Next Article in Journal
Snapping of the Subacromial Bursa: A New Cause of Shoulder Pain Demonstrated with Dynamic Ultrasound
Previous Article in Journal
Associations of Skin Autofluorescence with Diabetic Kidney Disease in Type 2 Diabetes
Previous Article in Special Issue
Immune Evasion in Stem Cell-Based Diabetes Therapy—Current Strategies and Their Application in Clinical Trials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Pluripotent Stem Cells: Recent Advances and Emerging Trends

1
Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
2
International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
3
Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
4
College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(4), 765; https://doi.org/10.3390/biomedicines13040765
Submission received: 14 March 2025 / Accepted: 19 March 2025 / Published: 21 March 2025
(This article belongs to the Special Issue Pluripotent Stem Cell: Current Understanding and Future Directions)
The field of induced pluripotent stem cells (iPSCs) continues to evolve, offering unprecedented potential for regenerative medicine, disease modeling, and therapeutic applications. All 10 featured research papers collected in this Special Issue demonstrate the assessment of distinct iPSC features, which include molecular regulations and differentiation protocols, in addition to therapeutic applications, along with genetic editing practices and immune evasion developments. This editorial presents a summary of the essential discoveries within all published works from this issue and establishes their widespread field impact together with upcoming trends. (1) Advances in iPSC-based regenerative medicine by Jin et al. (2023) provides a review of iPSC-based treatments for urethral regeneration, analyzing the drawbacks of conventional graft procedures [1]. The paper by Du et al. (2023) examines stem cell-signaling in treating intervertebral disk degeneration, and demonstrates iPSC-derived therapies as a promising musculoskeletal disorder solution [2]. (2) Molecular and genetic insights in iPSCs by Machado et al. (2023) examines the molecular functions of OCT4 and SOX2 during iPSC reprogramming by exploring their effects on epigenetic modifications while discussing their role in keeping stem cells pluripotent [3]. Zhang et al. (2023) present an extensive review about CRISPR-Cas9 gene editing of iPSCs, which demonstrates both advanced genome precision techniques and promising medical possibilities [4]. Saini et al. (2023) examine ATP-binding cassette proteins together with their regulatory functions for sustaining stem cell pluripotency while demonstrating the importance of intracellular transport systems [5]. (3) Neural differentiation and neurological applications: Neural differentiation research combined with applications in neuroscience by Yarkova et al. (2024) develops a distinct approach to detect endoplasmic reticulum stress in induced pluripotent stem cell-derived neurons for helping model neurodegenerative diseases [6]. Research by Lee et al. (2024) demonstrates how iPSCs can differentiate into neurons, which shows encouraging results in preclinical studies for treating Parkinson’s disease [7]. (4) Machine learning and computational applications by Vedeneeva et al. (2023) presents machine learning technology that automatically detects iPSC colonies for high-quality selection enhancement while improving differentiation outcomes [8]. According to Chung et al. in (5) Developmental and signaling pathways (2024), BMP signaling controls the differentiation process of neural crest cells and ectodermal placode cells, which helps advance knowledge of embryological pathways using iPSC technology [9]. (6) Immune evasion strategies in iPSC-based therapies by Mu-u-min et al. (2025) presents immune evasion techniques for stem cell-based diabetes therapy as they review methods including encapsulation, along with genetic modifications to escape immune rejection difficulties [10].
The field of iPSCs has recently made significant improvements in safety together with efficiency and clinical practice readiness [11,12]. Modern reprogramming methods have reduced genomic alterations through the development of safer non-integrative approaches, including messenger RNA (mRNA) transfection and Sendai virus delivery, along with small molecule-based reprogramming to replace traditional viral methods for generating clinical-grade iPSCs [13,14,15,16]. The cells receive reprogramming factor genes OCT4, SOX2, KLF4, and c-MYC through transient mRNA transfection procedures, which transiently expresses these factors [13,15,17]. The genome-changing risks during reprogramming are minimized by mRNA transfection because it does not lead to integration. Through this method, scientists obtain controlled gene expression and faster reprogramming kinetics. Sendai virus delivery technique is another powerful tool for reprogramming [18,19,20,21]. It is replication-deficient and does not integrate into the host genome, making it safer for generating clinically relevant iPSCs. This method has been widely adopted for generating GMP-compliant (Good Manufacturing Practice) iPSCs for clinical applications. Researchers investigated reprogramming using chemical substances instead of classical transcription factors because these substitutes reduce mutation risks and enhance the process efficiency [22,23]. Simultaneously, the creation of 3D organoid models has taken the power of iPSCs past traditional differentiation methods to produce real-life models of brain and liver together with gastrointestinal system structures that help scientists examine diseases, test new drugs, and develop regenerative medical treatments [24,25,26]. Scientists can now use CRISPR-Cas9 in combination with iPSC technology to develop personalized regenerative treatments, thanks to recent field revolutionization [27,28,29]. For example, researcher access to disease mechanisms is enhanced through the gene-editing of iPSCs that come from patients with genetic disorders to produce matching control lines for scientific studies [29,30]. Parkinson’s disease-specific neurons derived from iPSCs allow researchers to edit them for disease-progression investigation of key genes [31]. Additionally, CRISPR-based technologies serve as tools to fix genetic errors found in iPSCs extracted from patients before converting these cells into healthy transplantation-ready cells [32,33]. Research has shown that autologous cell therapy could become possible through dystrophin gene correction in Duchenne muscular dystrophy patient-derived iPSCs [34]. Moreover, the latest CRISPR systems, such as base editors and prime editors, enable exact gene modification that produces fewer errors while avoiding double-strand break formation to minimize unintended mutations. Finally, gene-edited iPSCs are now being used to create humanized disease models for drug screening, allowing for the identification of compounds that target disease-specific pathways [35,36,37,38]. Additionally, refined differentiation protocols leveraging key signaling pathways, including BMP, Wnt, and TGF-β, have enhanced the efficiency and reproducibility of iPSC-derived cardiomyocytes, neurons, and pancreatic β-cells, accelerating their potential clinical applications [39,40,41]. However, the main challenge that prevents iPSC-based therapies from implementation is immune rejection. Research teams may employ CRISPR-Cas9 strategies to engineer hypoalloreactive iPSCs by removing HLA class I and II molecules and reducing immune surveillance, while adding PD-L1 regulatory proteins for developing universal cell supplies that would not require immunologic suppressing drugs [42]. Multiple technological developments, together, are accelerating the usage of iPSC technology for translation applications, which is leading medicine toward broad personalization and regeneration at a scale not seen before.
Several issues continue to challenge the advancement of iPSCs, even though research conducted in this Special Issue shows great potential for their use. For example, the obstacles to moving iPSC-derived cell products toward clinical implementation include maintaining reproducibility and scalability. The risk of genetic instability and tumor formation in iPSC-derived cell therapies necessitates stringent safety assessments. Because iPSCs are widely utilized in disease modeling and tissue transplantation methods, the existing ethical rules, together with regulatory procedures, must adapt properly, such as through the implementation of AI with machine learning capabilities in the field of stem cell research. Computational methods described by Vedeneeva et al. (2023) hold great potential to transform current processes for quality checks and cell differentiation protocols [8]. Applied translation of iPSC-based treatments for medical use continues as a key objective that depends on united work between experts in stem cell biology and bioengineering and medical sciences.
The studies assembled in this Special Issue deliver valuable research about pluripotent stem cells together with their potential medical applications. The data proves the versatile nature of iPSCs, as they continue to drive advances in both regenerative medicine and gene therapy together with disease modeling applications. The combination of gene editing along with bioengineering methods and computational technologies will enhance the development process for safe and effective iPSC-based therapeutic options.
As a Guest Editor of this Special Issue, we express heartfelt thanks to all the authors, together with reviewers and researchers who advanced the discipline. iPSC cells have only just begun their path, yet this path leads directly to a new age of regenerative healthcare that will rely heavily on these cells to define its future developments.

Author Contributions

Conceptualization, writing, and reviewing: A.Y.L.W., A.E.A., Y.-Y.L. and H.-K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Science and Technology Council, Taiwan (grant number NSTC 112-2314-B-182A-045-MY3), and the Chang Gung Medical Foundation, Chang Gung Memorial Hospital, Taiwan (grant number CMRPG3M0283).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Jin, Y.; Zhao, W.; Yang, M.; Fang, W.; Gao, G.; Wang, Y.; Fu, Q. Cell-Based Therapy for Urethral Regeneration: A Narrative Review and Future Perspectives. Biomedicines 2023, 11, 2366. [Google Scholar] [CrossRef] [PubMed]
  2. Du, X.; Liang, K.; Ding, S.; Shi, H. Signaling Mechanisms of Stem Cell Therapy for Intervertebral Disc Degeneration. Biomedicines 2023, 11, 2467. [Google Scholar] [CrossRef] [PubMed]
  3. Machado, L.S.; Borges, C.M.; de Lima, M.A.; Sangalli, J.R.; Therrien, J.; Pessoa, L.V.F.; Fantinato Neto, P.; Perecin, F.; Smith, L.C.; Meirelles, F.V.; et al. Exogenous OCT4 and SOX2 Contribution to In Vitro Reprogramming in Cattle. Biomedicines 2023, 11, 2577. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, Z.; Bao, X.; Lin, C.P. Progress and Prospects of Gene Editing in Pluripotent Stem Cells. Biomedicines 2023, 11, 2168. [Google Scholar] [CrossRef]
  5. Saini, P.; Anugula, S.; Fong, Y.W. The Role of ATP-Binding Cassette Proteins in Stem Cell Pluripotency. Biomedicines 2023, 11, 1868. [Google Scholar] [CrossRef]
  6. Yarkova, E.S.; Grigor’eva, E.V.; Medvedev, S.P.; Tarasevich, D.A.; Pavlova, S.V.; Valetdinova, K.R.; Minina, J.M.; Zakian, S.M.; Malakhova, A.A. Detection of ER Stress in iPSC-Derived Neurons Carrying the p.N370S Mutation in the GBA1 Gene. Biomedicines 2024, 12, 744. [Google Scholar] [CrossRef]
  7. Lee, D.H.; Lee, E.C.; Lee, J.Y.; Lee, M.R.; Shim, J.W.; Oh, J.S. Neuronal Cell Differentiation of iPSCs for the Clinical Treatment of Neurological Diseases. Biomedicines 2024, 12, 1350. [Google Scholar] [CrossRef]
  8. Vedeneeva, E.; Gursky, V.; Samsonova, M.; Neganova, I. Morphological Signal Processing for Phenotype Recognition of Human Pluripotent Stem Cells Using Machine Learning Methods. Biomedicines 2023, 11, 3005. [Google Scholar] [CrossRef]
  9. Chung, K.; Millet, M.; Rouillon, L.; Zine, A. Timing and Graded BMP Signalling Determines Fate of Neural Crest and Ectodermal Placode Derivatives from Pluripotent Stem Cells. Biomedicines 2024, 12, 2262. [Google Scholar] [CrossRef]
  10. Mu, U.M.R.B.A.; Diane, A.; Allouch, A.; Al-Siddiqi, H.H. Immune Evasion in Stem Cell-Based Diabetes Therapy-Current Strategies and Their Application in Clinical Trials. Biomedicines 2025, 13, 383. [Google Scholar] [CrossRef]
  11. Wang, A.Y.L. Human Induced Pluripotent Stem Cell-Derived Exosomes as a New Therapeutic Strategy for Various Diseases. Int. J. Mol. Sci. 2021, 22, 1769. [Google Scholar] [CrossRef] [PubMed]
  12. Loh, C.Y.; Wang, A.Y.; Kao, H.K.; Cardona, E.; Chuang, S.H.; Wei, F.C. Episomal Induced Pluripotent Stem Cells Promote Functional Recovery of Transected Murine Peripheral Nerve. PLoS ONE 2016, 11, e0164696. [Google Scholar] [CrossRef] [PubMed]
  13. Warren, L.; Lin, C. mRNA-Based Genetic Reprogramming. Mol. Ther. 2019, 27, 729–734. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, A.Y.L. Application of Modified mRNA in Somatic Reprogramming to Pluripotency and Directed Conversion of Cell Fate. Int. J. Mol. Sci. 2021, 22, 8148. [Google Scholar] [CrossRef]
  15. Warren, L.; Manos, P.D.; Ahfeldt, T.; Loh, Y.H.; Li, H.; Lau, F.; Ebina, W.; Mandal, P.K.; Smith, Z.D.; Meissner, A.; et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 2010, 7, 618–630. [Google Scholar] [CrossRef]
  16. Wang, A.Y.L.; Loh, C.Y.Y. Episomal Induced Pluripotent Stem Cells: Functional and Potential Therapeutic Applications. Cell Transplant. 2019, 28, 112S–131S. [Google Scholar] [CrossRef]
  17. Li, J.; Song, W.; Pan, G.; Zhou, J. Advances in understanding the cell types and approaches used for generating induced pluripotent stem cells. J. Hematol. Oncol. 2014, 7, 50. [Google Scholar] [CrossRef]
  18. Silva, M.; Daheron, L.; Hurley, H.; Bure, K.; Barker, R.; Carr, A.J.; Williams, D.; Kim, H.W.; French, A.; Coffey, P.J.; et al. Generating iPSCs: Translating cell reprogramming science into scalable and robust biomanufacturing strategies. Cell Stem Cell 2015, 16, 13–17. [Google Scholar] [CrossRef]
  19. Nishino, K.; Arai, Y.; Takasawa, K.; Toyoda, M.; Yamazaki-Inoue, M.; Sugawara, T.; Akutsu, H.; Nishimura, K.; Ohtaka, M.; Nakanishi, M.; et al. Epigenetic-scale comparison of human iPSCs generated by retrovirus, Sendai virus or episomal vectors. Regen. Ther. 2018, 9, 71–78. [Google Scholar] [CrossRef]
  20. Nakanishi, M.; Otsu, M. Development of Sendai virus vectors and their potential applications in gene therapy and regenerative medicine. Curr. Gene Ther. 2012, 12, 410–416. [Google Scholar] [CrossRef]
  21. Trokovic, R.; Weltner, J.; Nishimura, K.; Ohtaka, M.; Nakanishi, M.; Salomaa, V.; Jalanko, A.; Otonkoski, T.; Kyttala, A. Advanced feeder-free generation of induced pluripotent stem cells directly from blood cells. Stem Cells Transl. Med. 2014, 3, 1402–1409. [Google Scholar] [CrossRef] [PubMed]
  22. Lin, T.; Wu, S. Reprogramming with Small Molecules instead of Exogenous Transcription Factors. Stem Cells Int. 2015, 2015, 794632. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, J.; Sun, S.; Deng, H. Chemical reprogramming for cell fate manipulation: Methods, applications, and perspectives. Cell Stem Cell 2023, 30, 1130–1147. [Google Scholar] [CrossRef] [PubMed]
  24. Silva-Pedrosa, R.; Salgado, A.J.; Ferreira, P.E. Revolutionizing Disease Modeling: The Emergence of Organoids in Cellular Systems. Cells 2023, 12, 930. [Google Scholar] [CrossRef]
  25. Kim, J.; Koo, B.K.; Knoblich, J.A. Human organoids: Model systems for human biology and medicine. Nat. Rev. Mol. Cell Biol. 2020, 21, 571–584. [Google Scholar] [CrossRef]
  26. Yao, Q.; Cheng, S.; Pan, Q.; Yu, J.; Cao, G.; Li, L.; Cao, H. Organoids: Development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm 2024, 5, e735. [Google Scholar] [CrossRef]
  27. Gahwiler, E.K.N.; Motta, S.E.; Martin, M.; Nugraha, B.; Hoerstrup, S.P.; Emmert, M.Y. Human iPSCs and Genome Editing Technologies for Precision Cardiovascular Tissue Engineering. Front. Cell Dev. Biol. 2021, 9, 639699. [Google Scholar] [CrossRef]
  28. Walsh, C.; Jin, S. Induced Pluripotent Stem Cells and CRISPR-Cas9 Innovations for Treating Alpha-1 Antitrypsin Deficiency and Glycogen Storage Diseases. Cells 2024, 13, 1052. [Google Scholar] [CrossRef]
  29. McTague, A.; Rossignoli, G.; Ferrini, A.; Barral, S.; Kurian, M.A. Genome Editing in iPSC-Based Neural Systems: From Disease Models to Future Therapeutic Strategies. Front. Genome Ed. 2021, 3, 630600. [Google Scholar] [CrossRef]
  30. Soldner, F.; Laganiere, J.; Cheng, A.W.; Hockemeyer, D.; Gao, Q.; Alagappan, R.; Khurana, V.; Golbe, L.I.; Myers, R.H.; Lindquist, S.; et al. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations. Cell 2011, 146, 318–331. [Google Scholar] [CrossRef]
  31. Pinjala, P.; Tryphena, K.P.; Prasad, R.; Khatri, D.K.; Sun, W.; Singh, S.B.; Gugulothu, D.; Srivastava, S.; Vora, L. CRISPR/Cas9 assisted stem cell therapy in Parkinson’s disease. Biomater. Res. 2023, 27, 46. [Google Scholar] [CrossRef] [PubMed]
  32. Burnight, E.R.; Gupta, M.; Wiley, L.A.; Anfinson, K.R.; Tran, A.; Triboulet, R.; Hoffmann, J.M.; Klaahsen, D.L.; Andorf, J.L.; Jiao, C.; et al. Using CRISPR-Cas9 to Generate Gene-Corrected Autologous iPSCs for the Treatment of Inherited Retinal Degeneration. Mol. Ther. 2017, 25, 1999–2013. [Google Scholar] [CrossRef] [PubMed]
  33. Jackow, J.; Guo, Z.; Hansen, C.; Abaci, H.E.; Doucet, Y.S.; Shin, J.U.; Hayashi, R.; DeLorenzo, D.; Kabata, Y.; Shinkuma, S.; et al. CRISPR/Cas9-based targeted genome editing for correction of recessive dystrophic epidermolysis bullosa using iPS cells. Proc. Natl. Acad. Sci. USA 2019, 116, 26846–26852. [Google Scholar] [CrossRef] [PubMed]
  34. Li, H.L.; Fujimoto, N.; Sasakawa, N.; Shirai, S.; Ohkame, T.; Sakuma, T.; Tanaka, M.; Amano, N.; Watanabe, A.; Sakurai, H.; et al. Precise correction of the dystrophin gene in duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9. Stem Cell Rep. 2015, 4, 143–154. [Google Scholar] [CrossRef]
  35. Bassett, A.R. Editing the genome of hiPSC with CRISPR/Cas9: Disease models. Mamm. Genome 2017, 28, 348–364. [Google Scholar] [CrossRef]
  36. Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef]
  37. Okano, H.; Morimoto, S. iPSC-based disease modeling and drug discovery in cardinal neurodegenerative disorders. Cell Stem Cell 2022, 29, 189–208. [Google Scholar] [CrossRef]
  38. Cerneckis, J.; Cai, H.; Shi, Y. Induced pluripotent stem cells (iPSCs): Molecular mechanisms of induction and applications. Signal Transduct. Target. Ther. 2024, 9, 112. [Google Scholar] [CrossRef]
  39. Funa, N.S.; Mjoseng, H.K.; de Lichtenberg, K.H.; Raineri, S.; Esen, D.; Egeskov-Madsen, A.R.; Quaranta, R.; Jorgensen, M.C.; Hansen, M.S.; van Cuyl Kuylenstierna, J.; et al. TGF-beta modulates cell fate in human ES cell-derived foregut endoderm by inhibiting Wnt and BMP signaling. Stem Cell Rep. 2024, 19, 973–992. [Google Scholar] [CrossRef]
  40. Fujimori, K.; Matsumoto, T.; Kisa, F.; Hattori, N.; Okano, H.; Akamatsu, W. Escape from Pluripotency via Inhibition of TGF-beta/BMP and Activation of Wnt Signaling Accelerates Differentiation and Aging in hPSC Progeny Cells. Stem Cell Rep. 2017, 9, 1675–1691. [Google Scholar] [CrossRef]
  41. Pushpan, C.K.; Kumar, S.R. iPSC-Derived Cardiomyocytes as a Disease Model to Understand the Biology of Congenital Heart Defects. Cells 2024, 13, 1430. [Google Scholar] [CrossRef]
  42. Park, H.; Kang, Y.K.; Shim, G. CRISPR/Cas9-Mediated Customizing Strategies for Adoptive T-Cell Therapy. Pharmaceutics 2024, 16, 346. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, A.Y.L.; Aviña, A.E.; Liu, Y.-Y.; Kao, H.-K. Pluripotent Stem Cells: Recent Advances and Emerging Trends. Biomedicines 2025, 13, 765. https://doi.org/10.3390/biomedicines13040765

AMA Style

Wang AYL, Aviña AE, Liu Y-Y, Kao H-K. Pluripotent Stem Cells: Recent Advances and Emerging Trends. Biomedicines. 2025; 13(4):765. https://doi.org/10.3390/biomedicines13040765

Chicago/Turabian Style

Wang, Aline Yen Ling, Ana Elena Aviña, Yen-Yu Liu, and Huang-Kai Kao. 2025. "Pluripotent Stem Cells: Recent Advances and Emerging Trends" Biomedicines 13, no. 4: 765. https://doi.org/10.3390/biomedicines13040765

APA Style

Wang, A. Y. L., Aviña, A. E., Liu, Y.-Y., & Kao, H.-K. (2025). Pluripotent Stem Cells: Recent Advances and Emerging Trends. Biomedicines, 13(4), 765. https://doi.org/10.3390/biomedicines13040765

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop