Open access peer-reviewed chapter

Induced Pluripotent Stem Cells: Advances and Applications in Regenerative Medicine

Written By

Igor Kizub, Andrii Rozhok and Ganna Bilousova

Submitted: 10 September 2022 Reviewed: 01 December 2022 Published: 28 December 2022

DOI: 10.5772/intechopen.109274

From the Edited Volume

Possibilities and Limitations in Current Translational Stem Cell Research

Edited by Diana Kitala

Chapter metrics overview

192 Chapter Downloads

View Full Metrics

Abstract

Reprogramming adult somatic cells into induced pluripotent stem cells (iPSCs) through the ectopic expression of reprogramming factors offers truly personalized cell-based therapy options for numerous human diseases. The iPSC technology also provides a platform for disease modeling and new drug discoveries. Similar to embryonic stem cells, iPSCs can give rise to any cell type in the body and are amenable to genetic correction. These properties of iPSCs allow for the development of permanent corrective therapies for many currently incurable disorders. In this chapter, we summarize recent progress in the iPSC field with a focus on potential clinical applications of these cells.

Keywords

  • cell differentiation
  • cell-based therapy
  • genetic correction
  • induced pluripotent stem cells
  • iPSCs
  • regenerative medicine
  • stem cell reprogramming

1. Introduction

Direct reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) through the ectopic expression of reprogramming factors [1, 2, 3, 4, 5] has had a dramatic impact on the field of regenerative medicine and has opened a new era in research and therapy. Human iPSCs represent an unprecedented source of patient-specific pluripotent stem cells suitable for disease modeling and tissue replacement therapy.

Stem cells (SCs) have the ability to self-renew through cell division and can differentiate into various cell types. Based on their origin, SCs are divided into embryonic SCs (ESCs), induced pluripotent SCs (iPSCs), and adult SCs. ESCs are pluripotent cells derived from the inner cell mass of the blastocyst. They can give rise to tissues of the three germ layers and are regarded as a renewable potent cell source for the regeneration of all bodily tissues [6]. Adult SCs are multipotent cells of adult tissues that are also essential for regenerative medicine [7]. iPSCs share many similarities with ECSs, including pluripotency, differentiation potential, and the capability to form teratomas and viable chimeras [8].

Human ESCs are isolated by the use of surplus in vitro fertilization embryos [9]. Therefore, unlike the iPSC technology, ESC-based techniques do not allow for the generation of genetically diverse patient-specific cells. Additionally, the use of ESCs is obstructed by the need to destroy human embryos in the process of cell isolation, which raises ethical considerations. Primary human ESCs, therefore, are a suboptimal SC source for therapy and tissue engineering. ESC-based cell therapies may also result in immune rejection, which theoretically can be avoided if autologous iPSC-derived cells are used instead.

Similar to ESCs, iPSCs can proliferate indefinitely and differentiate into all three germ layers. Thus, the iPSC technology solves many problems associated with the use of ESCs and provides an unlimited source of autologous pluripotent SCs, which can be genetically corrected, differentiated into adult lineages, and returned to the same patient as an autograft [10]. Significant advances have been achieved in recent years in improving the safety of the iPSC technology; thus, expanding the opportunities for its clinical application. However, despite the tremendous potential of iPSCs, extensive analyses of their safety and reliability are still required. This chapter discusses the current progress and prospects of using the iPSC technology in tissue replacement therapy and as a tool for studying human pathologies.

Advertisement

2. Reprograming of somatic cells into iPSC

The first attempts to derive pluripotent SCs from adult cells were stimulated by early experiments that demonstrated the feasibility of reprogramming adult frog somatic cell nuclei by the cytoplasm of an enucleated unfertilized frog oocyte [11]. Later findings showed that reprogramming of somatic cells back to the pluripotent state is possible by transferring somatic cell nuclei into oocytes or by fusing somatic cells with pluripotent SCs [12, 13, 14]. Finally, the successful cloning of Dolly the sheep showed the feasibility of complete reprogramming of a mammalian somatic nucleus back to a pluripotent state from which it can develop a new animal [15].

2.1 Main reprograming factors

The success of animal cloning has demonstrated that unfertilized eggs and ESCs contain a set of factors that can confer pluripotency to somatic cells. Oct4, Sox2, c-Myc, and Klf4 were later identified by Takahashi and Yamanaka as sufficient to induce pluripotency in mouse somatic cells, resulting in iPSCs that were functionally equivalent to mouse ESCs [1, 2].

OCT4 is a key transcriptional factor, which maintains pluripotency in both early embryos and ESCs [16]. The level of OCT4 expression is vital for regulating pluripotency and it can both activate or repress the promoter of the REX1 gene, also a critical regulator of pluripotency [17]. The transcription factor SOX2 (sex determining region Y (SRY)-box2) is also essential for maintaining cell pluripotency. It comprises a regulatory complex with OCT4 and REX1 that cooperatively binds to DNA to activate transcription of other pluripotency factors [18]. The proto-oncogene c-MYC has multiple downstream targets that enhance cell proliferation, resulting in SC self-renewal [19]. The c-MYC protein can also induce global histone acetylation allowing OCT4 and SOX2 to bind to otherwise inaccessible sites [20]. Krüppel-like factor 4 (KLF4) is an oncogene that contributes to the long-term maintenance of the ESC phenotype. The role of KLF4 in cell reprogramming is probably to downregulate the expression of the tumor suppressor protein p53 [21]. KLF4 can also activate transcription by interacting with histone acetyltransferases, suppressing cell proliferation, and reciprocally acting with c-MYC [22].

The combination of OCT4, SOX2, KLF4, and c-MYC (abbreviated as OSKM), is widely used to reprogram various types of somatic cells into a pluripotent state. Under specific conditions, reprogramming can also be achieved without c-MYC or with only one or two factors from the OSKM set [23, 24, 25]. An alternative combination of OCT4, SOX2, NANOG, and LIN28 has also been shown to be sufficient to reprogram human cells into iPSCs [5]. Additional factors can be used in combination with OSKM to enhance reprogramming efficiencies, such as LIN28, human telomerase reverse transcriptase (hTERT), and SV40 large T antigen [26, 27, 28]. Regardless of their combination, reprogramming factors remodel the epigenetic configuration of somatic cells in a way that allows for the conversion of these mature somatic cells into immature pluripotent SCs. Mechanisms of reprogramming into iPSCs are reviewed in detail by Meir and Li [29].

2.2 Reprograming approaches

Early approaches to obtain iPSCs relied on the use of integrating retro- and lentiviral vectors to deliver reprogramming factors into somatic cells [1]. However, the expression of these exogenous factors is only essential in the initial step of reprogramming, and their silencing must occur to ensure the stability of iPSCs [2]. The use of retroviral vectors can not only result in the reactivation of exogenous reprogramming factors and in turn destabilization of iPSCs [2] but also increase the risk of insertional mutagenesis and cancer transformation in iPSC-derived tissues. The development of integration-free reprogramming approaches made the production of iPSCs safer for potential clinical applications. Somatic cells have been successfully reprogrammed into iPSCs using episomal plasmids encoding the reprogramming factors and adenoviral vectors [30, 31]. A number of other non-integrating vectors of viral origins have been utilized for reprogramming, such as those based on Sendai virus [32], Epstein–Barr virus [26], and various circular plasmid constructs [33, 34]. While these methods produce integration-free iPSCs, many of these approaches suffer from extremely low efficiency, which hampers their potential clinical application [35].

The generation of iPSCs has been recently accomplished with defined chemicals that can functionally replace reprogramming factors [36]. iPSCs can also be generated by fusing reprogramming factors with cell-penetrating proteins that allow for the efficient transport of reprogramming proteins through cell membrane [37]. Despite their safety, these DNA-free approaches also suffer from low reprogramming efficiency.

A more promising approach for the transgene-free generation of iPSCs can be the use of synthetic modified mRNAs encoding the reprogramming factors. This approach has been shown to reprogram a variety of cell lines with an efficiency superior to that of other integration-free approaches [38]. The disadvantage of this method is that RNAs have to be delivered into the cells daily during the reprogramming process. MicroRNA (miRNAs), such as miR-200c, miR-291-3p, miR-294, miR-295, and miR-302a-d, have also been shown to significantly enhance the efficiency of pluripotency induction [39]. When combined with modified mRNAs encoding reprogramming factors, miR-367 and miR-302a-d have been shown to increase the reprogramming efficiency of human primary fibroblasts to an unprecedented level, and reprogramming up to 90.7% of individually plated cells [40].

2.3 Reprogramming process

Different cell types have been used for the generation of iPSCs, albeit with different reprogramming efficiencies. Fibroblasts are the most commonly used cells due to their availability and easy culture conditions. Keratinocytes, melanocytes, blood cells, hepatocytes, and gastric epithelial cells are also suitable for reprogramming [41].

The reprogramming process has been extensively studied in fibroblasts and has been shown to follow an organized sequence of events, which begins with the downregulation of somatic gene expression [42]. The first step requires a phenotype transition initiated by the activation of the early pluripotency stage-specific embryonic antigen (SSEA1) and alkaline phosphatase, and the inactivation of the differentiation-related antigen Thy-1 (CD90), followed by the activation of NANOG and OCT4 [31, 42]. OCT4, SOX2, and NANOG further induce the expression of stemness genes, such as STAT3 and ZIC3, and repress differentiation-associated genes [17, 43]. The expression level and balance of reprogramming factors are also important for iPSC generation. For example, the increased relative expression of OCT4 enhances reprogramming efficiency [44].

Reprogramming somatic cells often results in the generation of heterogeneous iPSCs with different molecular phenotypes and differentiation potentials [45]. The duration of the reprogramming process also affects the characteristics of the resulting iPSC. Prolonged cultivation of iPSCs yields phenotypes closer to those of ESCs as compared to cells in the early phase of reprogramming. This suggests that reprogramming continues even after iPSCs have been established [46]. Alterations in epigenetic modifications, such as DNA methylation, are also important for iPSC induction [47]. Interestingly, epigenetic profiling of iPSCs has revealed that reprogrammed cells retain epigenetic marks of the cell type of origin [48] although these marks disappear upon continued passaging [42].

Advertisement

3. Clinical applications of iPSCs

The rapid progress of the iPSC technology increases efforts to translate autologous iPSC-based therapies into the clinic. While only a few clinical trials have directly tested the delivery of iPSC-derived cells into patients, the technology continues to develop and researchers from virtually every field develop iPSC-based cell therapies for relevant diseases. These therapies are still at different stages of development and rely on the feasibility to derive functional somatic cell types from iPSCs. Examples of the efforts toward the development of iPSC-based therapies for a variety of tissues and organs are summarized for selected fields below.

3.1 Dermatology

The skin may represent an ideal tissue for testing novel iPSC-based therapies. It is readily accessible, highly proliferative, and can be easily monitored. Multiple skin cell lineages have been generated from iPSCs, such as keratinocytes [49], melanocytes [50], fibroblasts [51], and ectodermal precursor cells [52]. Mouse iPSC-derived keratinocytes display characteristics similar to those of primary keratinocytes and can regenerate differentiated epidermis and skin appendages when grafted together with mouse fibroblasts into athymic mice [49]. Human iPSC-derived keratinocytes have also been shown to establish functional organotypic skin in culture and 3D skin models [53]. These and other studies have demonstrated the potential of iPSCs to generate autologous donor cells for cell-based therapies for skin diseases.

As an essential step toward future clinical use, in vitro 3D skin equivalents have been generated using iPSC-derived components. These equivalents exhibit normal skin morphology, stratification, and terminal differentiation [54], and can potentially be used for drug screening. Skin organoids with stratified epidermal and dermal skin layers that are able to spontaneously produce de novo hair and sebaceous glands have also been generated from murine [55] and human [56] iPSCs.

Many of the most devastating forms of inherited skin diseases that are caused by known mutations, such as Epidermolysis Bullosa (EB), can be treated with genetically corrected iPSC-derived cells [54]. EB is a group of inherited skin blistering diseases that results in severe blistering and scarring [57]. Precise gene editing techniques, such as those based on CRISPR (clustered regularly interspaced short palindromic repeats)/CRISPR-associate (Cas) systems, can be used for the generation of these patient-specific genetically corrected iPSCs. These corrected iPSCs can then be differentiated into skin cells and transplanted back to the same patient in need of treatment. Similar strategies can be implemented for a variety of other diseases affecting many other organs (Figure 1). In fact, the generation of iPSCs, coupled with gene targeting, can solve many obstacles that are associated with gene correction in somatic cells. Unlike somatic cells, iPSCs can be expanded indefinitely, allowing for easier selection and expansion of corrected clones. In addition, iPSCs derived from very old patients can differentiate into “rejuvenated” cells [58].

Figure 1.

Cell therapy strategies using iPSC-derived cells. Patient-specific primary cells of different origin can be isolated, cultured in vitro, reprogrammed to iPSCs, and if needed, genetically corrected. Genetically corrected or unmodified iPSC clones can be differentiated into desired cell lineages. These iPSC-derived cells can then be used for either autologous transplantation or the development of new therapeutic strategies, for example, by testing new drug modalities.

While currently there are no approved clinical trials to test iPSC-based therapies for the treatment of skin diseases, EB is likely to be the first skin disease to benefit from the iPSC-based therapy due to the severity of this disorder. For example, Anthony Oro’s team at Stanford University has received an award from the California Institute for Regenerative Medicine (CIRM) to translate an iPSC-based gene correction therapy for the severe recessive dystrophic form of EB into the clinic by producing transplantable epidermal sheets from genetically corrected EB iPSCs [59]. Dr. Oro’s team is currently generating data for an investigational new drug application (IND) with the Food and Drug Administration (FDA) to initiate a clinical trial.

3.2 Vascular therapy

Endothelial cells, pericytes, and vascular smooth muscle cells have been derived from animal and human iPSCs [60, 61]. iPSCs can also be directed into cord-blood endothelial colony-forming cells that can be used to derive highly proliferative blood vessel-forming cells applicable for the restoration of endothelial function in patients with vascular diseases [62].

Coronary artery disease continues to be the leading cause of death and morbidity around the world, with the existing therapy being not always efficient. Studies have shown that transplanted iPSCs can promote angiogenesis and effective tissue revascularization [63].

In diabetes, prolonged hyperglycemia causes aberrant angiogenesis in both micro- and macro-vessels, resulting in deficient functionality of endothelial progenitor cells and leading to decreased neovascularization. iPSC-derived endothelial cells have been widely explored as a model to study the mechanisms and novel treatments for endothelial dysfunction in type 1 and 2 diabetes and maturity-onset diabetes of the young (MODY) [64, 65].

iPSC-derived endothelial cells have also been used to study the mechanisms of macular degeneration and ischemic retinopathies [66, 67]. iPSC-derived spinal motor neurons and cerebral microvascular endothelial cells seeded into the spinal cord lead to vascular-neural interaction with specific maturation effects of endothelial cells on the neural tissue [68].

Vascular grafts have been successfully developed from iPSC-derived cells, recapitulating the cellular composition and orientation, as well as the anti-inflammatory properties, of functional blood vessels [69, 70].

Endothelial derived from iPSCs are yet to be evaluated in clinical trials. However, these cells are now successfully used for drug testing [71].

3.3 Cardiology

The recent advances in iPSC reprogramming into cardiomyocytes and other types of cardiac cells have provided potential avenues for cardiac repair, and functional cardiomyocytes have been successfully generated from iPSCs [72]. iPSC-derived cardiomyocyte-like cells demonstrate spontaneous contractility and exhibit molecular and structural similarities to cardiomyocytes.

Many studies have focused on testing iPSC-derived cells for post-myocardial infarction repair [73], which is one of the leading causes of morbidity and death throughout the world. In patients with extensive myocardial infarction, more than a billion cardiomyocytes can be lost, overwhelming the heart’s repair capacity. Such massive cell death in the myocardium initiates the replacement of cardiomyocytes with fibrous tissue, resulting in heart failure. Beating iPSC-derived cardiomyocytes have been generated from patients with hypertrophic cardiomyopathy associated with diastolic dysfunction to study the cellular mechanisms and potential therapeutic targets of diastolic dysfunction [74].

Patient-specific iPSC-derived cardiomyocytes have also been generated from patients with different types of diabetes and used for modeling and studying the molecular mechanisms underlying diabetic cardiomyopathies [75]. Human iPSC-derived cardiomyocytes, endothelial cells, and cardiac fibroblasts have been generated and integrated in beating 3D cardiac microtissues as a platform for cardiovascular disease modeling [76].

Human iPSC lines have also been generated from patient-derived cells to study ventricular and atrial arrhythmias, which often lead to sudden cardiac death [77].

A few clinical trials to test the efficacy of iPSC-derived cardiomyocytes have been initiated. For example, an Osaka University spin-off company, Cuorips, Inc. has recently initiated a clinical trial to determine the efficacy and safety of a human allogeneic iPSC-derived cardiomyocyte sheet for ischemic cardiomyopathy patients (NCT04696328). Heartseed, Inc. is currently testing iPSC-derived cardiomyocyte spheroids in patients with severe heart failure in Phase I/II study (NCT04945018). Since both studies are still ongoing, no results have been reported.

3.4 Skeletal muscle regeneration

Converting iPSCs into skeletal muscle cells can offer a tool for in vitro modeling of muscular diseases and potential hope for patients afflicted with skeletal muscle dystrophic diseases. Myogenic progenitor cells have been derived from iPSCs in many studies [78, 79].

Skeletal muscle satellite cells, which drive skeletal muscle regeneration, have been shown to play an important role in the early regeneration of damaged skeletal muscles in muscular dystrophies and have been generated from human iPSCs for the identification of new therapeutic targets for the treatment of these disorders [80]. 3D functional skeletal muscle tissues have also been successfully generated from human iPSC-derived skeletal myotubes with sarcomeric structures as an in vitro model of contractile myofibrils for disease modeling and drug screening to study muscular and neuromuscular diseases [78].

Developing treatments for muscular dystrophy is a priority topic for researchers. Vita Therapeutics, Inc. has recently received an orphan drug designation from the FDA to initiate a clinical study that will test the efficacy of iPSC-derived myogenic stem cells to treat Duchenne muscular dystrophy [59]. More iPSC-based therapies for muscular dystrophies are currently in development [81].

3.5 Neurology

Several types of well-differentiated and functional populations of neural cells and neuronal multipotent progenitors have been generated from human and murine iPSCs. These progenitors have also been tested in cell replacement studies in rodent models with promising results [82]. Spinal neural progenitors have been differentiated from human iPSCs and together with human iPSC-derived brain microvascular endothelial cells were included in the dual-channel spinal cord chip system as an in vitro model of human vascularized motor neuron tissue [68]. Similarly, the blood–brain barrier chip system has been created for modeling neurological disorders and drug screening [83]. Cells in such systems can be generated from the same iPSC donor source, producing an isogenic in vitro model [84].

iPSCs are becoming an important source for the development of personalized therapeutic and preclinical strategies for research focusing on neurodegeneration. Parkinson’s disease is one of the most common neurodegenerative disorders, resulting from the loss of dopamine neurons in substantia nigra. Cell replacement therapy, such as the transplantation of iPSC-derived neural progenitors, provides an alternative treatment strategy for Parkinson’s disease (PD) [85]. An ongoing clinical trial in Japan is testing iPSC-derived dopaminergic neurons for PD [86]. In 2018, midbrain dopaminergic progenitor cells derived from autologous iPSCs were successfully transplanted into the brain of a patient with PD, and clinical symptoms improved in this patient at 18 to 24 months after implantation [85]. Aspen Neuroscience is currently developing two iPSC-based therapies for PD: an autologous iPSC-derived dopaminergic neuron therapy for idiopathic PD and an autologous gene-corrected iPSC-derived dopaminergic neuron therapy for genetic PD [87]. These and other therapies currently in development provide hope in the treatment of many neurological conditions.

3.6 Hearing loss

Hearing loss is a common impairment in humans that mainly results from the irreversible loss of sensory hair cells and auditory neurons. Patient-specific iPSCs are a promising tool for the regeneration of sensory hair cells and spiral ganglion neurons of the affected cochlea. iPSCs can be successfully reprogrammed into otic epithelial progenitors and otic neuroprogenitors that can subsequently be differentiated into inner ear hair cells [88, 89]. Functional cochlear supporting cells that can be important therapeutic targets for the treatment of hereditary deafness have also been successfully generated from mouse iPSCs [90]. Human iPSC-derived neural progenitors have been shown to innervate early postnatal cochlear hair cells in vitro, forming functional synapses [91].

Sensorineural hearing loss is a prevalent form of deafness, commonly arising from damage to the cochlear sensory hair cells and degeneration of the spiral ganglion neurons. iPSCs can serve as an autologous source of replacement neurons in an injured cochlea for the treatment of sensorineural hearing loss and as a model system to develop therapies to treat hereditary hearing loss [89]. While many iPSC-based treatment options are being developed in research settings, there are no approved clinical trials for hearing loss using iPSCs.

3.7 Ophthalmology

Transplantation of ocular cells derived from both autologous and allogeneic iPSCs in animal models and clinical trials showed great promise for cell-based therapies and disease modeling in ophthalmology. Using patient’s own cells and the ability to correct disease-related gene mutations in patient-derived iPSCs provided a powerful approach for the treatment of ophthalmologic diseases. Various ocular cells have been generated from iPSCs, including corneal epithelial progenitor cells capable of terminal differentiation toward mature corneal epithelial-like cells [92], conjunctival epithelial cells, and conjunctival goblet-like cells [93], retinal pigment epithelial cells [92], photoreceptors, and retinal ganglion cells among others [94]. Human iPSCs can, in an autonomous manner, recapitulate the main steps of retinal development and form 3D retinal cups containing all major retinal cell types arranged in layers via retinal progenitors [95].

iPSCs hold a promise for the treatment of various degenerative eye disorders by filling clinical gaps in the use of adult limbal SCs or ESCs [96]. iPSC-derived photoreceptor cells and retinal pigment epithelium cells provide a cell replacement therapy for visual impairment associated with inherited retinal degeneration and age-related degeneration of photoreceptors [97].

Similar to the skin, the eye may represent an ideal tissue for testing iPSC therapies: it is relatively easy to monitor and access. Unsurprisingly, retinal pigment epithelium (RPE) cells derived from iPSCs were the first autologous iPSC-derived cell type to be transplanted into a human patient [85]. These RPE cells were used to treat age-related macular degeneration (AMD), the leading cause of vision loss in the elderly. A 4-year follow-up demonstrated that the iPSC-derived RPE sheets transplanted into the right eye of a 77-year-old patient had survived post-engraftment. While no improvement in vision was noted, the patient’s vision remained stable, emphasizing the safety of the iPSC-based therapy to treat eye diseases [98]. Additional iPSC-based therapies are being assessed for the treatment of AMD in clinical trials in Japan [99] and in the United States by the team at the National Institute of Health (NCT04339764). The use of iPSC-derived photoreceptors to cure blindness is also being tested in preclinical research [59].

3.8 Bone and cartilage regeneration

While autologous bone grafting remains the main approach to reconstruct bone defects, the risk of bone resorption, infection, and insufficient amount of tissue available for transplantation is high. Therefore, iPSC technologies may provide a suitable alternative to grafting autologous bone/cartilage-forming cells. Functional osteoblasts, osteocytes, and chondrocytes have been generated from iPSCs [100, 101].

Human and animal iPSCs, as well as iPSC-derived mesenchymal stem/stromal cells (MSCs), have been differentiated into osteoblast- and osteocyte-like cells, which could be transplanted to achieve bone formation or regeneration of calvarial bone defects in in vivo animal models [102]. For bone tissue regeneration, engineered bioactive scaffolds provide mechanical support and components that mimic the extracellular matrix for iPSC-derived osteogenic cell grafting, increasing their adhesion, growth, and survival [103].

iPSCs also represent a potential source of viable chondroprogenitors for articular cartilage repair and engineering [104]. The regenerative potential of iPSCs is attractive for the therapy of intervertebral disc degeneration—a common cause of musculoskeletal diseases, such as low back pain, which is often attributed to a reduced number of nucleus pulposus cells that form the intervertebral disc. Current treatment strategies fail to replenish nucleus pulposus cells. The latter, however, have been successfully differentiated from iPSCs [105].

The development of iPSC-based therapies for bone and cartilage diseases is still ongoing. However, Australian Cynata Therapeutics has already tested allogeneic MSCs derived from iPSCs in a clinical trial for graft vs. host disease (GvHD) (NCT02923375). The infusion of these iPSC-derived MSCs was well tolerated by patients and promoted encouraging improvement in symptoms of GvHD [106]. Cynata therapeutics is also initiating a Phase III trial that will use iPSC-derived MSCs for the treatment of osteoarthritis [107].

3.9 Dentistry

Human iPSCs can potentially be a source to derive human odontoblasts for tissue engineering and regenerative therapy for the treatment of dental pulp damage [108]. The tooth is formed by sequential reciprocal interactions between epithelial cells originating from mesenchymal cells and surface ectoderm cells from cranial neural crest. iPSCs can be differentiated into neural crest-like cells, which in turn can be differentiated into odontogenic mesenchymal cells, odontoblast progenitors, and odontoblasts suitable for transplantation [109].

Periodontal disease is an important health problem that ultimately leads to tooth loss. An alternative to the existing artificially manufactured tooth replacements is the generation of complete or partial biological replacements, consisting of living periodontal tissues. Animal and human iPSCs and iPSC-derived MSCs transplanted to animals with a model of molar defects demonstrate periodontal tissue regeneration. Transplanted neural crest-like cells derived from iPSCs have also been shown to form a well-organized vascularized dentin-pulp complex and calcified tooth-like structures, demonstrating the feasibility for iPSCs use in dental tissue regeneration [108]. However, extensive research is still needed to advance the iPSC technology into dental applications.

3.10 Nephrology and urology

Renal failure is one of the most common causes of mortality and morbidity in the world. An iPSC-based cell therapy may offer an alternative therapeutic approach to kidney transplantation.

iPSCs have been successfully differentiated into nephrogenic intermediate mesoderm and renal progenitor cells [110, 111]. These cells pose the ability to differentiate into multiple cell types that constitute the adult kidney, such as metanephric mesenchyme cells, metanephric stromal cells, nephric duct, ureteric bud cells, proximal tubular cells, mature glomerular podocytes, and other types of cells capable of forming renal tubule-like structures [110]. iPSC-derived kidney organoids have been created containing multiple cell types and mimicking nephrogenesis that have a potential for regenerative medicine and personalized therapy [112]. iPSC-derived cells are also used to recellularize decellularized kidney scaffolds as an approach to bioengineering human replacement kidneys [113]. iPSCs generated from patients with specific kidney disorders have also been used in disease modeling [114]. The iPSC technology is now widely used to model kidney diseases and perform drug screening. However, no clinical trials have been approved to date.

3.11 Pulmonology

Lung transplantation remains the only treatment for many severe lung diseases. The use of iPSCs may be an effective strategy for developing patient-specific cells for lung cell therapy and lung tissue engineering as an alternative to whole-organ transplantation. iPSC-based models of lung diseases can also help to better understand lung pathologies and identify new therapeutic approaches.

Human alveolar and airway epithelial and basal cells have been successfully generated from iPSCs [115, 116]. Airway basal cells, in particular, can give rise to other airway lineages, such as secretory and ciliated cells, and can restore airway functionality [117].

Human iPSC-derived type II alveolar epithelial cells are capable of repopulating acellular lung matrices prepared from rat and human adult lungs, adhering and proliferating to form alveolar structures as a 3D lung tissue model of the distal lung regions [118]. Recently, scaffold-free structures for airway regeneration were also created using 3D bioprinting and a combination of human native chondrocytes, MSCs, and iPSC-derived endothelial cells [119].

Thus, significant progress has been achieved in deriving alveolar epithelial cells from iPSCs. However the complexity of lung tissue prevents rapid development and clinical translation of iPSC-based therapies for lung diseases.

3.12 Hepatology

Currently, liver transplants represent the only way to treat patients suffering from terminal liver failure. However, liver transplantation is associated with numerous problems, such as graft failure. As an alternative to the donor liver, human iPSCs may provide a promising source of hepatocytes for autologous cell therapy. iPSCs have been differentiated into hepatocytes [120]. Since the function of hepatocytes depends on their position in the liver globule, methods have been developed to generate iPSC-derived hepatocytes with zone-specific hepatic properties [121].

In addition to hepatocytes, the liver parenchyma also consists of many types of nonparenchymal cells, which are essential for maintaining hepatic metabolic activity and other functions. Human iPSC-derived hepatic progenitors have been differentiated into multiple liver cell types and produce functional liver models [122]. In vitro liver models are crucial for the study of liver diseases and development of effective therapies. Since liver transplantation is contingent on organ availability and other constraints, transplantable iPSC-derived cells and vascularized 3D organoids capable of repopulating and restoring liver functions have been developed as an alternative [123].

Hepatocyte transplantation is one of the most attractive approaches for the treatment of liver failure [124], and patient-specific iPSC-derived hepatic cells are expected to be used for patient-specific transplantations. The transplantation of hepatocyte-like cells differentiated from genetically corrected iPSCs has also been shown to ameliorate inherited liver diseases in a mouse model [125]. While iPSC-derived hepatocytes have not yet been translated into clinical trials, these cells are currently being used for screening drug hepatoxicity [126].

3.13 Gastroenterology

The gastrointestinal tract is one of the largest and most active systems, which not only breaks down and absorbs macromolecules from the lumen but also functions as an endocrine organ that regulates digestion and metabolism. The gastrointestinal system requires integrated neuronal, lymphatic, immune, and vascular tissues to function properly. The iPSC technology provides unique opportunities for modeling human diseases and novel therapeutic approaches in regenerative gastroenterology. Human iPSC-derived intestinal and gastric cells, as well as generated in vitro human organoids, may facilitate drug screening and modeling of gastrointestinal diseases. On the other hand, intestinal cell models can be widely used to study drug absorption and metabolism.

iPSCs can be differentiated into various types of intestinal cells and can even form multicell type intestinal tissue [127]. Among other cell types, human iPSCs have been differentiated into gastric epithelial cells and acid-secreting parietal cells [128], mature exocrine pancreatic cells [129], as well as directed along the gastric endocrine cell fate path [127]. Human iPSCs can also be efficiently differentiated into neural crest SCs and various subtypes of mature enteric neurons [130]. Enterocytes derived from human iPSCs have been used as a model system for predicting the pharmacokinetics of the human intestine and drug absorption and metabolism [131, 132].

The developed cell culture protocols allow for the derivation of self-organizing multicellular intestinal organoids from iPSCs, which resemble in vivo intestinal crypts [133]. Such organoids, which are composed of various intestinal cells, represent a physiologically relevant in vitro model for basic studies of intestinal development and pathophysiology, as well as a tool in personalized regenerative medicine and drug development. Bioengineering the intestine on a vascularized native scaffold can also be a promising approach for intestinal regeneration in patients with intestinal failure [134]. Intestinal 3D organoids derived from human primary digestive samples are currently being tested in a clinical trial to treat ulcerative colitis at Rennes University Hospitals (NCT05294107). The successful completion of this study will pave way for approval to use iPSC-derived intestinal organoids for the treatment of intestinal diseases in clinical trials.

3.14 Metabolic disorders

Several major types of diabetes are caused by the destruction and decrease in the number of functional insulin-producing β-cells. Therefore, the generation of functional insulin-secreting pancreatic β-cells represents an important goal for the treatment of various types of diabetes. Functional insulin-secreting pancreatic β-cells have been successfully generated from healthy human iPSCs [135], providing an important cell source for personalized drug screening and cell transplantation therapy in diabetes. Human iPSC-derived β-cells exhibit many of the properties of functional pancreatic β-cells, such as expression of specific transcription factors and the presence of mature endocrine secretory granules [136].

Brown adipocytes are promising cell targets for the treatment of obesity and type 2 diabetes due to their ability to actively drain and oxidize circulating glucose and triglycerides, which can prevent hyperglycemia and hypertriglyceridemia. Because of the scarcity of brown adipocytes in adults, iPSCs may be an important potential source of these cells and their progenitors for transplantation. Human iPSCs have been successfully differentiated into adipocytes [137].

MODY is a monogenic autosomal dominant disease caused by a mutation in one of the specific genes. Various mutations in each of these genes affect pancreatic β-cells, resulting in their dysfunction and diabetes development. However, despite extensive research, the mechanism by which the mutant MODY gene results in monogenic diabetes is not yet clear [138]. iPSCs have been generated from patients with different types of MODY to establish a human-based model for studying the molecular manifestations and mechanisms of these diseases [139].

iPSCs harboring disease-specific gene mutations have also been generated from somatic cells of patients with a few types of lipodystrophy and other inherited metabolic disorders for studying the pathogenesis of these diseases [140, 141].

While iPSC-based therapies for metabolic diseases are still being developed, the progress achieved by Melton’s group in using human ESCs to derive functional allogeneic insulin-producing β-cells [142] has created a strong foundation for the use of iPSCs in treating type 1 diabetes. Vertex Pharmaceuticals, Inc. is currently testing the glucose-responsive allogeneic β-cells generated from human ESCs in combination with immunosuppressive therapy in Phase I clinical trial (NCT04786262). The preliminary results of this clinical trial show engraftment and functionality of implanted hESC-derived β-cells [143].

3.15 Gland regeneration

Hypofunction of salivary glands causes various life-disrupting effects. With no satisfactory therapy available, the therapeutic and regenerative potential of iPSCs has been explored for the treatment of salivary gland dysfunction [144].

Inflammatory and degenerative changes in the lacrimal gland often lead to the development of severe dry eye syndrome, a complex disease resulting in visual acuity disruption. Currently, only palliative treatments for this disease exist. iPSCs have been used for developing a therapy for lacrimal gland tissue injuries [145].

The thymus plays a significant role in the establishment of immunological self-tolerance and is required for the generation of T cell-mediated immunity. Thyroid progenitors, thymic epithelial cells, and thymic organoids derived from iPSCs can completely regenerate the thymus in vivo and demonstrate the potential for regenerative therapy in patients with immunodeficiency and hypothyroidism [146]. Despite the progress in generating iPSC-derived thymic cells, a poor understanding of thymus biology impedes the clinical translation of these cells.

The mammary gland is a primary target for carcinogenesis, and regenerative therapy for damaged mammary gland tissues is the best way to restore breast functions. iPSCs have been successfully reprogrammed into mammary SCs [147]. In addition, human mammary-like organoids have been produced from iPSCs. Such organoids have been shown to regenerate mammary glands upon transplantation [148]. To date, no clinical trials have been approved for the use of iPSCs in gland regeneration.

Advertisement

4. Conclusion

Despite tremendous progress achieved in the iPSC field, broad applications of iPSC-based therapies will take time to establish. Nevertheless, considerable advances have been made in deriving iPSCs from patients, differentiating them into tissues of interest, and using them as a platform for studying the mechanisms of diseases. The development of iPSC-based therapies is just emerging and only a limited number of clinical studies using the transplantation of iPSC-derived cells have been initiated to date. However, clinical studies related to the iPSC technology are not limited to the studies described above. Other studies worth mentioning include the successful use of autologous iPSC-derived platelets for the treatment of aplastic anemia [149] and the derivation of an off-the-shelf allogeneic chimeric antigen receptor (CAR) T-cell therapy targeting B-cell malignancies developed by Fate Therapeutics (NCT04629729). An iPSC-derived, off-the-shelf, CAR natural killer (NK) cell therapy is currently being tested in a Phase I clinic trial for refractory B-cell lymphoma (NCT 04245722) and is showing promising therapeutic efficacy [150]. Other studies have been reviewed by Kim et al. [99].

Even though iPSCs hold great potential in the field of regenerative medicine and personalized medicine, a number of challenges hinder the widespread clinical applications of these cells. These challenges include the safety of methodologies for the generation, gene correction, and differentiation of iPSCs and the high cost associated with the technology.

The first major challenge is reprogramming and gene correction methods that are known to introduce undesired genetic modifications into the patient genome. Other limitations include the heterogeneity of iPSC lines that impairs the consistency of differentiation during manufacturing of iPSC-based cell products. Establishing selection criteria for iPSC-derived cells, such as cell-specific markers, proliferation rate, lifespan, and genomic analyses, help minimize the variability of iPSC-derived cells. Just like ESCs, iPSCs are also predisposed to forming teratomas when undifferentiated. The available cell purification technologies often do not guarantee the complete eradication of undifferentiated iPSCs during manufacturing. Although many problems concerning the clinical applications of iPSCs still remain, iPSC-based therapies have a tremendous therapeutic potential for many diseases that are difficult to treat otherwise.

Advertisement

Acknowledgments

We are grateful for funding support from the National Institutes of Health (R21 AR074642 and U01AR075932). We also thank Epidermolysis Bullosa (EB) Research Partnership, the EB Medical Research Foundation, the Cure EB Charity, Dystrophic Epidermolysis Bullosa Research Association (DEBRA) International, and the Gates Frontiers Fund.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663-676. DOI: 10.1016/j.cell.2006.07.024
  2. 2. Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448(7151):313-317. DOI: 10.1038/nature05934
  3. 3. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861-872. DOI: 10.1016/j.cell.2007.11.019
  4. 4. Wernig M, Meissner A, Foreman R, Brambrink T, Ku M, Hochedlinger K, et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature. 2007;448(7151):318-324. DOI: 10.1038/nature05944
  5. 5. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, et al. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318(5858):1917-1920. DOI: 10.1126/science.1151526
  6. 6. Gepstein L. Derivation and potential applications of human embryonic stem cells. Circulation Research. 2002;91(10):866-876. DOI: 10.1161/01.res.0000041435.95082.84
  7. 7. Zomer HD, Vidane AS, Goncalves NN, Ambrosio CE. Mesenchymal and induced pluripotent stem cells: General insights and clinical perspectives. Stem Cells Cloning. 2015;8:125-134. DOI: 10.2147/SCCAA.S88036
  8. 8. Kimbrel EA, Lanza R. Pluripotent stem cells: The last 10 years. Regenerative Medicine. 2016;11(8):831-847. DOI: 10.2217/rme-2016-0117
  9. 9. Mitalipova M, Palmarini G. Isolation and characterization of human embryonic stem cells. Methods in Molecular Biology. 2006;331:55-76. DOI: 10.1385/1-59745-046-4:55
  10. 10. Takahashi K. Cellular reprogramming--lowering gravity on Waddington's epigenetic landscape. Journal of Cell Science 2012;125(Pt 11):2553-2560. DOI: 10.1242/jcs.084822
  11. 11. Briggs R, King TJ. Transplantation of living nuclei from blastula cells into enucleated Frogs' eggs. Proceedings of the National Academy of Sciences of the United States of America. 1952;38(5):455-463. DOI: 10.1073/pnas.38.5.455
  12. 12. Gurdon JB, Elsdale TR, Fischberg M. Sexually mature individuals of Xenopus laevis from the transplantation of single somatic nuclei. Nature. 1958;182(4627):64-65. DOI: 10.1038/182064a0
  13. 13. Wakayama T, Perry AC, Zuccotti M, Johnson KR, Yanagimachi R. Full-term development of mice from enucleated oocytes injected with cumulus cell nuclei. Nature. 1998;394(6691):369-374. DOI: 10.1038/28615
  14. 14. Ambrosi DJ, Rasmussen TP. Reprogramming mediated by stem cell fusion. Journal of Cellular and Molecular Medicine. 2005;9(2):320-330. DOI: 10.1111/j.1582-4934.2005.tb00358.x
  15. 15. Wilmut I, Schnieke AE, McWhir J, Kind AJ, Campbell KH. Viable offspring derived from fetal and adult mammalian cells. Nature. 1997;385(6619):810-813. DOI: 10.1038/385810a0
  16. 16. Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nature Genetics. 2000;24(4):372-376. DOI: 10.1038/74199
  17. 17. Ben-Shushan E, Thompson JR, Gudas LJ, Bergman Y. Rex-1, a gene encoding a transcription factor expressed in the early embryo, is regulated via Oct-3/4 and Oct-6 binding to an octamer site and a novel protein, Rox-1, binding to an adjacent site. Molecular and Cellular Biology. 1998;18(4):1866-1878. DOI: 10.1128/MCB.18.4.1866
  18. 18. Johansson H, Simonsson S. Core transcription factors, Oct4, Sox2 and Nanog, individually form complexes with nucleophosmin (Npm1) to control embryonic stem (ES) cell fate determination. Aging (Albany NY). 2010;2(11):815-822. DOI: 10.18632/aging.100222
  19. 19. Adhikary S, Eilers M. Transcriptional regulation and transformation by Myc proteins. Nature Reviews. Molecular Cell Biology. 2005;6(8):635-645. DOI: 10.1038/nrm1703
  20. 20. Fernandez PC, Frank SR, Wang L, Schroeder M, Liu S, Greene J, et al. Genomic targets of the human c-Myc protein. Genes & Development. 2003;17(9):1115-1129. DOI: 10.1101/gad.1067003
  21. 21. Rowland BD, Bernards R, Peeper DS. The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nature Cell Biology. 2005;7(11):1074-1082. DOI: 10.1038/ncb1314
  22. 22. Evans PM, Zhang W, Chen X, Yang J, Bhakat KK, Liu C. Kruppel-like factor 4 is acetylated by p300 and regulates gene transcription via modulation of histone acetylation. The Journal of Biological Chemistry. 2007;282(47):33994-34002. DOI: 10.1074/jbc.M701847200
  23. 23. Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi T, et al. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nature Biotechnology. 2008;26(1):101-106. DOI: 10.1038/nbt1374
  24. 24. Kim JB, Sebastiano V, Wu G, Arauzo-Bravo MJ, Sasse P, Gentile L, et al. Oct4-induced pluripotency in adult neural stem cells. Cell. 2009;136(3):411-419. DOI: 10.1016/j.cell.2009.01.023
  25. 25. Huangfu D, Osafune K, Maehr R, Guo W, Eijkelenboom A, Chen S, et al. Induction of pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nature Biotechnology. 2008;26(11):1269-1275. DOI: 10.1038/nbt.1502
  26. 26. Chou BK, Mali P, Huang X, Ye Z, Dowey SN, Resar LM, et al. Efficient human iPS cell derivation by a non-integrating plasmid from blood cells with unique epigenetic and gene expression signatures. Cell Research. 2011;21(3):518-529. DOI: 10.1038/cr.2011.12
  27. 27. Utikal J, Polo JM, Stadtfeld M, Maherali N, Kulalert W, Walsh RM, et al. Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature. 2009;460(7259):1145-1148. DOI: 10.1038/nature08285
  28. 28. Yang DH, Moss EG. Temporally regulated expression of Lin-28 in diverse tissues of the developing mouse. Gene Expression Patterns. 2003;3(6):719-726. DOI: 10.1016/s1567-133x(03)00140-6
  29. 29. Meir YJ, Li G. Somatic Reprogramming-Above and Beyond Pluripotency. Cells. 2021;10:11. DOI: 10.3390/cells10112888
  30. 30. Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of mouse induced pluripotent stem cells without viral vectors. Science. 2008;322(5903):949-953. DOI: 10.1126/science.1164270
  31. 31. Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K. Induced pluripotent stem cells generated without viral integration. Science. 2008;322(5903):945-949. DOI: 10.1126/science.1162494
  32. 32. Fusaki N, Ban H, Nishiyama A, Saeki K, Hasegawa M. Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome. Proceedings of the Japan Academy. Series B, Physical and Biological Sciences. 2009;85(8):348-362. DOI: 10.2183/pjab.85.348
  33. 33. Narsinh KH, Jia F, Robbins RC, Kay MA, Longaker MT, Wu JC. Generation of adult human induced pluripotent stem cells using nonviral minicircle DNA vectors. Nature Protocols. 2011;6(1):78-88. DOI: 10.1038/nprot.2010.173
  34. 34. Wang W, Lin C, Lu D, Ning Z, Cox T, Melvin D, et al. Chromosomal transposition of PiggyBac in mouse embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(27):9290-9295. DOI: 10.1073/pnas.0801017105
  35. 35. Rao MS, Malik N. Assessing iPSC reprogramming methods for their suitability in translational medicine. Journal of Cellular Biochemistry. 2012;113(10):3061-3068. DOI: 10.1002/jcb.24183
  36. 36. Guan J, Wang G, Wang J, Zhang Z, Fu Y, Cheng L, et al. Chemical reprogramming of human somatic cells to pluripotent stem cells. Nature. 2022;605(7909):325-331. DOI: 10.1038/s41586-022-04593-5
  37. 37. Kim D, Kim CH, Moon JI, Chung YG, Chang MY, Han BS, et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell. 2009;4(6):472-476. DOI: 10.1016/j.stem.2009.05.005
  38. 38. Warren L, Manos PD, Ahfeldt T, Loh YH, Li H, Lau F, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010;7(5):618-630. DOI: 10.1016/j.stem.2010.08.012
  39. 39. Judson RL, Babiarz JE, Venere M, Blelloch R. Embryonic stem cell-specific microRNAs promote induced pluripotency. Nature Biotechnology. 2009;27(5):459-461. DOI: 10.1038/nbt.1535
  40. 40. Kogut I, McCarthy SM, Pavlova M, Astling DP, Chen X, Jakimenko A, et al. High-efficiency RNA-based reprogramming of human primary fibroblasts. Nature Communications. 2018;9(1):745. DOI: 10.1038/s41467-018-03190-3
  41. 41. Raab S, Klingenstein M, Liebau S, Linta L. A comparative view on human somatic cell sources for iPSC generation. Stem Cells International. 2014;2014:768391. DOI: 10.1155/2014/768391
  42. 42. Polo JM, Anderssen E, Walsh RM, Schwarz BA, Nefzger CM, Lim SM, et al. A molecular roadmap of reprogramming somatic cells into iPS cells. Cell. 2012;151(7):1617-1632. DOI: 10.1016/j.cell.2012.11.039
  43. 43. Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell. 2005;122(6):947-956. DOI: 10.1016/j.cell.2005.08.020
  44. 44. Papapetrou EP, Tomishima MJ, Chambers SM, Mica Y, Reed E, Menon J, et al. Stoichiometric and temporal requirements of Oct4, Sox2, Klf4, and c-Myc expression for efficient human iPSC induction and differentiation. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(31):12759-12764. DOI: 10.1073/pnas.0904825106
  45. 45. Chan EM, Ratanasirintrawoot S, Park IH, Manos PD, Loh YH, Huo H, et al. Live cell imaging distinguishes bona fide human iPS cells from partially reprogrammed cells. Nature Biotechnology. 2009;27(11):1033-1037. DOI: 10.1038/nbt.1580
  46. 46. Aasen T, Raya A, Barrero MJ, Garreta E, Consiglio A, Gonzalez F, et al. Efficient and rapid generation of induced pluripotent stem cells from human keratinocytes. Nature Biotechnology. 2008;26(11):1276-1284. DOI: 10.1038/nbt.1503
  47. 47. Doi A, Park IH, Wen B, Murakami P, Aryee MJ, Irizarry R, et al. Differential methylation of tissue- and cancer-specific CpG island shores distinguishes human induced pluripotent stem cells, embryonic stem cells and fibroblasts. Nature Genetics. 2009;41(12):1350-1353. DOI: 10.1038/ng.471
  48. 48. Kim K, Zhao R, Doi A, Ng K, Unternaehrer J, Cahan P, et al. Donor cell type can influence the epigenome and differentiation potential of human induced pluripotent stem cells. Nature Biotechnology. 2011;29(12):1117-1119. DOI: 10.1038/nbt.2052
  49. 49. Bilousova G, Chen J, Roop DR. Differentiation of mouse induced pluripotent stem cells into a multipotent keratinocyte lineage. The Journal of Investigative Dermatology. 2011;131(4):857-864. DOI: 10.1038/jid.2010.364
  50. 50. Ohta S, Imaizumi Y, Okada Y, Akamatsu W, Kuwahara R, Ohyama M, et al. Generation of human melanocytes from induced pluripotent stem cells. PLoS One. 2011;6(1):e16182. DOI: 10.1371/journal.pone.0016182
  51. 51. Hewitt KJ, Shamis Y, Hayman RB, Margvelashvili M, Dong S, Carlson MW, et al. Epigenetic and phenotypic profile of fibroblasts derived from induced pluripotent stem cells. PLoS One. 2011;6(2):e17128. DOI: 10.1371/journal.pone.0017128
  52. 52. Veraitch O, Kobayashi T, Imaizumi Y, Akamatsu W, Sasaki T, Yamanaka S, et al. Human induced pluripotent stem cell-derived ectodermal precursor cells contribute to hair follicle morphogenesis in vivo. The Journal of Investigative Dermatology. 2013;133(6):1479-1488. DOI: 10.1038/jid.2013.7
  53. 53. Ebner-Peking P, Krisch L, Wolf M, Hochmann S, Hoog A, Vari B, et al. Self-assembly of differentiated progenitor cells facilitates spheroid human skin organoid formation and planar skin regeneration. Theranostics. 2021;11(17):8430-8447. DOI: 10.7150/thno.59661
  54. 54. Sebastiano V, Zhen HH, Haddad B, Bashkirova E, Melo SP, Wang P, et al. Human COL7A1-corrected induced pluripotent stem cells for the treatment of recessive dystrophic epidermolysis bullosa. Science Translational Medicine. 2014;6(264):264ra163. DOI: 10.1126/scitranslmed.3009540
  55. 55. Toyoshima KE, Ogawa M, Tsuji T. Regeneration of a bioengineered 3D integumentary organ system from iPS cells. Nature Protocols. 2019;14(5):1323-1338. DOI: 10.1038/s41596-019-0124-z
  56. 56. Lee J, Rabbani CC, Gao H, Steinhart MR, Woodruff BM, Pflum ZE, et al. Hair-bearing human skin generated entirely from pluripotent stem cells. Nature. 2020;582(7812):399-404. DOI: 10.1038/s41586-020-2352-3
  57. 57. Fine JD. Inherited epidermolysis bullosa. Orphanet Journal of Rare Diseases. 2010;5:12. DOI: 10.1186/1750-1172-5-12
  58. 58. Lapasset L, Milhavet O, Prieur A, Besnard E, Babled A, Ait-Hamou N, et al. Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes & Development. 2011;25(21):2248-2253. DOI: 10.1101/gad.173922.111
  59. 59. Madrid M, Sumen C, Aivio S, Saklayen N. Autologous induced pluripotent stem cell-based cell therapies: Promise, progress, and challenges. Current Protocols. 2021;1(3):e88. DOI: 10.1002/cpz1.88
  60. 60. Bajpai VK, Mistriotis P, Loh YH, Daley GQ , Andreadis ST. Functional vascular smooth muscle cells derived from human induced pluripotent stem cells via mesenchymal stem cell intermediates. Cardiovascular Research. 2012;96(3):391-400. DOI: 10.1093/cvr/cvs253
  61. 61. Orlova VV, Drabsch Y, Freund C, Petrus-Reurer S, van den Hil FE, Muenthaisong S, et al. Functionality of endothelial cells and pericytes from human pluripotent stem cells demonstrated in cultured vascular plexus and zebrafish xenografts. Arteriosclerosis, Thrombosis, and Vascular Biology. 2014;34(1):177-186. DOI: 10.1161/ATVBAHA.113.302598
  62. 62. Prasain N, Lee MR, Vemula S, Meador JL, Yoshimoto M, Ferkowicz MJ, et al. Differentiation of human pluripotent stem cells to cells similar to cord-blood endothelial colony-forming cells. Nature Biotechnology. 2014;32(11):1151-1157. DOI: 10.1038/nbt.3048
  63. 63. Clayton ZE, Tan RP, Miravet MM, Lennartsson K, Cooke JP, Bursill CA, et al. Induced pluripotent stem cell-derived endothelial cells promote angiogenesis and accelerate wound closure in a murine excisional wound healing model. Bioscience Reports. 2018;38(4). DOI: 10.1042/BSR20180563
  64. 64. Kachamakova-Trojanowska N, Stepniewski J, Dulak J. Human iPSCs-derived endothelial cells with mutation in HNF1A as a model of maturity-onset diabetes of the young. Cell. 2019;8(11):1440. DOI: 10.3390/cells8111440
  65. 65. Samuel R, Daheron L, Liao S, Vardam T, Kamoun WS, Batista A, et al. Generation of functionally competent and durable engineered blood vessels from human induced pluripotent stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(31):12774-12779. DOI: 10.1073/pnas.1310675110
  66. 66. Songstad AE, Wiley LA, Duong K, Kaalberg E, Flamme-Wiese MJ, Cranston CM, et al. Generating iPSC-derived choroidal endothelial cells to study age-related macular degeneration. Investigative Ophthalmology & Visual Science. 2015;56(13):8258-8267. DOI: 10.1167/iovs.15-17073
  67. 67. Cho H, Macklin BL, Lin YY, Zhou L, Lai MJ, Lee G, et al. iPSC-derived endothelial cell response to hypoxia via SDF1a/CXCR4 axis facilitates incorporation to revascularize ischemic retina. JCI Insight. 2020;5(6):e131828. DOI: 10.1172/jci.insight.131828
  68. 68. Sances S, Ho R, Vatine G, West D, Laperle A, Meyer A, et al. Human iPSC-derived endothelial cells and microengineered organ-Chip enhance neuronal development. Stem Cell Reports. 2018;10(4):1222-1236. DOI: 10.1016/j.stemcr.2018.02.012
  69. 69. Nakayama KH, Joshi PA, Lai ES, Gujar P, Joubert LM, Chen B, et al. Bilayered vascular graft derived from human induced pluripotent stem cells with biomimetic structure and function. Regenerative Medicine. 2015;10(6):745-755. DOI: 10.2217/rme.15.45
  70. 70. Generali M, Casanova EA, Kehl D, Wanner D, Hoerstrup SP, Cinelli P, et al. Autologous endothelialized small-caliber vascular grafts engineered from blood-derived induced pluripotent stem cells. Acta Biomaterialia. 2019;97:333-343. DOI: 10.1016/j.actbio.2019.07.032
  71. 71. Sayed N, Liu C, Ameen M, Himmati F, Zhang JZ, Khanamiri S, et al. Clinical trial in a dish using iPSCs shows lovastatin improves endothelial dysfunction and cellular cross-talk in LMNA cardiomyopathy. Science Translational Medicine. 2020;12(554):eaax9276. DOI: 10.1126/scitranslmed.aax9276
  72. 72. Dubois NC, Craft AM, Sharma P, Elliott DA, Stanley EG, Elefanty AG, et al. SIRPA is a specific cell-surface marker for isolating cardiomyocytes derived from human pluripotent stem cells. Nature Biotechnology. 2011;29(11):1011-1018. DOI: 10.1038/nbt.2005
  73. 73. Ward MC, Gilad Y. A generally conserved response to hypoxia in iPSC-derived cardiomyocytes from humans and chimpanzees. eLife. 2019;8:8. DOI: 10.7554/eLife.42374
  74. 74. Wu H, Yang H, Rhee JW, Zhang JZ, Lam CK, Sallam K, et al. Modelling diastolic dysfunction in induced pluripotent stem cell-derived cardiomyocytes from hypertrophic cardiomyopathy patients. European Heart Journal. 2019;40(45):3685-3695. DOI: 10.1093/eurheartj/ehz326
  75. 75. Drawnel FM, Boccardo S, Prummer M, Delobel F, Graff A, Weber M, et al. Disease modeling and phenotypic drug screening for diabetic cardiomyopathy using human induced pluripotent stem cells. Cell Reports. 2014;9(3):810-821. DOI: 10.1016/j.celrep.2014.09.055
  76. 76. Giacomelli E, Bellin M, Sala L, van Meer BJ, Tertoolen LG, Orlova VV, et al. Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells. Development. 2017;144(6):1008-1017. DOI: 10.1242/dev.143438
  77. 77. Pecha S, Yorgan K, Rohl M, Geertz B, Hansen A, Weinberger F, et al. Human iPS cell-derived engineered heart tissue does not affect ventricular arrhythmias in a Guinea pig cryo-injury model. Scientific Reports. 2019;9(1):9831. DOI: 10.1038/s41598-019-46409-z
  78. 78. Jiwlawat S, Lynch E, Glaser J, Smit-Oistad I, Jeffrey J, Van Dyke JM, et al. Differentiation and sarcomere formation in skeletal myocytes directly prepared from human induced pluripotent stem cells using a sphere-based culture. Differentiation. 2017;96:70-81. DOI: 10.1016/j.diff.2017.07.004
  79. 79. Quattrocelli M, Swinnen M, Giacomazzi G, Camps J, Barthelemy I, Ceccarelli G, et al. Mesodermal iPSC-derived progenitor cells functionally regenerate cardiac and skeletal muscle. The Journal of Clinical Investigation. 2015;125(12):4463-4482. DOI: 10.1172/JCI82735
  80. 80. Song KY, Guo XM, Wang HQ , Zhang L, Huang SY, Huo YC, et al. MBNL1 reverses the proliferation defect of skeletal muscle satellite cells in myotonic dystrophy type 1 by inhibiting autophagy via the mTOR pathway. Cell Death & Disease. 2020;11(7):545. DOI: 10.1038/s41419-020-02756-8
  81. 81. Sun C, Serra C, Lee G, Wagner KR. Stem cell-based therapies for Duchenne muscular dystrophy. Experimental Neurology. 2020;323:113086. DOI: 10.1016/j.expneurol.2019.113086
  82. 82. Wernig M, Zhao JP, Pruszak J, Hedlund E, Fu D, Soldner F, et al. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson's disease. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(15):5856-5861. DOI: 10.1073/pnas.0801677105
  83. 83. Lippmann ES, Al-Ahmad A, Azarin SM, Palecek SP, Shusta EV. A retinoic acid-enhanced, multicellular human blood-brain barrier model derived from stem cell sources. Scientific Reports. 2014;4:4160. DOI: 10.1038/srep04160
  84. 84. Canfield SG, Stebbins MJ, Faubion MG, Gastfriend BD, Palecek SP, Shusta EV. An isogenic neurovascular unit model comprised of human induced pluripotent stem cell-derived brain microvascular endothelial cells, pericytes, astrocytes, and neurons. Fluids Barriers CNS. 2019;16(1):25. DOI: 10.1186/s12987-019-0145-6
  85. 85. Schweitzer JS, Song B, Herrington TM, Park TY, Lee N, Ko S, et al. Personalized iPSC-derived dopamine progenitor cells for Parkinson's disease. The New England Journal of Medicine. 2020;382(20):1926-1932. DOI: 10.1056/NEJMoa1915872
  86. 86. Takahashi J. iPS cell-based therapy for Parkinson's disease: A Kyoto trial. Regenerative Therapy. 2020;13:18-22. DOI: 10.1016/j.reth.2020.06.002
  87. 87. Loring JF. Autologous induced pluripotent stem cell-derived neurons to treat Parkinson's disease. Stem Cells and Development. 2018;27(14):958-959. DOI: 10.1089/scd.2018.0107
  88. 88. Lahlou H, Lopez-Juarez A, Fontbonne A, Nivet E, Zine A. Modeling human early otic sensory cell development with induced pluripotent stem cells. PLoS One. 2018;13(6):e0198954. DOI: 10.1371/journal.pone.0198954
  89. 89. Takeda H, Hosoya M, Fujioka M, Saegusa C, Saeki T, Miwa T, et al. Engraftment of human pluripotent stem cell-derived progenitors in the inner ear of prenatal mice. Scientific Reports. 2018;8(1):1941. DOI: 10.1038/s41598-018-20277-5
  90. 90. Fukunaga I, Fujimoto A, Hatakeyama K, Kurebayashi N, Ikeda K, Kamiya K. Generation of functional CX26-gap-junction-plaque-forming cells with spontaneous Ca(2+) transients via a gap junction characteristic of developing cochlea. Current Protocols in Stem Cell Biology. 2019;51(1):e100. DOI: 10.1002/cpsc.100
  91. 91. Gunewardene N, Crombie D, Dottori M, Nayagam BA. Innervation of Cochlear hair cells by human induced pluripotent stem cell-derived neurons In vitro. Stem Cells International. 2016;2016:1781202. DOI: 10.1155/2016/1781202
  92. 92. Hayashi R, Ishikawa Y, Sasamoto Y, Katori R, Nomura N, Ichikawa T, et al. Co-ordinated ocular development from human iPS cells and recovery of corneal function. Nature. 2016;531(7594):376-380. DOI: 10.1038/nature17000
  93. 93. Shibata S, Hayashi R, Kudo Y, Okubo T, Imaizumi T, Katayama T, et al. Cell-type-specific adhesiveness and proliferation propensity on Laminin isoforms enable purification of iPSC-derived corneal epithelium. Stem Cell Reports. 2020;14(4):663-676. DOI: 10.1016/j.stemcr.2020.02.008
  94. 94. Parameswaran S, Balasubramanian S, Babai N, Qiu F, Eudy JD, Thoreson WB, et al. Induced pluripotent stem cells generate both retinal ganglion cells and photoreceptors: Therapeutic implications in degenerative changes in glaucoma and age-related macular degeneration. Stem Cells. 2010;28(4):695-703. DOI: 10.1002/stem.320
  95. 95. Zhu J, Reynolds J, Garcia T, Cifuentes H, Chew S, Zeng X, et al. Generation of transplantable retinal photoreceptors from a current good manufacturing practice-manufactured human induced pluripotent stem cell line. Stem Cells Translational Medicine. 2018;7(2):210-219. DOI: 10.1002/sctm.17-0205
  96. 96. Uyama H, Mandai M, Takahashi M. Stem-cell-based therapies for retinal degenerative diseases: Current challenges in the establishment of new treatment strategies. Development, Growth & Differentiation. 2021;63(1):59-71. DOI: 10.1111/dgd.12704
  97. 97. Yang JM, Chung S, Yun K, Kim B, So S, Kang S, et al. Long-term effects of human induced pluripotent stem cell-derived retinal cell transplantation in Pde6b knockout rats. Experimental & Molecular Medicine. 2021;53(4):631-642. DOI: 10.1038/s12276-021-00588-w
  98. 98. Takagi S, Mandai M, Gocho K, Hirami Y, Yamamoto M, Fujihara M, et al. Evaluation of transplanted autologous induced pluripotent stem cell-derived retinal pigment epithelium in exudative age-related macular degeneration. Ophthalmol Retina. 2019;3(10):850-859. DOI: 10.1016/j.oret.2019.04.021
  99. 99. Kim JY, Nam Y, Rim YA, Ju JH. Review of the current trends in clinical trials involving induced pluripotent stem cells. Stem Cell Reviews and Reports. 2022;18(1):142-154. DOI: 10.1007/s12015-021-10262-3
  100. 100. Bilousova G, Jun du H, King KB, De Langhe S, Chick WS, Torchia EC, et al. Osteoblasts derived from induced pluripotent stem cells form calcified structures in scaffolds both in vitro and in vivo. Stem Cells. 2011;29(2):206-216. DOI: 10.1002/stem.566
  101. 101. Hosseini FS, Soleimanifar F, Khojasteh A, Ardeshirylajimi A. Promoting osteogenic differentiation of human-induced pluripotent stem cells by releasing Wnt/beta-catenin signaling activator from the nanofibers. Journal of Cellular Biochemistry. 2019;120(4):6339-6346. DOI: 10.1002/jcb.27921
  102. 102. Ye JH, Xu YJ, Gao J, Yan SG, Zhao J, Tu Q , et al. Critical-size calvarial bone defects healing in a mouse model with silk scaffolds and SATB2-modified iPSCs. Biomaterials. 2011;32(22):5065-5076. DOI: 10.1016/j.biomaterials.2011.03.053
  103. 103. Ardeshirylajimi A, Dinarvand P, Seyedjafari E, Langroudi L, Adegani FJ, Soleimani M. Enhanced reconstruction of rat calvarial defects achieved by plasma-treated electrospun scaffolds and induced pluripotent stem cells. Cell and Tissue Research. 2013;354(3):849-860. DOI: 10.1007/s00441-013-1693-8
  104. 104. Dicks A, Wu CL, Steward N, Adkar SS, Gersbach CA, Guilak F. Prospective isolation of chondroprogenitors from human iPSCs based on cell surface markers identified using a CRISPR-Cas9-generated reporter. Stem Cell Research & Therapy. 2020;11(1):66. DOI: 10.1186/s13287-020-01597-8
  105. 105. Tang R, Jing L, Willard VP, Wu CL, Guilak F, Chen J, et al. Differentiation of human induced pluripotent stem cells into nucleus pulposus-like cells. Stem Cell Research & Therapy. 2018;9(1):61. DOI: 10.1186/s13287-018-0797-1
  106. 106. Bloor AJC, Patel A, Griffin JE, Gilleece MH, Radia R, Yeung DT, et al. Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: A phase I, multicenter, open-label, dose-escalation study. Nature Medicine. 2020;26(11):1720-1725. DOI: 10.1038/s41591-020-1050-x
  107. 107. Zhang J, Chen M, Liao J, Chang C, Liu Y, Padhiar AA, et al. Induced pluripotent stem cell-derived mesenchymal stem cells hold lower heterogeneity and great promise in biological research and clinical applications. Frontiers in Cell and Development Biology. 2021;9:716907. DOI: 10.3389/fcell.2021.716907
  108. 108. Malhotra N. Induced pluripotent stem (iPS) cells in dentistry: A review. International Journal of Stem Cells. 2016;9(2):176-185. DOI: 10.15283/ijsc16029
  109. 109. Seki D, Takeshita N, Oyanagi T, Sasaki S, Takano I, Hasegawa M, et al. Differentiation of odontoblast-like cells from mouse induced pluripotent stem cells by Pax9 and Bmp4 transfection. Stem Cells Translational Medicine. 2015;4(9):993-997. DOI: 10.5966/sctm.2014-0292
  110. 110. Araoka T, Mae S, Kurose Y, Uesugi M, Ohta A, Yamanaka S, et al. Efficient and rapid induction of human iPSCs/ESCs into nephrogenic intermediate mesoderm using small molecule-based differentiation methods. PLoS One. 2014;9(1):e84881. DOI: 10.1371/journal.pone.0084881
  111. 111. Imberti B, Tomasoni S, Ciampi O, Pezzotta A, Derosas M, Xinaris C, et al. Renal progenitors derived from human iPSCs engraft and restore function in a mouse model of acute kidney injury. Scientific Reports. 2015;5:8826. DOI: 10.1038/srep08826
  112. 112. Takasato M, Er PX, Chiu HS, Maier B, Baillie GJ, Ferguson C, et al. Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature. 2015;526(7574):564-568. DOI: 10.1038/nature15695
  113. 113. Figliuzzi M, Bonandrini B, Remuzzi A. Decellularized kidney matrix as functional material for whole organ tissue engineering. Journal of Applied Biomaterial and Functional Material. 2017;15(4):e326-ee33. DOI: 10.5301/jabfm.5000393
  114. 114. Shimizu T, Mae SI, Araoka T, Okita K, Hotta A, Yamagata K, et al. A novel ADPKD model using kidney organoids derived from disease-specific human iPSCs. Biochemical and Biophysical Research Communications. 2020;529(4):1186-1194. DOI: 10.1016/j.bbrc.2020.06.141
  115. 115. Abo KM, Ma L, Matte T, Huang J, Alysandratos KD, Werder RB, et al. Human iPSC-derived alveolar and airway epithelial cells can be cultured at air-liquid interface and express SARS-CoV-2 host factors. bioRxiv. 2020. DOI: 10.1101/2020.06.03.132639
  116. 116. Hawkins FJ, Suzuki S, Beermann ML, Barilla C, Wang R, Villacorta-Martin C, et al. Derivation of airway basal stem cells from human pluripotent stem cells. Cell Stem Cell. 2021;28(1):79-95 e8. DOI: 10.1016/j.stem.2020.09.017
  117. 117. Rock JR, Onaitis MW, Rawlins EL, Lu Y, Clark CP, Xue Y, et al. Basal cells as stem cells of the mouse trachea and human airway epithelium. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(31):12771-12775. DOI: 10.1073/pnas.0906850106
  118. 118. Ghaedi M, Calle EA, Mendez JJ, Gard AL, Balestrini J, Booth A, et al. Human iPS cell-derived alveolar epithelium repopulates lung extracellular matrix. The Journal of Clinical Investigation. 2013;123(11):4950-4962. DOI: 10.1172/JCI68793
  119. 119. Taniguchi D, Matsumoto K, Machino R, Takeoka Y, Elgalad A, Taura Y, et al. Human lung microvascular endothelial cells as potential alternatives to human umbilical vein endothelial cells in bio-3D-printed trachea-like structures. Tissue & Cell. 2020;63:101321. DOI: 10.1016/j.tice.2019.101321
  120. 120. Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle MA, Duris C, et al. Highly efficient generation of human hepatocyte-like cells from induced pluripotent stem cells. Hepatology. 2010;51(1):297-305. DOI: 10.1002/hep.23354
  121. 121. Mitani S, Takayama K, Nagamoto Y, Imagawa K, Sakurai F, Tachibana M, et al. Human ESC/iPSC-derived hepatocyte-like cells achieve zone-specific hepatic properties by modulation of WNT signaling. Molecular Therapy. 2017;25(6):1420-1433. DOI: 10.1016/j.ymthe.2017.04.006
  122. 122. Koui Y, Kido T, Ito T, Oyama H, Chen SW, Katou Y, et al. An In vitro human liver model by iPSC-derived parenchymal and non-parenchymal cells. Stem Cell Reports. 2017;9(2):490-498. DOI: 10.1016/j.stemcr.2017.06.010
  123. 123. Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature. 2013;499(7459):481-484. DOI: 10.1038/nature12271
  124. 124. Nagamoto Y, Takayama K, Ohashi K, Okamoto R, Sakurai F, Tachibana M, et al. Transplantation of a human iPSC-derived hepatocyte sheet increases survival in mice with acute liver failure. Journal of Hepatology. 2016;64(5):1068-1075. DOI: 10.1016/j.jhep.2016.01.004
  125. 125. Sin YY, Ballantyne LL, Richmond CR, Funk CD. Transplantation of gene-edited hepatocyte-like cells modestly improves survival of Arginase-1-deficient mice. Molecular Therapy and Nucleic Acids. 2018;10:122-130. DOI: 10.1016/j.omtn.2017.11.012
  126. 126. Jin M, Yi X, Liao W, Chen Q , Yang W, Li Y, et al. Advancements in stem cell-derived hepatocyte-like cell models for hepatotoxicity testing. Stem Cell Research & Therapy. 2021;12(1):84. DOI: 10.1186/s13287-021-02152-9
  127. 127. Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470(7332):105-109. DOI: 10.1038/nature09691
  128. 128. DeLaForest A, Quryshi AF, Frolkis TS, Franklin OD, Battle MA. GATA4 is required for budding morphogenesis of posterior foregut endoderm in a model of human stomach development. Frontiers in Medicine (Lausanne). 2020;7:44. DOI: 10.3389/fmed.2020.00044
  129. 129. Ito K, Matsuura K, Mihara Y, Sakamoto Y, Hasegawa K, Kokudo N, et al. Delivery of pancreatic digestive enzymes into the gastrointestinal tract by pancreatic exocrine tissue transplant. Scientific Reports. 2019;9(1):5922. DOI: 10.1038/s41598-019-42362-z
  130. 130. Workman MJ, Mahe MM, Trisno S, Poling HM, Watson CL, Sundaram N, et al. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system. Nature Medicine. 2017;23(1):49-59. DOI: 10.1038/nm.4233
  131. 131. Iwao T, Kodama N, Kondo Y, Kabeya T, Nakamura K, Horikawa T, et al. Generation of enterocyte-like cells with pharmacokinetic functions from human induced pluripotent stem cells using small-molecule compounds. Drug Metabolism and Disposition. 2015;43(4):603-610. DOI: 10.1124/dmd.114.062604
  132. 132. Ozawa T, Takayama K, Okamoto R, Negoro R, Sakurai F, Tachibana M, et al. Generation of enterocyte-like cells from human induced pluripotent stem cells for drug absorption and metabolism studies in human small intestine. Scientific Reports. 2015;5:16479. DOI: 10.1038/srep16479
  133. 133. McCracken KW, Howell JC, Wells JM, Spence JR. Generating human intestinal tissue from pluripotent stem cells in vitro. Nature Protocols. 2011;6(12):1920-1928. DOI: 10.1038/nprot.2011.410
  134. 134. Kitano K, Schwartz DM, Zhou H, Gilpin SE, Wojtkiewicz GR, Ren X, et al. Bioengineering of functional human induced pluripotent stem cell-derived intestinal grafts. Nature Communications. 2017;8(1):765. DOI: 10.1038/s41467-017-00779-y
  135. 135. Enderami SE, Kehtari M, Abazari MF, Ghoraeian P, Nouri Aleagha M, Soleimanifar F, et al. Generation of insulin-producing cells from human induced pluripotent stem cells on PLLA/PVA nanofiber scaffold. Artif Cells Nanomed Biotechnol. 2018;46(sup1):1062-1069. DOI: 10.1080/21691401.2018.1443466
  136. 136. Rajaei B, Shamsara M, Sanati MH. In vitro generation of glucose-responsive insulin-secreting cells from PDX1-overexpressing human-induced pluripotent stem cell derived from diabetic patient. ASAIO Journal. 2018;64(6):819-826. DOI: 10.1097/MAT.0000000000000728
  137. 137. Mohsen-Kanson T, Hafner AL, Wdziekonski B, Takashima Y, Villageois P, Carriere A, et al. Differentiation of human induced pluripotent stem cells into brown and white adipocytes: Role of Pax3. Stem Cells. 2014;32(6):1459-1467. DOI: 10.1002/stem.1607
  138. 138. Bonnefond A, Philippe J, Durand E, Dechaume A, Huyvaert M, Montagne L, et al. Whole-exome sequencing and high throughput genotyping identified KCNJ11 as the thirteenth MODY gene. PLoS One. 2012;7(6):e37423. DOI: 10.1371/journal.pone.0037423
  139. 139. Griscelli F, Ezanno H, Soubeyrand M, Feraud O, Oudrhiri N, Bonnefond A, et al. Generation of an induced pluripotent stem cell (iPSC) line from a patient with maturity-onset diabetes of the young type 3 (MODY3) carrying a hepatocyte nuclear factor 1-alpha (HNF1A) mutation. Stem Cell Research. 2018;29:56-59. DOI: 10.1016/j.scr.2018.02.017
  140. 140. Friesen M, Cowan CA. FPLD2 LMNA mutation R482W dysregulates iPSC-derived adipocyte function and lipid metabolism. Biochemical and Biophysical Research Communications. 2018;495(1):254-260. DOI: 10.1016/j.bbrc.2017.11.008
  141. 141. Matsubara M, Kanda H, Imamura H, Inoue M, Noguchi M, Hosoda K, et al. Analysis of mitochondrial function in human induced pluripotent stem cells from patients with mitochondrial diabetes due to the A3243G mutation. Scientific Reports. 2018;8(1):949. DOI: 10.1038/s41598-018-19264-7
  142. 142. Melton D. The promise of stem cell-derived islet replacement therapy. Diabetologia. 2021;64(5):1030-1036. DOI: 10.1007/s00125-020-05367-2
  143. 143. Jones PM, Persaud SJ. Beta-cell replacement therapy for type 1 diabetes: Closer and closer. Diabetic Medicine. 2022;39(6):e14834. DOI: 10.1111/dme.14834
  144. 144. Ono H, Obana A, Usami Y, Sakai M, Nohara K, Egusa H, et al. Regenerating salivary glands in the microenvironment of induced pluripotent stem cells. BioMed Research International. 2015;2015:293570. DOI: 10.1155/2015/293570
  145. 145. Zhang Y, Deng C, Qian J, Zhang M, Li X. Improvement of radiotherapy-induced lacrimal gland injury by induced pluripotent stem cell-derived conditioned medium via MDK and inhibition of the p38/JNK pathway. International Journal of Molecular Sciences. 2014;15(10):18407-18421. DOI: 10.3390/ijms151018407
  146. 146. Provin N, Giraud M. Differentiation of pluripotent stem cells into Thymic epithelial cells and generation of Thymic organoids: Applications for therapeutic strategies against APECED. Frontiers in Immunology. 2022;13:930963. DOI: 10.3389/fimmu.2022.930963
  147. 147. Li Y, Hong N, Zhang A, Chen W, Wang RH, Xu XL, et al. Enhancing mammary differentiation by overcoming lineage-specific epigenetic modification and signature gene expression of fibroblast-derived iPSCs. Cell Death & Disease. 2014;5:e1550. DOI: 10.1038/cddis.2014.499
  148. 148. Dai X, Wang X, Yang C, Huang M, Zhou Z, Qu Y, et al. Human fibroblasts facilitate the generation of iPSCs-derived mammary-like organoids. Stem Cell Research & Therapy. 2022;13(1):377. DOI: 10.1186/s13287-022-03023-7
  149. 149. Sugimoto N, Kanda J, Nakamura S, Kitano T, Hishizawa M, Kondo T, et al. iPLAT1: The first-in-human clinical trial of iPSC-derived platelets as a phase 1 autologous transfusion study. Blood. 2022;140:2398-2402. DOI: 10.1182/blood.2022017296
  150. 150. Sabbah M, Jondreville L, Lacan C, Norol F, Vieillard V, Roos-Weil D, et al. CAR-NK cells: A chimeric Hope or a promising therapy? Cancers (Basel). 2022;14(15):3839. DOI: 10.3390/cancers14153839

Written By

Igor Kizub, Andrii Rozhok and Ganna Bilousova

Submitted: 10 September 2022 Reviewed: 01 December 2022 Published: 28 December 2022