UBIGENE

A Comprehensive Overview of iPSC Gene Editing: Concepts, Applications, and Technical Advantages

application-banner
Application
type-image
Gene Editing Practical Tips
A Comprehensive Overview of iPSC Gene Editing: Concepts, Applications, and Technical Advantages
Published on: January 08, 2026

Induced pluripotent stem cells (iPSCs) are a special type of stem cell “engineered” through advanced technologies. Scientists obtain somatic cells from human sources, such as skin or blood cells, and employ gene reprogramming techniques. By introducing specific transcription factors (e.g., Oct4, Sox2, Klf4, c-Myc), the existing differentiation program of these highly specialized cells is reversed, effectively “turning back the clock” to restore them to a pluripotent state with unlimited differentiation potential.Using CRISPR/Cas9, disease-mimicking mutations can be introduced into iPSCs, or pathogenic mutations in iPSC disease models can be corrected. These edited iPSCs can then be differentiated into the desired cell types for research or therapeutic applications, making iPSC gene editing one of the hottest areas in stem cell research today.

I. What is iPSC Gene Editing?

iPSC gene editing refers to the integration of induced pluripotent stem cell (iPSC) technology with CRISPR/Cas9 genome editing. This approach enables precise modification of the iPSC genome for gene knockout, knock-in, point mutation, or gene correction. Leveraging the unique properties of iPSCs—unlimited proliferation and multi-lineage differentiation—modified iPSCs can be induced to differentiate into specific functional cell types, such as neurons, cardiomyocytes, or pancreatic β cells.These edited cells can then be applied across a wide range of fields, including basic research, disease modeling, drug development, and therapeutic interventions. Based on the core logic of "iPSCs as the platform, gene editing as the tool", iPSC gene editing demonstrates versatile applications from fundamental science to clinical translation, serving as a key driver of breakthroughs in biomedical research.

II. Application Scenarios of iPSC Gene Editing

Basic Research: The “All-in-One Platform” for Gene Function Studies

1.Overcoming Cell Source Limitations for Long-Term Studies

iPSCs can be reprogrammed from adult somatic cells, such as skin fibroblasts or peripheral blood mononuclear cells, eliminating the need for embryonic stem cells and minimizing ethical concerns while providing a broad and accessible cell source.By combining iPSC technology with CRISPR/Cas9 genome editing, precise modifications such as gene knockout, knock-in, or point mutations can be achieved. Modified iPSCs can proliferate indefinitely, providing stable cellular models for long-term tracking of gene function. For example, editing key genes in signaling pathways within iPSCs allows continuous observation of their effects on cell differentiation, proliferation, and apoptosis, effectively addressing the limitations of primary cells, which are difficult to maintain in long-term culture.

Figure 1. Continuous inactivation of p53 reduces the efficiency of induced pluripotent stem cell (iPSC) generation [3].

Figure 1. Continuous inactivation of p53 reduces the efficiency of induced pluripotent stem cell (iPSC) generation [3].

2.Multi-Lineage Differentiation Capabilities for Diverse Tissue and Organ Research

iPSCs can differentiate into a variety of functional cell types, including neurons, cardiomyocytes, hepatocytes, and pancreatic β cells. When combined with gene editing, precise models can be established with “specific gene modifications + specific tissue cell types.”For example, editing genes related to neural function followed by differentiation into neurons provides an ideal tool for studying gene function in neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease.

Disease Modeling: From “In Vitro Simulation” to “Precision Prediction”

1.Patient-Specific Disease Model Construction to Recapitulate Disease Mechanisms

Patient-derived iPSCs can be reprogrammed from somatic cells and combined with gene editing to validate causal relationships between disease-causing genes and disease phenotypes, overcoming species differences inherent in traditional animal models. For example, in patients with hereditary cardiomyopathy, somatic cells can be reprogrammed into iPSCs. Using gene editing to correct pathogenic mutations allows researchers to observe whether the differentiated cardiomyocytes regain normal function, directly confirming the pathogenic role of the gene. For rare diseases, iPSCs can be edited to introduce disease-causing mutations, providing models despite limited patient samples and facilitating mechanistic studies that would otherwise be difficult to perform.

Figure 2. Generation and characterization of engineered heart muscle (EHM) from rhesus macaque [4].

Figure 2. Generation and characterization of engineered heart muscle (EHM) from rhesus macaque [4].

2.Dynamic Tracking of Disease Progression to Elucidate Pathological Mechanisms

The iPSC differentiation process can recapitulate human embryonic development and the dynamic progression of diseases. Combined with gene editing, the temporal expression of pathogenic genes can be precisely controlled, allowing researchers to observe pathological changes from early to late stages of disease.For example, in spinal muscular atrophy (SMA) research, editing disease-causing genes in iPSCs and tracking their differentiation into motor neurons enables the observation of apoptosis and functional abnormalities over time. This approach provides critical insights into the temporal dynamics of SMA pathogenesis.

Figure 3. Editing disease-causing genes in iPSCs provides critical insights into SMA pathogenesis [5].

Figure 3. Editing disease-causing genes in iPSCs provides critical insights into SMA pathogenesis [5].

Drug Development: Enhancing Screening Efficiency and Clinical Translation

1.A “Precision Platform” for High-Throughput Drug Screening

Functional cells derived from gene-edited iPSCs can recapitulate disease-specific cellular phenotypes, serving as a platform for high-throughput drug screening and activity validation. Compared with traditional cell models, disease-specific iPSC-derived cells better mimic human physiology, increasing the translational potential of candidate compounds. For example, in cystic fibrosis (CF), iPSCs with gene edits in CF-related genes can be differentiated into airway epithelial cells. These cells enable rapid screening of compounds that restore ion channel function, significantly reducing drug development time and failure rates.

2.An “Alternative Model” for Drug Toxicity and Safety Assessment

iPSCs can be differentiated into hepatocytes, cardiomyocytes, renal tubular cells, and other drug metabolism-relevant cell types. Combined with gene editing, they can model genetic polymorphisms (e.g., drug-metabolizing enzyme variants) to evaluate drug toxicity across different genetic backgrounds.For instance, by editing key drug metabolism genes in iPSCs, researchers can predict patient-specific drug metabolism rates and toxicity risks, enabling personalized drug safety assessment and providing guidance for rational clinical use.

Figure 4. Establishment and functional characterization of iPSC-derived airway organoids carrying the CFTR S308X nonsense mutation [7]

Figure 4. Establishment and functional characterization of iPSC-derived airway organoids carrying the CFTR S308X nonsense mutation [7]

Cell Therapy: The “Powerful Combination” of Gene Editing and Stem Cell Therapy

1.Exploring Curative Treatments for Genetic Diseases

For monogenic disorders, pathogenic mutations in patient-derived iPSCs can be corrected using gene editing technologies. The modified iPSCs are then differentiated into functional cells and transplanted back into the patient, achieving a dual therapeutic effect of “gene correction + cell replacement.”For example, in sickle cell anemia, hematopoietic stem cell-like cells can be obtained from the patient and reprogrammed into iPSCs. CRISPR-based correction of the β-globin gene mutation followed by differentiation into hematopoietic stem cells allows transplantation back into the patient, restoring normal hematopoiesis. Such therapies are currently in clinical trials, offering a promising approach for curing genetic diseases.

2.Overcoming Immune Rejection in Cell Therapy

By knocking out human leukocyte antigen (HLA) genes in iPSCs, “universal” iPSC banks can be generated. Functional cells differentiated from these iPSCs can be transplanted without triggering host immune rejection, eliminating the need for patient-specific iPSCs and significantly reducing the cost and time for cell therapy preparation.For example, universal iPSC-derived cardiomyocytes can be used for heart failure patients, and universal iPSC-derived neurons can aid in nerve injury repair, laying the foundation for large-scale clinical applications of cell therapy.

3.Cell Replacement Therapy for Complex Diseases

For non-genetic disorders such as diabetes, Parkinson'’'s disease, or spinal cord injury, gene editing can optimize iPSC differentiation efficiency and functional stability. The edited iPSCs can then be differentiated into pancreatic β cells, dopaminergic neurons, neural stem cells, or other relevant cell types for transplantation.For instance, editing differentiation-related genes in iPSCs can increase the proportion and functionality of β cells, enabling them to replace damaged islet cells post-transplantation, providing long-term therapeutic benefits for patients with type 1 diabetes.

Figure 5. The INS c.188-31G>A mutation generates insulin mRNA isoforms but does not produce insulin in differentiated human β-like cells [8]

Figure 5. The INS c.188-31G>A mutation generates insulin mRNA isoforms but does not produce insulin in differentiated human β-like cells [8]

III. Conclusion: The “Synergistic Effect” of iPSCs and Gene Editing

The combination of iPSCs' pluripotency and gene editing's precision creates a powerful synergistic effect. This approach not only overcomes the limitations of traditional research models—such as poor physiological relevance and difficulty in long-term stable culture—but also advances disease research from phenotypic observation to mechanistic understanding, and therapeutic strategies from symptomatic relief to targeting the root cause of disease.

With continuous innovations in gene editing technologies (e.g., base editing and prime editing) and the maturation of iPSC culture and differentiation techniques, the application scenarios of iPSCs are expanding. They range from gene function validation in basic research, to individualized disease modeling, high-throughput drug screening in pharmaceutical development, and clinical translation in cell therapy.iPSCs are increasingly becoming the core platform in the field of gene editing, driving innovation in biomedical research and offering promising avenues to address complex challenges in human health.

Reference

[1]McTague A, Rossignoli G, Ferrini A, Barral S, Kurian MA. Genome Editing in iPSC-Based Neural Systems: From Disease Models to Future Therapeutic Strategies. Front Genome Ed. 2021 Mar 15;3:630600. doi: 10.3389/fgeed.2021.630600. PMID: 34713254; PMCID: PMC8525405.
[2]Xu L, Wang J, Liu Y, Xie L, Su B, Mou D, Wang L, Liu T, Wang X, Zhang B, Zhao L, Hu L, Ning H, Zhang Y, Deng K, Liu L, Lu X, Zhang T, Xu J, Li C, Wu H, Deng H, Chen H. CRISPR-Edited Stem Cells in a Patient with HIV and Acute Lymphocytic Leukemia. N Engl J Med. 2019 Sep 26;381(13):1240-1247. doi: 10.1056/NEJMoa1817426. Epub 2019 Sep 11. PMID: 31509667.
[3]Yi L, Lu C, Hu W, Sun Y, Levine AJ. Multiple roles of p53-related pathways in somatic cell reprogramming and stem cell differentiation. Cancer Res. 2012 Nov 1;72(21):5635-45. doi: 10.1158/0008-5472.CAN-12-1451. Epub 2012 Sep 10. PMID: 22964580.
[4]Jebran AF, Seidler T, Tiburcy M, Daskalaki M, Kutschka I, Fujita B, Ensminger S, Bremmer F, Moussavi A, Yang H, Qin X, Mißbach S, Drummer C, Baraki H, Boretius S, Hasenauer C, Nette T, Kowallick J, Ritter CO, Lotz J, Didié M, Mietsch M, Meyer T, Kensah G, Krüger D, Sakib MS, Kaurani L, Fischer A, Dressel R, Rodriguez-Polo I, Stauske M, Diecke S, Maetz-Rensing K, Gruber-Dujardin E, Bleyer M, Petersen B, Roos C, Zhang L, Walter L, Kaulfuß S, Yigit G, Wollnik B, Levent E, Roshani B, Stahl-Henning C, Ströbel P, Legler T, Riggert J, Hellenkamp K, Voigt JU, Hasenfuß G, Hinkel R, Wu JC, Behr R, Zimmermann WH. Engineered heart muscle allografts for heart repair in primates and humans. Nature. 2025 Mar;639(8054):503-511. doi: 10.1038/s41586-024-08463-0. Epub 2025 Jan 29. PMID: 39880949; PMCID: PMC11903342.
[5]Grass T, Dokuzluoglu Z, Buchner F, Rosignol I, Thomas J, Caldarelli A, Dalinskaya A, Becker J, Rost F, Marass M, Wirth B, Beyer M, Bonaguro L, Rodriguez-Muela N. Isogenic patient-derived organoids reveal early neurodevelopmental defects in spinal muscular atrophy initiation. Cell Rep Med. 2024 Aug 20;5(8):101659. doi: 10.1016/j.xcrm.2024.101659. Epub 2024 Jul 26. PMID: 39067446; PMCID: PMC11384962.
[6]Tsai PH, Yang YP, Wang CH, Liang KH, Yeh CL, Huang TY, Hwang TC, Lo WL, Lan YT, Ching LJ, Wang ML, Huo TI, Luo YH, Chiou SH, Chou SJ. Multimodal single-cell transcriptomics with patient-specific iPSC-derived airway organoids as a drug screening approach for cystic fibrosis with nonsense mutations. Biomed Pharmacother. 2025 Oct;191:118476. doi: 10.1016/j.biopha.2025.118476. Epub 2025 Aug 18. PMID: 40829314.
[7]Sato M, Saitoh I, Inada E. Efficient CRISPR/Cas9-based gene correction in induced pluripotent stem cells established from fibroblasts of patients with sickle cell disease. Stem Cell Investig. 2016 Nov 14;3:78. doi: 10.21037/sci.2016.11.05. PMID: 28066780; PMCID: PMC5182212.
[8]Panova AV, Klementieva NV, Sycheva AV, Korobko EV, Sosnovtseva AO, Krasnova TS, Karpova MR, Rubtsov PM, Tikhonovich YV, Tiulpakov AN, Kiselev SL. Aberrant Splicing of INS Impairs Beta-Cell Differentiation and Proliferation by ER Stress in the Isogenic iPSC Model of Neonatal Diabetes. Int J Mol Sci. 2022 Aug 8;23(15):8824. doi: 10.3390/ijms23158824. PMID: 35955956; PMCID: PMC9369396.

Related service

CRISPR-U™ Knockout Cell Line
Based on the CRISPR-U™ technique, Ubigene selects appropriate transfection methods (electroporation or viral transduction) according to different cell characteristics to transfer gRNA and Cas9 into cells.
Logo
E-mail: info@ubigene.com
WhatsAPP: +86 153 6067 3248
TEL: +86 153 6067 3248(Int'l)
Copyright © 2025 Ubigene. All rights reserved.