promise for the treatment of diverse human disorders. The incorporation of gene editing technologies, including ZFNs, TALENs, and the CRISPR/Cas system, in conjunction with progress in nanotechnology, is fundamentally transforming stem cell therapy and research. These innovations not only provide a glimmer of optimism for patients and healthcare practitioners but also possess the capacity to radically reshape medical treatment paradigms. Gene editing and nanotechnology synergistically enhance stem cell-based therapies' precision, efficiency, and applicability, offering transformative potential for treating complex diseases and advancing regenerative medicine. Nevertheless, it is important to acknowledge that these technologies also give rise to ethical considerations and possible hazards, such as inadvertent genetic modifications and the development of genetically modified organisms, therefore creating a new age of designer infants. This review emphasizes the crucial significance of gene editing technologies and nanotechnology in the progress of stem cell treatments, particularly for degenerative pathologies and injuries. It emphasizes their capacity to restructure and comprehensively revolutionize medical treatment paradigms, providing fresh hope and optimism for patients and healthcare practitioners.
Collapse
Affiliation(s)
- Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Rambod Barzigar
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India
| | - Razieh Bahrami Gahrouei
- Department of Pharmacy PES College, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| | - Elham Mohebbi
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, Springfield, IL, USA
| | - Khawaja Husnain Haider
- Sulaiman AlRajhi Medical School, Al Bukayriyah, AlQaseem, 52726, Kingdom of Saudi Arabia
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA.
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Mohammad Javad Mehran
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India.
| |
Collapse
|
19
|
Major RM, Juengst ET. Prenatal gene editing for neurodevelopmental diseases: Ethical considerations. Am J Hum Genet 2025; 112:201-214. [PMID: 39879986 PMCID: PMC11866956 DOI: 10.1016/j.ajhg.2025.01.003] [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: 09/16/2024] [Revised: 01/02/2025] [Accepted: 01/02/2025] [Indexed: 01/31/2025] Open
Abstract
Neurodevelopmental diseases (NDDs) are notoriously difficult to treat because clinical symptoms stem from developmental processes that begin before birth. Prenatal gene editing could fill the treatment gap for NDDs by targeting and permanently correcting the genetic variants that underlie these pathogenic developmental processes. At the same time, there is a risk of unintended edits to the fetus or the pregnant person that could result in serious adverse consequences that are difficult, if not impossible, to undo. This raises ethical concerns that make the development of prenatal gene editing especially challenging. To date, there are no frameworks for considering the steps necessary for an ethical path forward for prenatal gene editing specifically. The 60-year history of in utero therapy has included the development of frameworks for other therapies that can provide starting points for addressing the unique issues of prenatal gene editing. We identified 12 themes from 17 ethical frameworks, literature, consensus statements, and government reports on prenatal interventions that could set precedents for prenatal gene editing interventions. In considering these alongside current criteria for postnatal gene therapies for NDDs, we discuss a path forward for prenatal gene editing interventions of NDDs.
Collapse
Affiliation(s)
- Rami M Major
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Eric T Juengst
- Department of Social Medicine and Center for Bioethics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
20
|
Tariq H, Bukhari SZ, An R, Dong J, Ihsan A, Younis MR. Stem cell-derived exosome delivery systems for treating atherosclerosis: The new frontier of stem cell therapy. Mater Today Bio 2025; 30:101440. [PMID: 39866781 PMCID: PMC11758955 DOI: 10.1016/j.mtbio.2024.101440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide. As a chronic inflammatory disease with a complicated pathophysiology marked by abnormal lipid metabolism and arterial plaque formation, atherosclerosis is a major contributor to CVDs and can induce abrupt cardiac events. The discovery of exosomes' role in intercellular communication has sparked a great deal of interest in them recently. Exosomes are involved in strategic phases of the onset and development of atherosclerosis because they have been identified to control pathophysiologic pathways including inflammation, angiogenesis, or senescence. This review investigates the potential role of stem cell-derived exosomes in atherosclerosis management. We briefly introduced atherosclerosis and stem cell therapy including stem cell-derived exosomes. The biogenesis of exosomes along with their secretion and isolation have been elaborated. The design engineering of exosomes has been summarized to present how drug loading and surface modification with targeting ligands can improve the therapeutic and targeting capacity of exosomes, demonstrating atheroprotective action. Moreover, the mechanism of action (endothelial dysfunction, reduction of dyslipidemia, macrophage polarization, vascular calcification, and angiogenesis) of drug-loaded exosomes to treat atherosclerosis has been discussed in detail. In the end, a comparative and balanced viewpoint has been given regarding the current challenges and potential solutions to advance exosome engineering for cardiovascular therapeutic applications.
Collapse
Affiliation(s)
- Hassan Tariq
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| | - Syeda Zunaira Bukhari
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Ruibing An
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Jian Dong
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Muhammad Rizwan Younis
- Institute of Optical Functional Materials for Biomedical Imaging, School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Science, Taian, Shandong, 271016, PR China
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA, 90095, USA
- Department of Molecular, Cell and Developmental Biology, University of California - Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
21
|
Narasimha RB, Shreya S, Jayabal VA, Yadav V, Rath PK, Mishra BP, Kancharla S, Kolli P, Mandadapu G, Kumar S, Mohanty AK, Jena MK. Stem Cell Therapy for Diseases of Livestock Animals: An In-Depth Review. Vet Sci 2025; 12:67. [PMID: 39852942 PMCID: PMC11768649 DOI: 10.3390/vetsci12010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
Stem cells are unique, undifferentiated cells that have the ability to both replicate themselves and develop into specialized cell types. This dual capability makes them valuable in the development of regenerative medicine. Current development in stem cell research has widened their application in cell therapy, drug discovery, reproductive cloning in animals, and cell models for various diseases. Although there are substantial studies revealing the treatment of human degenerative diseases using stem cells, this is yet to be explored in livestock animals. Many diseases in livestock species such as mastitis, laminitis, neuromuscular disorders, autoimmune diseases, and some debilitating diseases are not covered completely by the existing drugs and treatment can be improved by using different types of stem cells like embryonic stem cells, adult stem cells, and induced pluripotent stem cells. This review mainly focuses on the use of stem cells for disease treatment in livestock animals. In addition to the diseases mentioned, the potential of stem cells can be helpful in wound healing, skin disease therapy, and treatment of some genetic disorders. This article explores the potential of stem cells from various sources in the therapy of livestock diseases and also their role in the conservation of endangered species as well as disease model preparation. Moreover, the future perspectives and challenges associated with the application of stem cells in livestock are discussed. Overall, the transformative impact of stem cell research on the livestock sector is comprehensively studied which will help researchers to design future research work on stem cells related to livestock diseases.
Collapse
Affiliation(s)
- Raghavendra B. Narasimha
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| | - Singireddy Shreya
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| | - Vijay Anand Jayabal
- Department of Animal Biotechnology, Madras Veterinary College, Tamil Nadu Veterinary and Animal Sciences University, Chennai 600051, Tamil Nadu, India;
| | - Vikas Yadav
- Department of Clinical Sciences, Clinical Research Centre, Skåne University Hospital, Lund University, SE 20213 Malmö, Sweden
| | - Prasana Kumar Rath
- College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar 751003, Odisha, India; (P.K.R.); (B.P.M.)
| | - Bidyut Prava Mishra
- College of Veterinary Science and AH, Odisha University of Agriculture and Technology, Bhubaneswar 751003, Odisha, India; (P.K.R.); (B.P.M.)
| | - Sudhakar Kancharla
- Devansh Lab Werks, 234 Aquarius Drive, Homewood, AL 35209, USA; (S.K.); (G.M.)
| | - Prachetha Kolli
- Microgen Health Inc., 14225 Sullyfield Cir Suite E, Chantilly, VA 20151, USA;
| | - Gowtham Mandadapu
- Devansh Lab Werks, 234 Aquarius Drive, Homewood, AL 35209, USA; (S.K.); (G.M.)
| | - Sudarshan Kumar
- Cell, Molecular and Proteomics Lab, Animal Biotechnology Centre, ICAR-National Dairy Research Institute (ICAR-NDRI), Karnal 132001, Haryana, India;
| | - Ashok Kumar Mohanty
- ICAR-Central Institute for Research on Cattle (ICAR-CIRC), Meerut 250001, Uttar Pradesh, India;
| | - Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India; (R.B.N.); (S.S.)
| |
Collapse
|
22
|
Wang Z, Zhao F, Lang H, Ren H, Zhang Q, Huang X, He C, Xu C, Tan C, Ma J, Duan S, Wang Z. Organoids in skin wound healing. BURNS & TRAUMA 2025; 13:tkae077. [PMID: 39759541 PMCID: PMC11697111 DOI: 10.1093/burnst/tkae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/30/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025]
Abstract
Stem cells (SCs) can self-replicate and differentiate into multiple lineages. Organoids, 3D cultures derived from SCs, can replicate the spatial structure and physiological characteristics of organs in vitro. Skin organoids can effectively simulate the physiological structure and function of skin tissue, reliably restoring the natural skin ecology in various in vitro environments. Skin organoids have been employed extensively in skin development and pathology research, offering valuable insights for drug screening. Moreover, they play crucial roles in skin regeneration and tissue repair. This in-depth review explores the construction and applications of skin organoids in wound healing, with a focus on their construction process, including skin appendage integration, and significant advancements in wound-healing research.
Collapse
Affiliation(s)
- Zitong Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Hongxin Lang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory of Stem Cell and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang, Liaoning 110013, China
| | - Haiyue Ren
- Department of Pathology, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No. 1 Hospital), No. 215 Zhongshan Street, Wuhan, Hubei 430022, China
| | - Qiqi Zhang
- Department of Pathology, Chengdu Third People's Hospital, No. 82 Qinglong Street, Chengdu, Sichuan 610031, China
| | - Xing Huang
- Department of Anaesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yantaxi Road, Xi'an, Shanxi 710061, China
| | - Cai He
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chengcheng Xu
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Chiyu Tan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Jiajie Ma
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Shu Duan
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang, Liaoning 110004, China
| |
Collapse
|
23
|
Taei A, Sajadi FS, Salahi S, Enteshari Z, Falah N, Shiri Z, Abasalizadeh S, Hajizadeh-Saffar E, Hassani SN, Baharvand H. The cell replacement therapeutic potential of human pluripotent stem cells. Expert Opin Biol Ther 2025; 25:47-67. [PMID: 39679436 DOI: 10.1080/14712598.2024.2443079] [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: 08/21/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION The remarkable ability of human pluripotent stem cells (hPSCs) to differentiate into specialized cells of the human body emphasizes their immense potential in treating various diseases. Advances in hPSC technology are paving the way for personalized and allogeneic cell-based therapies. The first-in-human studies showed improved treatment of diseases with no adverse effects, which encouraged the industrial production of this type of medicine. To ensure the quality, safety and efficacy of hPSC-based products throughout their life cycle, it is important to monitor and control their clinical translation through good practices (GxP) regulations. Understanding these rules in advance will help ensure that the industrial development of hPSC-derived products for widespread clinical implementation is feasible and progresses rapidly. AREAS COVERED In this review, we discuss the key translational obstacles of hPSCs, outline the current hPSC-based clinical trials, and present a workflow for putative clinical hPSC-based products. Finally, we highlight some future therapeutic opportunities for hPSC-derivatives. EXPERT OPINION hPSC-based products continue to show promise for the treatment of a variety of diseases. While clinical trials support the relative safety and efficacy of hPSC-based products, further investigation is required to explore the clinical challenges and achieve exclusive regulations for hPSC-based cell therapies.
Collapse
Affiliation(s)
- Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fatemeh-Sadat Sajadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Sarvenaz Salahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Enteshari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasrin Falah
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Shiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeed Abasalizadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
24
|
Gantner CW, Weatherbee BAT, Wang Y, Zernicka-Goetz M. Assembly of a stem cell-derived human postimplantation embryo model. Nat Protoc 2025; 20:67-91. [PMID: 39261744 DOI: 10.1038/s41596-024-01042-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 06/24/2024] [Indexed: 09/13/2024]
Abstract
The embryonic and extraembryonic tissue interactions underlying human embryogenesis at implantation stages are not currently understood. We have generated a pluripotent stem cell-derived model that mimics aspects of peri-implantation development, allowing tractable experimentation otherwise impossible in the human embryo. Activation of the extraembryonic lineage-specific transcription factors GATA6 and SOX17 (hypoblast factors) or GATA3 and TFAP2C (encoding AP2γ; trophoblast factors) in human embryonic stem (ES) cells drive conversion to extraembryonic-like cells. When combined with wild-type ES cells, self-organized embryo-like structures form in the absence of exogenous factors, termed human inducible embryoids (hiEmbryoids). The epiblast-like domain of hiEmbryoids polarizes and differentiates in response to extraembryonic-secreted extracellular matrix and morphogen cues. Extraembryonic mesenchyme, amnion and primordial germ cells are specified in hiEmbryoids in a stepwise fashion. After establishing stable inducible ES lines and converting ES cells to RSeT culture media, the protocol takes 7-10 d to generate hiEmbryoids. Generation of hiEmbryoids can be performed by researchers with basic expertise in stem cell culture.
Collapse
Affiliation(s)
- Carlos W Gantner
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Bailey A T Weatherbee
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Yuntao Wang
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Magdalena Zernicka-Goetz
- Mammalian Embryo and Stem Cell Group, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Stem Cell Embryo Models Group, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
25
|
Yazew BG, Alemu BW, Walle TA. Factors associated with knowledge and practice of breast self-examination among female governmental school teachers in Gondar Town, Northwest Ethiopia, 2019. Front Oncol 2024; 14:1481714. [PMID: 39777343 PMCID: PMC11703965 DOI: 10.3389/fonc.2024.1481714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
Background Breast cancer is the most common cancer among women globally, and early detection through breast self-examination can improve survival rates. However, this practice is limited in developing countries like Ethiopia. Objective This study aimed to assess the factors influencing knowledge and practices related to breast self-examination among female governmental school teachers in Gondar Town, Northwest Ethiopia. Methods A cross-sectional study was conducted from May 1 to 30, 2019, with 422 female teachers in Gondar metropolis governmental schools selected through simple random sampling. Data were collected using a self-administered questionnaire and analyzed using EPI INFO version 7 and SPSS version 20. Result A total of 415 female teachers participated in the study, with a response rate of 98.3%. The mean age of respondents was 38.64 years. Only 41.9% had good knowledge of breast self-examination, while 14.5% reported good practice. Factors associated with knowledge included secondary education, higher degrees, and exposure to information. Factors influencing practice included having a degree or higher education and prior experience with breast self-examination. Conclusion and recommendation The study revealed low levels of knowledge and practice of breast self-examination among female teachers. Educational level was significantly associated with both knowledge and practice. Recommendations include implementing health education campaigns, organizing events like breast cancer awareness days, and forming support groups in schools to promote awareness and encourage regular breast self-examination among female teachers in Ethiopia.
Collapse
Affiliation(s)
| | - Biresaw Wassihun Alemu
- Department of Midwifery, College of Medicine and Health Sciences, Injibara University, Injibara, Ethiopia
| | - Tarkie Abebe Walle
- Department of Surgical Nursing, School of Nursing, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
26
|
Wong HPN, Selvakumar SV, Loh PY, Liau JYJ, Liau MYQ, Shelat VG. Ethical frontiers in liver transplantation. World J Transplant 2024; 14:96687. [PMID: 39697458 PMCID: PMC11438941 DOI: 10.5500/wjt.v14.i4.96687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024] Open
Abstract
Liver transplantation represents a pivotal intervention in the management of end-stage liver disease, offering a lifeline to countless patients. Despite significant strides in surgical techniques and organ procurement, ethical dilemmas and debates continue to underscore this life-saving procedure. Navigating the ethical terrain surrounding this complex procedure is hence paramount. Dissecting the nuances of ethical principles of justice, autonomy and beneficence that underpin transplant protocols worldwide, we explore the modern challenges that plaques the world of liver transplantation. We investigate the ethical dimensions of organ transplantation, focusing on allocation, emerging technologies, and decision-making processes. PubMed, Scopus, Web of Science, Embase and Central were searched from database inception to February 29, 2024 using the following keywords: "liver transplant", "transplantation", "liver donation", "liver recipient", "organ donation" and "ethics". Information from relevant articles surrounding ethical discussions in the realm of liver transplantation, especially with regards to organ recipients and allocation, organ donation, transplant tourism, new age technologies and developments, were extracted. From the definition of death to the long term follow up of organ recipients, liver transplantation has many ethical quandaries. With new transplant techniques, societal acceptance and perceptions also play a pivotal role. Cultural, religious and regional factors including but not limited to beliefs, wealth and accessibility are extremely influential in public attitudes towards donation, xenotransplantation, stem cell research, and adopting artificial intelligence. Understanding and addressing these perspectives whilst upholding bioethical principles is essential to ensure just distribution and fair allocation of resources. Robust regulatory oversight for ethical sourcing of organs, ensuring good patient selection and transplant techniques, and high-quality long-term surveillance to mitigate risks is essential. Efforts to promote equitable access to transplantation as well as prioritizing patients with true needs are essential to address disparities. In conclusion, liver transplantation is often the beacon of hope for individuals suffering from end-stage liver disease and improves quality of life. The ethics related to transplantation are complex and multifaceted, considering not just the donor and the recipient, but also the society as a whole.
Collapse
Affiliation(s)
- Hoi Pong Nicholas Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Surya Varma Selvakumar
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Pei Yi Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Jovan Yi Jun Liau
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Matthias Yi Quan Liau
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Vishalkumar Girishchandra Shelat
- Department of General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
- Surgical Science Training Centre, Tan Tock Seng Hospital, Singapore 308433, Singapore
| |
Collapse
|
27
|
Galow AM, Agriesti F. Advances in Stem Cell Research-Insights from the Special Issue "Stem Cells in Health and Disease 2.0". Int J Mol Sci 2024; 25:13364. [PMID: 39769129 PMCID: PMC11677540 DOI: 10.3390/ijms252413364] [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: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Significant progress in stem cell research over the last few decades has opened up new avenues for exploring human physiology and pathophysiology [...].
Collapse
Affiliation(s)
- Anne-Marie Galow
- Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Francesca Agriesti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
28
|
Górska A, Trubalski M, Borowski B, Brachet A, Szymańczyk S, Markiewicz R. Navigating stem cell culture: insights, techniques, challenges, and prospects. Front Cell Dev Biol 2024; 12:1435461. [PMID: 39588275 PMCID: PMC11586186 DOI: 10.3389/fcell.2024.1435461] [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: 06/03/2024] [Accepted: 10/09/2024] [Indexed: 11/27/2024] Open
Abstract
Stem cell research holds huge promise for regenerative medicine and disease modeling, making the understanding and optimization of stem cell culture a critical aspect of advancing these therapeutic applications. This comprehensive review provides an in-depth overview of stem cell culture, including general information, contemporary techniques, encountered problems, and future perspectives. The article begins by explaining the fundamental characteristics of various stem cell types, elucidating the importance of proper culture conditions in maintaining pluripotency or lineage commitment. A detailed exploration of established culture techniques sheds light on the evolving landscape of stem cell culture methodologies. Common challenges such as genetic stability, heterogeneity, and differentiation efficiency are thoroughly discussed, with insights into cutting-edge strategies and technologies aimed at addressing these hurdles. Moreover, the article delves into the impact of substrate materials, culture media components, and biophysical cues on stem cell behavior, emphasizing the intricate interplay between the microenvironment and cell fate decisions. As stem cell research advances, ethical considerations and regulatory frameworks become increasingly important, prompting a critical examination of these aspects in the context of culture practices. Lastly, the article explores emerging perspectives, including the integration of artificial intelligence and machine learning in optimizing culture conditions, and the potential applications of stem cell-derived products in personalized medicine. This comprehensive overview aims to serve as a valuable resource for researchers and clinicians, fostering a deeper understanding of stem cell culture and its key role in advancing regenerative medicine and biomedical research.
Collapse
Affiliation(s)
- Aleksandra Górska
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Mateusz Trubalski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Bartosz Borowski
- Students Scientific Association, Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, Lublin, Poland
| | - Adam Brachet
- Student Scientific Association, Department of Forensic Medicine, Medical University of Lublin, Lublin, Poland
| | - Sylwia Szymańczyk
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Renata Markiewicz
- Occupational Therapy Laboratory, Chair of Nursing Development, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
29
|
Wei S, Cheng RJ, Li S, Lu C, Zhang Q, Wu Q, Zhao X, Tian X, Zeng X, Liu Y. MSC-microvesicles protect cartilage from degradation in early rheumatoid arthritis via immunoregulation. J Nanobiotechnology 2024; 22:673. [PMID: 39497131 PMCID: PMC11536868 DOI: 10.1186/s12951-024-02922-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Abstract
OBJECTIVE As research into preclinical rheumatoid arthritis (pre-RA) has advanced, a growing body of evidence suggests that abnormalities in RA-affected joint cartilage precede the onset of arthritis. Thus, early prevention and treatment strategies are imperative. In this study, we aimed to explore the protective effects of mesenchymal stem cell (MSC)-derived microvesicles (MVs) on cartilage degradation in a collagen-induced arthritis (CIA) mouse model. METHODS A CIA mouse model was established to observe early pathological changes in cartilage (days 21-25) through histological and radiological examinations. On day 22, MSCs-MVs were intravenously injected into the mice with CIA. Radiological, histological, and flow cytometric examinations were conducted to observe inflammation and cartilage changes in these mice compared to the mice with CIA and the control mice. In vitro, chondrocytes were cultured with inflammatory factors such as IL-1β and TNFα to simulate inflammatory damage to cartilage. After the addition of MVs, changes in inflammatory levels and collagen expression were measured via Western blotting, immunofluorescence, enzyme-linked immunosorbent assays (ELISAs), and quantitative PCR to determine the role of MVs in maintaining chondrocytes. RESULTS MSC-MVs expressed vesicular membrane proteins (CD63 and Annexin V) and surface markers characteristic of MSCs (CD44, CD73, CD90, and CD105). In the early stages of CIA in mice, a notable decrease in collagen content was observed in the joint cartilage. In mice with CIA, injection of MSCs-MVs resulted in a significant reduction in the peripheral blood levels of IL-1β, TNFα, and IL-6, along with a decrease in the ratio of proinflammatory T and B cells. Additionally, MSC-MVs downregulated the expression of IL-1β, TNFα, MMP-13, and ADAMTS-5 in cartilage while maintaining the stability of type I and type II collagen. These MVs also attenuated the destruction of cartilage, which was evident on imaging. In vitro experiments demonstrated that MSC-MVs effectively suppressed the secretion of the inflammatory factors IL-1β, TNFα, and IL-6 in stimulated peripheral blood mononuclear cells (PBMCs). CONCLUSIONS MSCs-MVs can inhibit the decomposition of the inflammation-induced cartilage matrix by regulating immune cell inflammatory factors to attenuate cartilage destruction. MSC-MVs are promising effective treatments for the early stages of RA.
Collapse
Affiliation(s)
- Shixiong Wei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Sujia Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chenyang Lu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiuhong Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xueting Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
30
|
Xue J, Wu D, Bao Y, Wu Y, Zhang X, Chen L. The Abnormal Proliferation of Midbrain Dopamine Cells From Human Pluripotent Stem Cells Is Induced by Exposure to the Tumor Microenvironment. CNS Neurosci Ther 2024; 30:e70117. [PMID: 39563017 PMCID: PMC11576488 DOI: 10.1111/cns.70117] [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: 02/03/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
AIMS Tumorigenicity is a significant concern in stem cell-based therapies. However, traditional tumorigenicity tests using animal models often produce inaccurate results. Consequently, a more sensitive method for assessing tumorigenicity is required. This study aimed to enhance sensitivity by exposing functional progenitors derived from human pluripotent stem cells (hPSCs) to the tumor microenvironment (TME) in vitro before transplantation, potentially making them more prone to abnormal proliferation or tumorigenicity. METHODS Midbrain dopamine (mDA) cells derived from hPSCs were exposed to the TME by coculturing with medulloblastoma. The cellular characteristics of these cocultured mDA cells were evaluated both in vitro and in vivo, and the mechanisms underlying the observed alterations were investigated. RESULTS Our findings demonstrated increased proliferation of cocultured mDA cells both in vitro and in vivo. Moreover, these proliferating cells showed a higher expression of Ki67 and SOX1, suggesting abnormal proliferation. The observed abnormal proliferation in cocultured mDA cells was attributed to the hyperactivation of proliferation-related genes, the JAK/STAT3 pathway, and cytokine stimulation. CONCLUSION This study indicates that exposing functional progenitors to the TME in vitro before transplantation can induce abnormal proliferation, thereby increasing the sensitivity of tumorigenicity tests.
Collapse
Affiliation(s)
- Jun Xue
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Dongyan Wu
- Institute of Neurology, Huashan Hospital, Shanghai Medical CollegeFudan UniversityShanghaiChina
- National Clinical Research Center for Aging and Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Yuting Bao
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Yifan Wu
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Xin Zhang
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain ScienceFudan UniversityShanghaiChina
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
31
|
Ivanovski S, Han P, Peters O, Sanz M, Bartold P. The Therapeutic Use of Dental Mesenchymal Stem Cells in Human Clinical Trials. J Dent Res 2024; 103:1173-1184. [PMID: 39370700 PMCID: PMC11562285 DOI: 10.1177/00220345241261900] [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: 10/08/2024] Open
Abstract
Mesenchymal stem cells (MSCs), characterized by their undifferentiated and multipotent nature, can be derived from various sources, including bone marrow, adipose, and dental tissues. Among these, dental MSCs (DSCs) exhibit universal MSC characteristics and are attracting considerable attention for regenerating oral and craniofacial tissues. This review provides a contemporary overview of recently published clinical studies using DSCs for various orodental and maxillofacial regenerative applications, including bone, periodontal, and endodontic regeneration. It also explores the utilization of DSCs in treating systemic conditions, exemplified by their application in managing conditions such as COVID-19 and osteoarthritis. The available evidence underscores the potential of DSCs and their secretome as efficacious tools in regenerative medicine for both dental and nondental clinical applications, supporting the continued promise of stem cell-based therapies. It is nevertheless evident that there are a number of important challenges that restrict the widespread utilization of DSCs, namely, difficulty in standardizing autologous preparations, insufficient cell surface marker characterization, high production costs, and regulatory compliance requirements. Further, the unique requirements of dental applications, especially complex structures such as the periodontium, where temporospatial control over the healing process is required, necessitate the combination of stem cells with appropriate scaffolds according to the principles of tissue engineering. There is currently insufficient evidence to support the clinical translation of DSCs into clinical practice, and phase 3 clinical trials with standardized protocols for cell sourcing, propagation, dosing, and delivery are required to move the field forward. In summary, this review provides a contemporary overview of the evolving landscape of stem cell therapy, offering insights into the latest developments and trends as well as the challenges that need to be addressed for the widespread application of DSC-based cell therapies.
Collapse
Affiliation(s)
- S. Ivanovski
- The University of Queensland, School of Dentistry, Brisbane, QLD, Australia
| | - P. Han
- The University of Queensland, School of Dentistry, Brisbane, QLD, Australia
- The University of Queensland, School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Brisbane, QLD, Australia
| | - O.A. Peters
- The University of Queensland, School of Dentistry, Brisbane, QLD, Australia
| | - M. Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, Faculty of Odontology, University Complutense of Madrid, Plaza Ramón y Cajalsn (Ciudad Universitaria), Madrid, Spain
| | - P.M. Bartold
- The University of Queensland, School of Dentistry, Brisbane, QLD, Australia
- The University of Adelaide, School of Dentistry, Adelaide, SA, Australia
| |
Collapse
|
32
|
Yang Z, Yang M, Rui S, Hao W, Wu X, Guo L, Armstrong DG, Yang C, Deng W. Exosome-based cell therapy for diabetic foot ulcers: Present and prospect. Heliyon 2024; 10:e39251. [PMID: 39498056 PMCID: PMC11532254 DOI: 10.1016/j.heliyon.2024.e39251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
Diabetic foot ulcers (DFUs) represent a serious complication of diabetes with high incidence, requiring intensive treatment, prolonged hospitalization, and high costs. It poses a severe threat to the patient's life, resulting in substantial burdens on patient and healthcare system. However, the therapy of DFUs remains challenging. Therefore, exploring cell-free therapies for DFUs is both critical and urgent. Exosomes, as crucial mediators of intercellular communication, have been demonstrated potentially effective in anti-inflammation, angiogenesis, cell proliferation and migration, and collagen deposition. These functions have been proven beneficial in all stages of diabetic wound healing. This review aims to summarize the role and mechanisms of exosomes from diverse cellular sources in diabetic wound healing research. In addition, we elaborate on the challenges for clinical application, discuss the advantages of membrane vesicles as exosome mimics in wound healing, and present the therapeutic potential of exosomes and their mimetic vesicles for future clinical applications.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Mengling Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Shunli Rui
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Wei Hao
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Xiaohua Wu
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Lian Guo
- Department of Endocrinology, School of Medicine, Chongqing University Three Gorges Central Hospital, Chongqing, 404000, China
| | - David G. Armstrong
- Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90033, USA
| | - Cheng Yang
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| | - Wuquan Deng
- Department of Endocrinology and Metabolism, School of Medicine, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, 400014, China
| |
Collapse
|
33
|
Xue J, Chu Y, Huang Y, Chen M, Sun M, Fan Z, Wu Y, Chen L. A tumorigenicity evaluation platform for cell therapies based on brain organoids. Transl Neurodegener 2024; 13:53. [PMID: 39472972 PMCID: PMC11520457 DOI: 10.1186/s40035-024-00446-5] [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: 05/27/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Tumorigenicity represents a critical challenge in stem cell-based therapies requiring rigorous monitoring. Conventional approaches for tumorigenicity evaluation are based on animal models and have numerous limitations. Brain organoids, which recapitulate the structural and functional complexity of the human brain, have been widely used in neuroscience research. However, the capacity of brain organoids for tumorigenicity evaluation needs to be further elucidated. METHODS A cerebral organoid model produced from human pluripotent stem cells (hPSCs) was employed. Meanwhile, to enhance the detection sensitivity for potential tumorigenic cells, we created a glioblastoma-like organoid (GBM organoid) model from TP53-/-/PTEN-/- hPSCs to provide a tumor microenvironment for injected cells. Midbrain dopamine (mDA) cells from human embryonic stem cells were utilized as a cell therapy product. mDA cells, hPSCs, mDA cells spiked with hPSCs, and immature mDA cells were then injected into the brain organoids and NOD SCID mice. The injected cells within the brain organoids were characterized, and compared with those injected in vivo to evaluate the capability of the brain organoids for tumorigenicity evaluation. Single-cell RNA sequencing was performed to identify the differential gene expression between the cerebral organoids and the GBM organoids. RESULTS Both cerebral organoids and GBM organoids supported maturation of the injected mDA cells. The hPSCs and immature mDA cells injected in the GBM organoids showed a significantly higher proliferative capacity than those injected in the cerebral organoids and in NOD SCID mice. Furthermore, the spiked hPSCs were detectable in both the cerebral organoids and the GBM organoids. Notably, the GBM organoids demonstrated a superior capacity to enhance proliferation and pluripotency of spiked hPSCs compared to the cerebral organoids and the mouse model. Kyoto Encyclopedia of Genes and Genomes analysis revealed upregulation of tumor-related metabolic pathways and cytokines in the GBM organoids, suggesting that these factors underlie the high detection sensitivity for tumorigenicity evaluation. CONCLUSIONS Our findings suggest that brain organoids could represent a novel and effective platform for evaluating the tumorigenic risk in stem cell-based therapies. Notably, the GBM organoids offer a superior platform that could complement or potentially replace traditional animal-based models for tumorigenicity evaluation.
Collapse
Affiliation(s)
- Jun Xue
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Youjun Chu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yanwang Huang
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ming Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Meng Sun
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Zhiqin Fan
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Yonghe Wu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
- Shanghai Clinical Research and Trial Center, Shanghai, 201210, China.
| | - Liang Chen
- Department of Neurosurgery, Huashan Hospital, MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200040, China.
- National Center for Neurological Disorders, Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
34
|
Shahbazi MN, Pasque V. Early human development and stem cell-based human embryo models. Cell Stem Cell 2024; 31:1398-1418. [PMID: 39366361 PMCID: PMC7617107 DOI: 10.1016/j.stem.2024.09.002] [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: 03/15/2024] [Revised: 06/18/2024] [Accepted: 09/02/2024] [Indexed: 10/06/2024]
Abstract
The use of stem cells to model the early human embryo promises to transform our understanding of developmental biology and human reproduction. In this review, we present our current knowledge of the first 2 weeks of human embryo development. We first focus on the distinct cell lineages of the embryo and the derivation of stem cell lines. We then discuss the intercellular crosstalk that guides early embryo development and how this crosstalk is recapitulated in vitro to generate stem cell-based embryo models. We highlight advances in this fast-developing field, discuss current limitations, and provide a vision for the future.
Collapse
Affiliation(s)
| | - Vincent Pasque
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; Leuven Stem Cell Institute & Leuven Institute for Single-Cell Omics (LISCO), Leuven, Belgium.
| |
Collapse
|
35
|
Hamazaki N, Yang W, Kubo CA, Qiu C, Martin BK, Garge RK, Regalado SG, Nichols EK, Pendyala S, Bradley N, Fowler DM, Lee C, Daza RM, Srivatsan S, Shendure J. Retinoic acid induces human gastruloids with posterior embryo-like structures. Nat Cell Biol 2024; 26:1790-1803. [PMID: 39164488 PMCID: PMC11469962 DOI: 10.1038/s41556-024-01487-8] [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: 02/05/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Gastruloids are a powerful in vitro model of early human development. However, although elongated and composed of all three germ layers, human gastruloids do not morphologically resemble post-implantation human embryos. Here we show that an early pulse of retinoic acid (RA), together with later Matrigel, robustly induces human gastruloids with posterior embryo-like morphological structures, including a neural tube flanked by segmented somites and diverse cell types, including neural crest, neural progenitors, renal progenitors and myocytes. Through in silico staging based on single-cell RNA sequencing, we find that human RA-gastruloids progress further than other human or mouse embryo models, aligning to E9.5 mouse and CS11 cynomolgus monkey embryos. We leverage chemical and genetic perturbations of RA-gastruloids to confirm that WNT and BMP signalling regulate somite formation and neural tube length in the human context, while transcription factors TBX6 and PAX3 underpin presomitic mesoderm and neural crest, respectively. Looking forward, RA-gastruloids are a robust, scalable model for decoding early human embryogenesis.
Collapse
Affiliation(s)
- Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
| | - Wei Yang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Connor A Kubo
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Beth K Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Riddhiman K Garge
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Samuel G Regalado
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Eva K Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sriram Pendyala
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Nicholas Bradley
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Douglas M Fowler
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Choli Lee
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Riza M Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Seattle Hub for Synthetic Biology, Seattle, WA, USA.
- Howard Hughes Medical Institute, Seattle, WA, USA.
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| |
Collapse
|
36
|
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
|
37
|
Gunjan, Himanshu, Pandey RP, Mukherjee R, Chang CM. Advanced meta-analysis on therapeutic strategies of mesenchymal derived exosome for diabetic chronic wound healing and tissue remodeling. Mol Cell Probes 2024; 77:101974. [PMID: 39038766 DOI: 10.1016/j.mcp.2024.101974] [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: 06/10/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Exosome (EXOs) are rapidly being identified as key mediators of cell-to-cell communication. They convey biologically active molecules to target cells, serve important roles in a range of physiological and pathological processes, and have enormous potential as novel therapeutic strategies. METHODS Preclinical research published between 2019 and 2023 provided the study's data searched on different medline search engine, and clinicaltrials.gov was searched for clinical data. These papers were chosen because they are relevant to the research of mesenchymal stem cell-derived exosomes (MSC-EXOs). Thematic synthesis and meta-analysis were used to perform the meta-analysis of diabetic wound healing. RESULTS For data extraction, a total of 18 preclinical and 4 clinical trials were selected. Preclinical investigations involving EXOs across various animal wound healing models showed promising potential for treatment. Specifically, following EXO treatment, there was a notable correlation with wound closure rates, with a pooled proportion of 46 % (95 % CI: 0.34; 0.59) and τ2 of 0.0593 after 3 ± 2 days, 54 % (95 % CI: 0.43; 0.65) and τ2 of 0.0465 after 7 ± 2 days, and 69 % (95 % CI: 0.62; 0.76) and τ2 of 0.0221 after 14 ± 2 days, with an egger's test p-value of <0.01. Further investigation into heterogeneity was conducted through subgroup analysis based on the source of EXO and the animal model utilized in the study. CONCLUSIONS EXOs are proving to be viable platforms for the treatment of a wide range of disorders in clinical trials. MSC-EXOs exhibited significant diabetic wound healing capabilities across diverse outcomes including wound closure, increase angiogenesis, immunomodulatory ability and skin regeneration with its typical structure and functions.
Collapse
Affiliation(s)
- Gunjan
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1(st) Road, Guishan District, Taoyuan City, 33302, Taiwan (R.O.C)
| | - Himanshu
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1(st) Road, Guishan District, Taoyuan City, 33302, Taiwan (R.O.C)
| | - Ramendra Pati Pandey
- School of Health Sciences and Technology (SoHST), UPES, Bidholi, Dehradun, 248007, Uttarakhand, India
| | - Riya Mukherjee
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1(st) Road, Guishan District, Taoyuan City, 33302, Taiwan (R.O.C)
| | - Chung-Ming Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259, Wenhua 1(st) Road, Guishan District, Taoyuan City, 33302, Taiwan (R.O.C); Master & Ph.D. Program in Biotechnology Industry, Chang Gung University, No. 259, Wenhua 1(st) Road, Guishan District, Taoyuan City, 33302, Taiwan (R.O.C); Department of Medical Biotechnology and Laboratory Science, Chang Gung University, No. 259, Wenhua 1(st) Road, Guishan District, Taoyuan City, 33302, Taiwan (R.O.C).
| |
Collapse
|
38
|
Isasi R, Bentzen HB, Fabbri M, Fuhr A, Glover JC, Mah N, Mascalzoni D, Mueller S, Seltmann S, Kurtz A. Dynamic governance: A new era for consent for stem cell research. Stem Cell Reports 2024; 19:1233-1241. [PMID: 39151430 PMCID: PMC11411296 DOI: 10.1016/j.stemcr.2024.07.006] [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: 03/21/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024] Open
Abstract
Governance infrastructures streamline scientific and ethical provenance verification of human pluripotent stem cell (SC) lines. Yet, scientific developments (e.g., SC-derived embryo models, organoids) challenge research governance approaches to stored biospecimens, questioning the validity of informed consent (IC) models. Likewise, e-health platforms are driving major transformations in data processing, prompting a reappraisal of IC. Given these developments, participatory research platforms are identified as effective tools to promote longitudinal engagement, interactive decision-making, and dynamic governance. Learning from European initiatives piloting dynamic IC for biobanking and SC research, this Perspective explores the benefits and challenges of implementing dynamic IC and governance for SC.
Collapse
Affiliation(s)
- Rosario Isasi
- Interdisciplinary Stem Cell Institute and Dr. John T. Macdonald Foundation Department of Human Genetics, Miami, FL, USA.
| | - Heidi B Bentzen
- Centre for Medical Ethics, Faculty of Medicine, University of Oslo, Oslo, Norway and Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Morris Fabbri
- Interdisciplinary Stem Cell Institute and Dr. John T. Macdonald Foundation Department of Human Genetics, Miami, FL, USA
| | - Antonie Fuhr
- Fraunhofer Institute für Biomedizinische Technik (IBMT), Joseph-von-Fraunhofer Weg 1, 66280 Sulzbach, Germany
| | - Joel C Glover
- Department of Molecular Medicine, University of Oslo, and Department of Immunology and Transfusion Medicine, Oslo University Hospital, Oslo, Norway
| | - Nancy Mah
- Fraunhofer Institute für Biomedizinische Technik (IBMT), Joseph-von-Fraunhofer Weg 1, 66280 Sulzbach, Germany
| | - Deborah Mascalzoni
- Centre for Research Ethics and Bioethics (CRB), Uppsala University, Uppsala, Sweden
| | - Sabine Mueller
- Fraunhofer Institute für Biomedizinische Technik (IBMT), Joseph-von-Fraunhofer Weg 1, 66280 Sulzbach, Germany
| | - Stefanie Seltmann
- Fraunhofer Institute für Biomedizinische Technik (IBMT), Joseph-von-Fraunhofer Weg 1, 66280 Sulzbach, Germany
| | - Andreas Kurtz
- Fraunhofer Institute für Biomedizinische Technik (IBMT), Joseph-von-Fraunhofer Weg 1, 66280 Sulzbach, Germany
| |
Collapse
|
39
|
Ore A, Angelastro JM, Giulivi C. Integrating Mitochondrial Biology into Innovative Cell Therapies for Neurodegenerative Diseases. Brain Sci 2024; 14:899. [PMID: 39335395 PMCID: PMC11429837 DOI: 10.3390/brainsci14090899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
The role of mitochondria in neurodegenerative diseases is crucial, and recent developments have highlighted its significance in cell therapy. Mitochondrial dysfunction has been implicated in various neurodegenerative disorders, including Alzheimer's, Parkinson's, amyotrophic lateral sclerosis, and Huntington's diseases. Understanding the impact of mitochondrial biology on these conditions can provide valuable insights for developing targeted cell therapies. This mini-review refocuses on mitochondria and emphasizes the potential of therapies leveraging mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, stem cell-derived secretions, and extracellular vesicles. Mesenchymal stem cell-mediated mitochondria transfer is highlighted for restoring mitochondrial health in cells with dysfunctional mitochondria. Additionally, attention is paid to gene-editing techniques such as mito-CRISPR, mitoTALENs, mito-ZNFs, and DdCBEs to ensure the safety and efficacy of stem cell treatments. Challenges and future directions are also discussed, including the possible tumorigenic effects of stem cells, off-target effects, disease targeting, immune rejection, and ethical issues.
Collapse
Affiliation(s)
- Adaleiz Ore
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
- Department of Chemical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James M. Angelastro
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
- University of California Medical Investigations of Neurodevelopmental Disorders Institute (MIND Institute), University of California Health, Sacramento, CA 95817, USA
| |
Collapse
|
40
|
Lunshof JE, Rijssenbeek J. Collaborative ethics: innovating collaboration between ethicists and life scientists. Nat Methods 2024; 21:1571-1574. [PMID: 38902517 DOI: 10.1038/s41592-024-02320-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Affiliation(s)
- Jeantine E Lunshof
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, USA.
- Department of Genetics, Harvard Medical School, Boston, USA.
| | - Julia Rijssenbeek
- Philosophy Group, Wageningen University & Research, Wageningen, the Netherlands
| |
Collapse
|
41
|
Iwamoto-Stohl LK, Petelski AA, Meglicki M, Fu A, Khan S, Specht H, Huffman G, Derks J, Jorgensen V, Weatherbee BAT, Weberling A, Gantner CW, Mandelbaum RS, Paulson RJ, Lam L, Ahmady A, Vasquez ES, Slavov N, Zernicka-Goetz M. Proteome asymmetry in mouse and human embryos before fate specification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.26.609777. [PMID: 39253500 PMCID: PMC11383291 DOI: 10.1101/2024.08.26.609777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Pre-patterning of the embryo, driven by spatially localized factors, is a common feature across several non-mammalian species 1-4 . However, mammals display regulative development and thus it was thought that blastomeres of the embryo do not show such pre-patterning, contributing randomly to the three lineages of the blastocyst: the epiblast, primitive endoderm and trophectoderm that will generate the new organism, the yolk sac and placenta respectively 4-6 . Unexpectedly, early blastomeres of mouse and human embryos have been reported to have distinct developmental fates, potential and heterogeneous abundance of certain transcripts 7-12 . Nevertheless, the extent of the earliest intra-embryo differences remains unclear and controversial. Here, by utilizing multiplexed and label-free single-cell proteomics by mass-spectrometry 13 , we show that 2-cell mouse and human embryos contain an alpha and a beta blastomere as defined by differential abundance of hundreds of proteins exhibiting strong functional enrichment for protein synthesis, transport, and degradation. Such asymmetrically distributed proteins include Gps1 and Nedd8, depletion or overexpression of which in one blastomere of the 2-cell embryo impacts lineage segregation. These protein asymmetries increase at 4-cell stage. Intriguingly, halved mouse zygotes display asymmetric protein abundance that resembles alpha and beta blastomeres, suggesting differential proteome localization already within zygotes. We find that beta blastomeres give rise to a blastocyst with a higher proportion of epiblast cells than alpha blastomeres and that vegetal blastomeres, which are known to have a reduced developmental potential, are more likely to be alpha. Human 2-cell blastomeres also partition into two clusters sharing strong concordance with clusters found in mouse, in terms of differentially abundant proteins and functional enrichment. To our knowledge, this is the first demonstration of intra-zygotic and inter-blastomere proteomic asymmetry in mammals that has a role in lineage segregation.
Collapse
|
42
|
Ibi Y, Nishinakamura R. Generating kidney organoids based on developmental nephrology. Eur J Cell Biol 2024; 103:151450. [PMID: 39137450 DOI: 10.1016/j.ejcb.2024.151450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
Over the past decade, the induction protocols for the two types of kidney organoids (nephron organoids and ureteric bud organoids) from pluripotent stem cells (PSCs) have been established based on the knowledge gained in developmental nephrology. Kidney organoids are now used for disease modeling and drug screening, but they also have potential as tools for clinical transplantation therapy. One of the options to achieve this goal would be to assemble multiple renal progenitor cells (nephron progenitor, ureteric bud, stromal progenitor) to reproduce the organotypic kidney structure from PSCs. At least from mouse PSCs, all the three progenitors have been induced and assembled into such "higher order" kidney organoids. We will provide an overview of the developmental nephrology required for the induction of renal progenitors and discuss recent advances and remaining challenges of kidney organoids for clinical transplantation therapy.
Collapse
Affiliation(s)
- Yutaro Ibi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
43
|
Tan JP, Liu X, Polo JM. Reprogramming fibroblast into human iBlastoids. Nat Protoc 2024; 19:2298-2316. [PMID: 38632379 DOI: 10.1038/s41596-024-00984-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 02/12/2024] [Indexed: 04/19/2024]
Abstract
The study of early human embryogenesis has relied on the use of blastocysts donated to research or simple stem cell culture systems such as pluripotent and trophoblast stem cells, which have been seminal in shedding light on many key developmental processes. However, simple culture systems lack the necessary complexity to adequately model the spatiotemporal, cellular and molecular dynamics occurring during the early phases of embryonic development. As such, an in vitro model of the human blastocyst is advantageous in many aspects to decipher human embryogenesis. Here we describe a step-by-step protocol for the generation of induced blastoids (iBlastoids), an in vitro integrated model of the human blastocyst derived via somatic reprogramming. This protocol details the workflow for reprogramming of human dermal fibroblasts and subsequent generation of iBlastoids using the reprogramming intermediates, which together takes ~27 days (21 days for reprogramming and 6 days for iBlastoid generation). We also discuss several characterization/functional assays that can be used on the iBlastoids. We believe that a person trained in cell culture with ~1 year of experience with human somatic cell and reprogramming/cell differentiation assays would be able to perform this protocol. In short, the iBlastoids present an alternative tool as a model to the blastocyst to facilitate the scientific community in the exploration of early human development.
Collapse
Affiliation(s)
- Jia Ping Tan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Xiaodong Liu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.
- Adelaide Centre for Epigenetics, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
- The South Australian Immunogenomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
44
|
Li C, Yao L, He F, Hua K. Transplantation of acellular amniotic membrane seeded with adipose-derived mesenchymal stem cells in a rat model of intrauterine adhesion. Ann Med Surg (Lond) 2024; 86:4463-4474. [PMID: 39118771 PMCID: PMC11305806 DOI: 10.1097/ms9.0000000000002249] [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: 12/05/2023] [Accepted: 05/27/2024] [Indexed: 08/10/2024] Open
Abstract
Objective This study aimed to investigate the role of acellular amniotic membrane (AAM) loaded with adipose-derived mesenchymal stem cells (ADSCs) for the treatment of intrauterine adhesion. Methods One hundred twenty female Spargue-Dawley rats were randomly divided into four groups: sham operation group (the uterus was picked out and incised without treatment), intrauterine adhesion group, the experimental group treated with AAM, and experimental group treated with AAM loaded with ADSCs. Histological and immunohistochemical analysis were performed on 3, 7, and 14 days after surgery to evaluate the degree of uterine fibrosis and regeneration of injured endometrium. RNA sequencing and real-time PCR were used to explore the potential mechanism by which ADSCs modulated immune response and promoted endometrial regeneration. Results On 14 days after surgery, the endometrial thickness, number of glands, and degree of fibrosis reduction in the ADSCs/AAM group was higher than those in the AAM group, and similar to the sham operation group. RNA sequencing analysis showed that ADSCs can modulate local immune responses and promote the formation of functional endometrium. Meanwhile, we found that ADSCs significantly decreased the levels of pro-inflammatory cytokines (TNF-α and IL-1β) and increased the levels of anti-inflammatory cytokines (bFGF and IL-6). Conclusion Our results demonstrated that AAM loaded with ADSCs can result in the regeneration of injured endometrium and fibrosis reduction. Meanwhile, ADSCs also regulated the immune microenvironment, which was beneficial to functional endometrial recovery.
Collapse
Affiliation(s)
- Chunbo Li
- Department of Obstetrics and Gynecology, The Obstetrics & Gynecology Hospital of Fudan University, Shanghai
| | - Liangfeng Yao
- Department of Obstetrics and Gynecology, Zhangzhou Municipal Hospital of Fujian Province, Zhangzhou
| | - Fengquan He
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of HongHe Hani and Yi Autonomous Prefecture, Yunnan, China
| | - Keqin Hua
- Department of Obstetrics and Gynecology, The Obstetrics & Gynecology Hospital of Fudan University, Shanghai
| |
Collapse
|
45
|
Barlevy D, Juengst E, Kahn J, Moreno J, Lambert L, Charo A, Chneiweiss H, Farooque M, Guston DH, Hyun I, Knoepfler PS, Selin C, Wilbanks R, Zaghlula M, Scott CT. Governing with public engagement: an anticipatory approach to human genome editing. SCIENCE & PUBLIC POLICY 2024; 51:680-691. [PMID: 39035203 PMCID: PMC11258878 DOI: 10.1093/scipol/scae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/21/2024] [Indexed: 07/23/2024]
Abstract
In response to calls for public engagement on human genome editing (HGE), which intensified after the 2018 He Jiankui scandal that resulted in the implantation of genetically modified embryos, we detail an anticipatory approach to the governance of HGE. By soliciting multidisciplinary experts' input on the drivers and uncertainties of HGE development, we developed a set of plausible future scenarios to ascertain publics values-specifically, their hopes and concerns regarding the novel technology and its applications. In turn, we gathered a subset of multidisciplinary experts to propose governance recommendations for HGE that incorporate identified publics' values. These recommendations include: (1) continued participatory public engagement; (2) international harmonization and transparency of multiple governance levers such as professional and scientific societies, funders, and regulators; and (3) development of a formal whistleblower framework.
Collapse
Affiliation(s)
- Dorit Barlevy
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX 77030, United States
| | - Eric Juengst
- Center for Bioethics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Jeffrey Kahn
- Berman Institute of Bioethics, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Jonathan Moreno
- Department of Medical Ethics and Health Policy, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Lauren Lambert
- College of Global Futures, Arizona State University, Tempe, AZ 85287, United States
| | - Alta Charo
- Law School, University of Wisconsin–Madison, Madison, WI 53706, United States
| | - Hervé Chneiweiss
- Neuroscience, Institute of Biology Paris Seine, CNRS UMR8246, INSERM U1130, Sorbonne Université, Paris 75252, France
| | - Mahmud Farooque
- Consortium for Science, Policy & Outcomes, Arizona State University, Washington, DC 20006, United States
- School for the Future of Innovation in Society, Arizona State University, Tempe, AZ 85281, United States
| | - David H Guston
- School for the Future of Innovation in Society, Arizona State University, Tempe, AZ 85281, United States
- Julie Ann Wrigley Global Futures Laboratory, Arizona State University, Tempe, AZ 85287, United States
| | - Insoo Hyun
- Museum of Science, Boston, MA 02114, United States
- Center for Bioethics, Harvard Medical School, Boston, MA 02115, United States
| | - Paul S Knoepfler
- Department of Cell Biology & Human Anatomy, UC Davis School of Medicine, Davis, CA 95616, United States
| | - Cynthia Selin
- School for the Future of Innovation in Society, Arizona State University, Tempe, AZ 85281, United States
- School of Sustainability, Arizona State University, Tempe, AZ 85281, United States
| | - Rebecca Wilbanks
- University Writing Program, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Manar Zaghlula
- Innovative Genomics Institute, University of California, Berkeley, CA 97404, United States
| | - Christopher Thomas Scott
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
46
|
Juguilon C, Wu JC. The role of the International Society for Stem Cell Research (ISSCR) guidelines in disease modeling. Dis Model Mech 2024; 17:dmm050947. [PMID: 38973350 PMCID: PMC11261629 DOI: 10.1242/dmm.050947] [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: 07/09/2024] Open
Abstract
Human stem cell-based modeling systems are valuable tools that can greatly improve the clinical translation of basic research. Importantly, the successful application of human stem cell-based models to biomedical research depends on the widespread adoption of ethical principles and practical standards. To achieve this outcome, the International Society for Stem Cell Research (ISSCR) provides a comprehensive set of recommendations that aim to promote the ethical usage of human stem cells and to ensure rigor and reproducibility within the field. Understanding and implementing these recommendations should be a top priority for investigators around the world.
Collapse
Affiliation(s)
- Cody Juguilon
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Medicine (Division of Cardiovascular Medicine), Stanford University, Stanford, CA 94305, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Medicine (Division of Cardiovascular Medicine), Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
47
|
Schmidt TT, Tyer C, Rughani P, Haggblom C, Jones JR, Dai X, Frazer KA, Gage FH, Juul S, Hickey S, Karlseder J. High resolution long-read telomere sequencing reveals dynamic mechanisms in aging and cancer. Nat Commun 2024; 15:5149. [PMID: 38890299 PMCID: PMC11189484 DOI: 10.1038/s41467-024-48917-7] [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: 10/25/2023] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
Telomeres are the protective nucleoprotein structures at the end of linear eukaryotic chromosomes. Telomeres' repetitive nature and length have traditionally challenged the precise assessment of the composition and length of individual human telomeres. Here, we present Telo-seq to resolve bulk, chromosome arm-specific and allele-specific human telomere lengths using Oxford Nanopore Technologies' native long-read sequencing. Telo-seq resolves telomere shortening in five population doubling increments and reveals intrasample, chromosome arm-specific, allele-specific telomere length heterogeneity. Telo-seq can reliably discriminate between telomerase- and ALT-positive cancer cell lines. Thus, Telo-seq is a tool to study telomere biology during development, aging, and cancer at unprecedented resolution.
Collapse
Affiliation(s)
| | - Carly Tyer
- Oxford Nanopore Technologies, Inc., New York, NY, USA
| | | | - Candy Haggblom
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Jeffrey R Jones
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Xiaoguang Dai
- Oxford Nanopore Technologies, Inc., New York, NY, USA
| | - Kelly A Frazer
- Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA, 92093-0761, USA
| | - Fred H Gage
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Sissel Juul
- Oxford Nanopore Technologies, Inc., New York, NY, USA
| | - Scott Hickey
- Oxford Nanopore Technologies, Inc., New York, NY, USA.
| | - Jan Karlseder
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
48
|
Kuang YH, Zhu W, Lin G, Cheng LM, Qin Q, Huang ZJ, Shi YL, Zhang CL, Xu JH, Yan KX, Lv CZ, Li W, Han Q, Stambler I, Lim LW, Chakrabarti S, Ulfhake B, Min KJ, Ellison-Hughes G, Cho WC, Jin K, Yao D, Lu C, Zhao RC, Chen X. Expert Consensus on the Application of Stem Cells in Psoriasis Research and Clinical Trials. Aging Dis 2024; 16:1363-1377. [PMID: 39012666 DOI: 10.14336/ad.2024.0012] [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: 01/03/2024] [Accepted: 06/04/2024] [Indexed: 07/17/2024] Open
Abstract
Psoriasis is an immune-mediated, chronic, relapsing, inflammatory, systemic disease induced by individual-environmental interactions, and is often lifelong because of the difficulty of treatment. In recent years, a variety of targeted therapies, including biologics, have improved the lesions and quality of life of most psoriasis patients, but they still do not address the problem of relapse and may be associated with decreased efficacy or adverse events such as infections over time. Therefore, there is an urgent need for breakthroughs in psoriasis treatment and in relapse-delaying and non-pharmacologic strategies, and stem cell therapy for psoriasis has emerged. In recent years, research on stem cell therapy for psoriasis has received a lot of attention, however, there is no reference standard as well as consensus in this field of research. Therefore, according to the latest consensus and guidelines, combined with relevant literature reports, clinical practice experience and the results of discussions with experts, this consensus specifies the types of stem cells commonly used in the treatment of psoriasis, the methods, dosages, and routes of stem cell therapy for psoriasis, as well as the clinical evaluations (efficacy and safety) of stem cell therapy for psoriasis. In addition, this consensus also provides normative standards for the processes of collection, preparation, preservation and quality control of stem cells and their related products, as well as recommendations for the management of stem cells during infusion for the treatment of psoriasis. This consensus provides the latest specific reference standards and practice guidelines for the field of stem cell therapy for psoriasis.
Collapse
Affiliation(s)
- Ye-Hong Kuang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| | - Wu Zhu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering Research Center of Human Stem Cell, Changsha, China
| | - La-Mei Cheng
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, China
- National Engineering Research Center of Human Stem Cell, Changsha, China
| | - Qun Qin
- The Office of Drug Clinical Trials Institution, Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Jun Huang
- Center for Clinical Pharmacology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yu-Ling Shi
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, China
| | - Chun-Lei Zhang
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Peking University Third Hospital, Beijing, China
| | - Jin-Hua Xu
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke-Xiang Yan
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Cheng-Zhi Lv
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
- Department of Dermatology, Dalian Dermatosis Hospital, Dalian, China
| | - Wei Li
- Department of Dermatology, Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| | - Qin Han
- International Society on Aging and Disease, Fort Worth, TX, USA
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Ilia Stambler
- International Society on Aging and Disease, Fort Worth, TX, USA
- Department of Science, Technology and Society, Bar Ilan University, Ramat Gan, Israel
| | - Lee Wei Lim
- International Society on Aging and Disease, Fort Worth, TX, USA
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Sasanka Chakrabarti
- International Society on Aging and Disease, Fort Worth, TX, USA
- Maharishi Markandeshwar Deemed University, Mullana-Ambala, India
| | - Brun Ulfhake
- International Society on Aging and Disease, Fort Worth, TX, USA
- Karolinska University Hospital, Stockholm, Sweden
| | - Kyung-Jin Min
- International Society on Aging and Disease, Fort Worth, TX, USA
- Department of Biological Sciences, Inha University, Incheon, Republic of Korea
| | - Georgina Ellison-Hughes
- International Society on Aging and Disease, Fort Worth, TX, USA
- School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Kunlin Jin
- International Society on Aging and Disease, Fort Worth, TX, USA
- University of North Texas Health Science Center, Bryan, TX, USA
| | - Danni Yao
- Department of Dermatology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | | | - Robert Chunhua Zhao
- International Society on Aging and Disease, Fort Worth, TX, USA
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- China Dermatologist Association, China
- Chinese Society of Dermatology, China
| |
Collapse
|
49
|
Mecca R, Tang S, Jones C, Coward K. The limitations of testicular organoids: are they truly as promising as we believe? Reprod Fertil Dev 2024; 36:RD23216. [PMID: 38935835 DOI: 10.1071/rd23216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Organoid systems have revolutionised various facets of biological research by offering a three-dimensional (3D), physiologically relevant in vitro model to study complex organ systems. Over recent years, testicular organoids have been publicised as promising platforms for reproductive studies, disease modelling, drug screening, and fertility preservation. However, the full potential of these systems has yet to be realised due to inherent limitations. This paper offers a comprehensive analysis of the current challenges associated with testicular organoid models. Firstly, we address the inability of current organoid systems to fully replicate the intricate spatial organisation and cellular diversity of the in vivo testis. Secondly, we scrutinise the fidelity of germ cell maturation within the organoids, highlighting incomplete spermatogenesis and epigenetic inconsistencies. Thirdly, we consider the technical challenges faced during organoid culture, including nutrient diffusion limits, lack of vasculature, and the need for specialised growth factors. Finally, we discuss the ethical considerations surrounding the use of organoids for human reproduction research. Addressing these limitations in combination with integrating complementary approaches, will be essential if we are to advance our understanding of testicular biology and develop novel strategies for addressing reproductive health issues in males.
Collapse
Affiliation(s)
- R Mecca
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - S Tang
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - C Jones
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - K Coward
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Level 3, Women's Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK
| |
Collapse
|
50
|
Yui H, Yashiro Y, Muto K, Watanabe S, Kiya Y, Inoue Y, Yamagata Z. Opinions on research involving human embryo models by researchers and the general public. Regen Ther 2024; 26:9-13. [PMID: 38798744 PMCID: PMC11126766 DOI: 10.1016/j.reth.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/21/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Rules and ethical considerations regarding research on embryo models have been debated across numerous countries. In this paper, we provide insights from our attitude survey conducted among Japanese researchers, including members of the Japanese Society for Regenerative Medicine, and among the general public residing in Japan, the US, the UK, Canada, and Australia. Our survey revealed that many researchers expressed the need for clear guidelines for embryo model research. Furthermore, a minority but significant portion of the general public in each country expressed opposition to research on embryo models but did not oppose research involving real embryos.
Collapse
Affiliation(s)
- Hideki Yui
- RIKEN Center for Integrative Medical Sciences, Yokohama-shi, Kanagawa, Japan
- Center for Birth Cohort Studies, University of Yamanashi, Chuo-shi, Yamanashi, Japan
| | - Yoshimi Yashiro
- Medical Innovation Center, Fujita Health University, Ota-ku, Tokyo, Japan
| | - Kaori Muto
- RIKEN Center for Integrative Medical Sciences, Yokohama-shi, Kanagawa, Japan
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Saori Watanabe
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Yukitaka Kiya
- The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Yusuke Inoue
- Department of Healthcare Ethics, Kyoto University School of Public Health, Kyoto-shi, Kyoto, Japan
| | - Zentaro Yamagata
- Center for Birth Cohort Studies, University of Yamanashi, Chuo-shi, Yamanashi, Japan
| |
Collapse
|