Open access peer-reviewed chapter

hiPSC-Based Tissue Organoid Regeneration

Written By

Ying Qu, Nur Yucer, Veronica J. Garcia, Armando E. Giuliano and Xiaojiang Cui

Submitted: 27 March 2018 Reviewed: 04 April 2018 Published: 06 June 2018

DOI: 10.5772/intechopen.76997

From the Edited Volume

Tissue Regeneration

Edited by Hussein Abdel hay El-Sayed Kaoud

Chapter metrics overview

2,502 Chapter Downloads

View Full Metrics

Abstract

Induced pluripotent stem cells (iPSCs) are generated from terminally differentiated cells and have the potential to differentiate to any organs originated from the embryonic germ layers. Extensive effort has been made to establish protocols for direct in vitro conversion of human iPSCs (hiPSCs) to different cell types/organs. Importantly, hiPSCs can be generated from patients with known genetic mutations that predispose to high-risks of specific disease development. Thus, the hiPSCs technology provides unlimited resources for creating patient-specific disease models. hiPSC-derived three-dimensional “organoid” models have recently emerged as a powerful tool to recapitulate the physiologically-relevant process of disease progression in vitro. In this chapter, we will discuss the current advancement of organoid regeneration from hiPSCs and the applications of hiPSCs-derived organoids. The limitations and challenges of this approach will also be discussed here.

Keywords

  • disease modeling
  • induced pluripotent stem cells
  • organoid
  • organ-on-chip
  • tissue regeneration

1. Introduction

Induced pluripotent stem cells (iPSCs), generated directly from terminally differentiated cells [1], can differentiate toward all three embryonic germ layers - ectoderm, mesoderm, and endoderm. iPSCs can give rise to diverse cell types such as neurons, cardiomyocytes, and hepatocytes under defined conditions [2, 3, 4], and thus may provide a useful tool for studying human organ development. Human iPSCs (hiPSCs) also open new avenues for patient-specific or personalized disease modeling and therapies [5]. In the following sections, we will summarize the current advances in hiPSC-derived organoid differentiation and discuss the applications of these hiPSC-derived organoids in pre-clinical and clinical areas.

Advertisement

2. Generation of hiPSC

2.1. Protocols for generating hiPSCs

Dr. Yamanaka first reported the generation of hiPSCs from fibroblasts using four transcriptional factors (POU5F1, SOX2, KLF4, and MYC) [6]. There are many protocols to further improve the original method. The first improvement was to minimizing the integration risks such as using non-integrating adenoviral vectors, transfection of mRNA, and using cell-penetrating peptide-tagged reprogramming factors [7]. Transgene-free hiPSC generation protocols have been published by multiple groups [8]. Using small molecules such as valproic acid, sodium butyrate, PD0325901, and others to create iPSCs has been reported [9, 10, 11]. Haase et al. reported a new non-transgenic protocol to generate hiPSCs from patient cord blood CD34+ cells using CytoTune™ Sendai reprogramming vectors under the exclusive usage of animal-derived component-free (ADCF) materials and components [12]. Recently, non-integrative and non-viral mRNA reprogramming technology has been reported for hiPSC generation [13]. Rapid, efficient, and safe strategies which are compliant with standard Good Manufacturing Practice (GMP) regulations pave the way for hiPSC clinical applications.

2.2. Genome editing of hiPSCs

Genome editing in hiPSCs provides a valuable tool for disease modeling, mechanism study, and gene therapy. A line of technology utilizing engineered nucleases consisting of sequence-specific DNA-binding domains attached to a non-specific DNA nuclease have been developed. These cutting-edge technologies allow researchers to manipulate entire genomes, including specific genes, intergenic regions, promoters, enhancers, silencers, and insulators. After zinc finger nucleases (ZFNs, first-generation) and transcription activator-like effector nucleases (TALENs, second-generation), the clustered regularly interspaced short palindromic repeats (CRISPR/Cas9) technology is the third-generation editing tool. Despite the difference in the nucleases, the common mechanisms involve inducing DNA double-strand breaks (DSBs) in targeted DNA. Compared to TALEN and ZFN, CRISPR/Cas9 has become the system of choice because of its features such as high feasibility, high affordability, and precise targeting.

Advertisement

3. hiPSC-based tissue regeneration

hiPSC-derived organoids are valuable resources and tools for disease modeling, organ development research, and therapy screening. The current established hiPSC-derived organoids are listed in Table 1 (adapted from Shi et al. [1]).

Organoids Applications Refs.
iPSC derived organoid model
Brain organoids Modeling autism disorder [14]
Modeling ALS disease [15]
Modeling Parkinson’s disease [16]
Modeling Zika virus infection [17]
Modeling Seckel syndrome [18]
Brain-region specific organoids Modeling Zika virus infection and human brain development disease [19]
Breast organoid Breast cancer research [20]
Cystic organoids Modeling Alagille syndrome, polycyctic liver disease and cystic fibrosis [21]
Fallopian tube organoids Ovarian cancer research [22]
Liver bud Organ-bud transplantation for regenerative medicine [23]
Lung organoids Lung development and lung disease modeling [22, 25]
Pancreas Pancreatic disease model [26]
Retinal organoids Modeling glaucoma [27]

Table 1.

Summary of hiPSC- and ESC-derived organoids, adapted from Shi et al. [1].

3.1. Ectoderm-derived tissues

Ectoderm is one of the three germ layers and the most exterior layer in the human embryo. It covers the outside of the embryo. The ectoderm gives rise to the central nervous system (the brain and spinal cord), the peripheral nervous system, the sensory epithelia of the eye, ear, and nose, the epidermis and its appendages (the nails and hair), the mammary glands, the hypophysis, the subcutaneous glands, and the enamel of the teeth (Figure 1).

Figure 1.

Summary of the organs originated from ectoderm.

3.1.1. Neuronal tissue regeneration

Several protocols have been developed for the de novo differentiation of hiPSC into cell types comprising the central nervous system (Figure 2). In general, protocols utilize either a monolayer culture condition, in which the neuroectoderm is further pushed toward spinal or cortical fates via neural rosette formation, or a three-dimensional culture system leading to the formation of neural organoids that again possess features of either cortical or spinal cell types. Here, we discuss the critical components of cortical and spinal organoid differentiation protocols.

Figure 2.

Specification of iPSC-derived neural tissue and exogenous factors used for derivation of organoids from iPS cells. Arrows indicate the temporal flow of tissue lineage as described by human development. Colored text denotes the small molecules that have been used to derive organoid models of each tissue (in gray) from iPSC spheroid or EB culture. Brain regions: CTX-Cortex and PNS: Peripheral nervous system. Growth factor and small molecules: BDNF, Brain-Derived Neurotrophic Factor; CNTF, Ciliary Neurotrophic Factor; CycA, Cyclophilin A; DSM, Dorsomorphin; EGF, Epidermal Growth Factor; FGF, Fibroblast Growth Factor; GDNF, Glial Cell-Derived Neurotrophic Factor; IWP2, WNT inhibitor; LDN, LDN-193189; NT3, Neurotrophin-3; SAG, Smoothened Agonist; SB, SB431542; SDF, Stromal cell-derived factor; SHH, Sonic Hedgehog; PMP, Purmorphamine (Adapted from Lullo and Krigstein [29]).

Successful formation of a cortical organoid depends upon the appropriate temporal- and regional-specific expression of several proteins and transcripts. The first method of this differentiation was presented by Lancaster et al. [28] in which the group relied upon intrinsic self-organization, a droplet of gelatinous matrix, and spinning bioreactors to drive neural-specific embryoid bodies toward developing cerebral structures.

Based on this initial protocol, several groups have published tangential methods that have improved the cortical organoid model. Pasca et al. [30] developed a differentiation method resulting in the generation of exclusively excitatory neurons reminiscent of the dorsal telencephalon, as well as the derivation of non-reactive astrocyte-like cells. Dual SMAD inhibition has been used to induce neural-ectoderm differentiation in suspension, which results in high-efficiency temporal and spatial organization of forebrain organoids [19, 30, 31]. Groups have also implemented transcriptional profiling to investigate the cell type composition of the mature organoid and to compare the overall maturity to that of the developing human brain [19, 30].

One of the most critical features for defining or characterizing a neuron from iPSCs is the neuron’s function and physiology. Many groups have assayed the physiology of the developed organoids, implementing calcium imaging or patch-clamp physiology to describe spontaneous activity and expression of specific neurotransmitter receptors [19, 28, 30]. With the addition of an exogenous matrix built of hydrogel, another model of cerebral organoids could reach a larger size over a shorter duration in culture and express the oligodendroglial-like marker Olig2 [32].

In addition to modeling the brain and its specific regions, neural organoids have also been developed for modeling the immature spinal cord and motor nerve units [33, 34]. Experimentation with different extracellular matrix components and rigidity led to the dorsal-ventral patterning of neural cysts within nine days of culture [33], resulting in the immature modeling of the human spinal cord. Another group described a method for generating motor nerve organoids that developed a polarized axon fascicle [34]. Although the above protocols have their limitations, it is evident that organoid technology is rapidly moving toward the goal of forming nervous systems.

3.1.2. Non-neural tissue regeneration

3.1.2.1. Skin

Skin is the largest organ of the body and provides a barrier to protect the interior from the external environment. Human skin is the first barrier system that is vital for homeostasis. Protocols using hiPSCs to generate human skin or skin components have been developed. Regeneration of human skin typically composed of 2D and 3D methods. Keratinocytes are major epithelial components in the skin. Researchers have developed protocols using defined medium and chemical/cytokines generate keratinocyte them. Itoh et al. used a combination of retinoic acid and bone morphogenetic protein (BMP) 4 to induce ectoderm epithelial cell differentiation from the patient-derived hiPSCs [35]. The same group later generated hiPSC-derived dermal fibroblasts, together with keratinocytes, to build 3D skin equivalents using an air-liquid interface culture [36]. The skin contains not only keratinocytes and fibroblasts, but also other skin appendages (eg. sweat glands, sebaceous glands, and hair follicle) and cells from different germ layer origins (fat cells, neurons, immune cells, muscles, blood vessels, and melanocytes. So far, the in vitro differentiation protocols have only been successful in developing dermal fibroblasts, keratinocytes, and melanocytes [37, 38]. Creating a full layer of skin tissue is still unfulfilled (Figure 3).

Figure 3.

The hiPSC-based regeneration strategy for a full-layered human skin.

3.1.2.2. Mammary gland

Mammary epithelial cells originate from non-neural/surface ectoderm cells, which co-exist with neural ectoderm cells at the same embryonic stage. Although mouse mammary gland development has been well studied, the human breast development is still poorly understood due to numerous differences between the mammary glands of the two species. In addition, questions regarding human mammary stem cell identity, mammary epithelial differentiation hierarchy, and the effects of ovarian hormones on mammary development are major obstacles for in vitro mammary gland regeneration.

Taking a cue from the understanding of human embryonic mammary gland development [39, 40], Qu et al. conceptualized that the first step for in vitro induction of mammary differentiation from hiPSCs was to pattern hiPSCs in to non-neural ectoderm, thus enriching mammary progenitors. The group developed a reliable two-step protocol to generate human mammary-like organoids from hiPSCs [20]. These organoids express luminal, basal, and breast-specific markers. Despite these novel findings, this in vitro system needs to be improved to fully recapitulate the formation of mammary ductal and alveolar structures.

3.2. Mesoderm-derived tissues

The mesoderm is formed through a process called gastrulation around the third week of embryonic development. Initially, mesoderm is segmented into three crucial compartments; the paraxial mesoderm (PM), the intermediate mesoderm (IM), and the lateral plate mesoderm (LPM) (Figure 4). The PM, also known as presomitic or somitic mesoderm, gives rise to embryonic structures of the sclerotome, myotome, and dermomyotome, which later develop into many adult tissues, including most of the axial skeleton, skeletal muscles, and connective tissues of the skin. The IM, which lies between the PM and LPM, differentiates into the urogenital duct system and gives rise to the kidneys, gonads, Wolffian (male) or Müllerian (female) ducts, fallopian tube, uterus, and the adrenal glands. The LPM is located on the side of the IM and is split horizontally into two layers: splanchnic mesoderm and somatic mesoderm. These layers contribute to the formation of the heart, blood vessels, and blood cells as well as to the connective tissue of the limbs and the space between these layers develop into the body cavity. This section summarizes the current hiPSC-derived 3D organoid differentiation research for tissues of mesodermal origin.

Figure 4.

Mesoderm subdivision and mesoderm-originated tissue development.

3.2.1. Kidney

Adult humans have a limited number of nephrons which do not increase during life but rather decay with age through attrition or disease. Currently, there is no known treatment accessible for nephron renewal in patients with chronic or end-stage kidney disease. Recent hiPSC-based tissue regeneration studies have provided the novel sources for nephron progenitor cell (NPC) production and potentially kidney regeneration. This section summarizes the current protocols to generate and maintain NPCs and 3D human kidney organoids.

The kidney is derived from IM which forms a urogenital ridge on either side of the aorta. Current multistep-directed differentiation methods are intended to recapitulate these crucial stages of renal embryonic development. Studies for the critical transcriptional regulation process and signaling pathways contribute to the better understanding of each stage of renal development (Figure 5). Importantly, these studies enable us to recognize the factors that direct cell fate decisions and have been the basis for establishing the current differentiation protocols and culture conditions [41, 42].

Figure 5.

Schematic representation of the differentiation stages into NPCs and kidney organoids and markers that identify each stage. GATA3, GATA Binding Protein 3; LAM, laminin; LHX1, LIM homeobox 1; LPM, lateral plate mesoderm; MIXL, Mix Paired-Like Homeobox; OCT4, POU class 5 homeobox1; OSR1, odd-skipped related transcription factor 1; PAX2, paired box 2; PAX8, paired box 8; SALL1, spalt-like transcription factor 1; SIX2, SIX homeobox 2; SOX2, SRY-box 2;WT1, Wilms tumor 1.

hiPSC-derived 3D human kidney organoids that led to the generation of more complex and partially self-organizing organoids [43, 44, 45, 46] have been established (Figure 6). Takasato et al. [43, 44] reported the generation of self-organizing kidney organoids using CHIR99021 and fibroblast growth factor (FGF) 9/heparin in a monolayer culture followed by cell pelleting. The transcriptional profiling of resulting organoids exhibits significant similarity to the first-trimester embryonic human kidney.

Figure 6.

Protocols for the directed differentiation of hiPSC into kidney organoid. The timescale is shown at the top (shortened after 20 days) and Day 0 marks the hiPSC stage. Oval images represent the obtained cell types-like in each stage, namely: Ep, epiblast; LPS, late primitive streak; NM, nascent mesoderm; PNM, posterior nascent mesoderm; PIM, posterior intermediate mesoderm; UE, ureteric epithelium; PA, pre-tubular aggregate; RV, renal vesicle. Growth factors and small molecules; BMP4, Bone Morphogenetic Protein bone 4; CHIR, CHIR99021; FGF, Fibroblast Growth Factor; RA, Retinoic Acid; VitD3, vitamin D3; WNT3A, Wingless Type Family member 3A; mTeSR1, defined iPSC medium and B27-Serum free cell culture supplements. Figure adapted from (Jacqueline Kai et al. [47]).

On the other hand, Morizane et al. [45] have patterned mesoderm into PM, IM, and LPMs and generated NPCs by mediating graded signals of GSK-3β inhibitor CHIR99021, Noggin, and Activin A. In this study, renal vesicle was formed by transiently treating the NPCs with the CHIR99021 and FGF9, following self-organizing differentiation into podocytes, proximal tubules, loop of Henle, and distal tubules in both 2D and 3D culture. Alternatively, a shorter and more straightforward protocol has been developed by the group of Bonventre and collaborators. Bonventre group et al. demonstrated the formation of nephron-like structures as well as endothelial-like cells that were arranged into cords and expressed the endothelial markers CD31 and von Willebrand factor [46].

3.2.2. Cardiomyocytes

Cardiovascular disease remains the leading cause of death worldwide. It encompasses an extensive range of clinical conditions due to genetics, physiologic and metabolic circumstances as well as drug toxicity. Most heart diseases are associated with severe damage to, or loss of, cardiomyocytes (CMs), and mammalian CMs have a limited regenerative capacity [48]. The recent advancements in the field of hiPSC-derived CMs (hiPSC-CM) offer unique opportunities for not only disease modeling and personalized drug efficacy/toxicity screening but also for stem cell-based cardiac regenerative therapy [49, 50, 51].

CMs arise from mesoderm, which is further specified into cardiac mesoderm and cardiac progenitor cells by three families of extracellular signaling molecules: WNT, FGF and TGFβ superfamily ligands (WNT3a, bone BMP4, Nodal and Activin A). The expression of these ligands in a spatiotemporal manner defines the mesodermal cell fate and prime CM differentiation [52, 53]. Several groups successfully mimic these signaling processes in vitro to generate hiPSC-CM, which is summarized in Table 2 (adapted from Smith et al. [54] and Burridge et al. [55]).

Method Culture condition Differentiation format Mesoderm induction Cardiac specification factors Cardiac Differentiation factors Ref.
Suspension EB in StemPro34 Knock-out serum Replacement (KSR)/FGF2 StemPRo 34 Activin A, BMP4
FGF2
VEGFA, DKK1 VEGFA, FGF2 [56]
VEGFA, DKK1, SB431542
Dorsomorphin
[57]
IWR1 Tri-iodothyrinine [58]
Monolayer Differentiation Monolayer on Matrigel with MEF RPM1 plus B27 Activin A, BMP4 RPM1 plus B27 RPM1 plus B27 [59]
Activin A, BMP4, FGF2 Noggin, RAi, DKK1 DKK1 [60]
RPM1 plus B27(−insulin) Activin A, BMP4, FGF2 VEGFA, DKK1 VEGFA, FGF2 [61]
(KSR)/FGF2 on MEF LI-APEL Activin A, BMP4, FGF2, VEGFA, SCF LI-BEL LI-BEL [62]
mTeSR RPM1 plus B27 Activin A, BMP4 IWR1 or IWP4 RPM1 plus B27 [63]
mTeSR+ROCKi RPM1 plus B27(−insulin) CHIR IWR1 or IWP4 RPM1 plus B27
Chemically defined E8 CDM3 CHIR/WNTC59 CDM3 CDM3 [65]

Table 2.

Methods for hiPSC-CM differentiations (adapted from Smith et al. [54] and Burridge et al. [55]).

The first generation of CM differentiation was established using ESCs and successfully applied to hiPSCs. While the traditional embryoid body protocol engaged serum-derived spontaneous differentiation into CM, its efficiency was very low, (∼1–5%). The second generation of CM differentiation aimed to recapitulate the embryonic developmental sequences in vitro by modulating stage-specific activation/inhibition of signaling pathways with recombinant protein, details described in Table 2. At the molecular level, each stage of iPSC-CM differentiation is characterized by sequential expression of specific sets of genes [53]. These protocols were much more efficient; however, they were expensive and exhibited high batch-to-batch variation.

The third-generation hiPSC-CM protocol is composed of sequential modulation of the Wnt signaling pathway: activation at an early stage with small molecules such as CHIR-99021 and then inhibition at a late stage with small molecules such as IWP2 [64, 65, 66]. These monolayer-based directed differentiation protocols generate CMs with high efficiency. On the other hand, maturation of these CMs became a major challenge for the use of de novo CMs in heart research, especially for disease modeling and drug testing [67, 68]. Thus, several studies used prolonged cell culture, electrical stimulation, mechanical stretch or hormonal stimulations to induce CM maturity [69, 70, 71, 72].

hiPSC-CM technology has transformed the field of cardiovascular research, especially the study of inherited and acquired cardiovascular diseases. Several heart diseases including long QT syndromes, catecholaminergic polymorphic ventricular tachycardia, and familial hypertrophic cardiomyopathy have been modeled using hiPSC-CM [73, 74, 75]. Patient-specific CM regeneration may hold the promise for stem cell-based cardiac therapy.

3.2.3. Fallopian tube

Ovarian cancer is the leading cause of gynecologic cancer-related deaths in the United States. Fallopian tube epithelia (FTE) has been identified as the origin of ovarian cancer [76]. The discovery of serous tubal intraepithelial carcinoma (STIC) lesions, a preneoplastic finding in the fallopian tube fimbriae of patients with BRCA mutations, supports the model of FTE origin of serous “ovarian” carcinoma [77].

Yucer et al. [22] developed a hiPSC-derived 3D human FTE model, mimicking the FTE development process via various intermediate stages toward mature FTE in 3D organoid culture. Female reproductive tract structures including fallopian tube epithelium arise from the Müllerian duct in parallel to the urinary system from IM of the urogenital ridge in the posterior primitive streak. Therefore, Yucer et al. [22] recapitulated Müllerian development starting with IM generation and further developed into fallopian tube epithelial precursors using pro-Müllerian growth factors. Each step of this differentiation is monitored through the expression of established markers (Figure 7). Further differentiation of the fallopian tube epithelial lineage was attained on a 3D growth platform, which enables the FTE organoid to self-organize into a convoluted luminal structure with secretory and ciliated cellular components [22].

Figure 7.

Schematic of iPSC derive FTE organoid model. The stepwise differentiation of FTE via various intermediate stages which are characterized by specific molecular signatures. BMP4, Bone Morphogenetic Protein bone 4; CHIR, CHIR99021; E2, Estrogen: P4, Progesterone; WNT, Wingless Type Family member.

hiPSC-derived 3D-FTE organoids model offers a faithful in vitro platform to investigate the fallopian tube origin of ovarian cancer and to explore early cancer pathogenesis and progression. This platform can also be used to study high-risk germline mutations including BRCA1/2, to identify the molecular signature and genetic alteration involved in carcinogenesis and ultimately uncover novel drug targets.

3.3. Endoderm-derived tissues

The endoderm gives rise to the epithelial lining of the gastrointestinal and respiratory tracts; the parenchyma of the tonsils, the liver, the thymus, the thyroid, the parathyroids, and the pancreas; the epithelial lining of the urinary bladder and urethra; and the epithelial lining of the tympanic cavity, tympanic antrum, and auditory tube (Figure 8).

Figure 8.

Summary of organs originated from endoderm.

3.3.1. Small intestine, stomach, and colon

The small intestinal tissue is composed of a single-layer of epithelial cells which form a lumen that is surounded surrounded by connective tissue. Functionally, the small intestine plays a central role in digestion and absorption of nutrients. There are different cell lineages (enterocytes, goblet cells, Paneth cells, tuft cells and enteroendocrine cells) in the small intestine with various functions such as exocrine, absorption, and protection. While diseases of the small intestine, such as tumor, inflammatory bowel disease (IBD), lactose intolerance are common, the study of these diseases have encountered difficulties due to the limitation of in vitro modeling systems.

The protocols used for intestinal organoid differentiation have been published by different groups [78, 79]. In general, hiPSCs are directed to differentiate into definitive endoderm followed by intestinal fate specification and development using Wnt3A and FGF4. Intestinal organoids, usually cultured in Matrigel, show a polarized, columnar epithelium that is patterned into villus-like structures and crypt-like proliferative zones that expresses intestinal stem cell markers. The epithelium contain functional enterocytes, as well as goblet, Paneth and enteroendocrine cells with a layer of mesenchymal cells. Yu et al. [80] reported a refined, non-Matrigel scaffold and 3D intestinal organoid culture protocol. The matrix-free system may improve the yield, decrease the time, and facilitate high-throughput approaches. The protocols used to generate intestines from hiPSCs are summarized in Figure 9.

Figure 9.

The protocols used to generate intestinal organoids using hiPSCs.

Gastric ulcer and gastric cancer affect 10% of the world’s population and there is no experimental model of the normal human gastric mucosa. The lack of proper models has hindered mechanistic studies, preventive approach testing, and disease modeling. Kyle et al. developed the first protocol directing hiPSCs to 3D gastric organoids by manipulating FGF, WNT, BMP, retinoic acid and EGF signaling pathways [81]. These organoids formed primitive gastric gland- and pit-like domains, proliferative zones containing LGR5-expressing cells, surface and antral mucous cells, and a diversity of gastric endocrine cells.

Recent studies have successfully generated colonic organoids from hiPSCs. Following the similar differentiation path to intestinal organoids, hiPSCs were sequentially differentiated into definitive endoderm, hindgut endoderm, and colonic organoids. The colonic organoid differentiation was conducted in a Matrigel 3D culture. Jorge et al. [82] modified the protocol used for small intestinal organoid differentiation by adding BMP2 in the Matrigel culture stage. BMP signaling can promote posterior fate in human gut tube cultures. Another group [83] reported a different approach by supplementing the inhibitors CHIR99021 and LDN193189, and EGF in the Matrigel culture stage. These hiPSCs-derived colonic organoids exhibit crypt-like structure formed by a polarized epithelium consisting of colon stem cells, goblet, and endocrine cells and a layer of supportive mesodermal tissue.

3.3.2. Lung

The regeneration of lung epithelial cells/organoids has applications in regenerative medicine, modeling of lung disease, drug screening and studies of human lung development. The lung is composed of endoderm-derived epithelial cells surrounded by mesenchymal-derived stromal cells. Lung epithelial cell differentiation follows the path of definitive endoderm to anterior foregut endoderm. Then the Nkx2.1+ endoderm will bud from the ventral side of the anterior foregut to form the primitive lung bud, which will form the respiratory tree. Signals from the mesenchyme to the epithelium are critical in cell specification, determination, and differentiation, and are essential for proper development and maturation of the lung [84].

A number of studies show the differentiation of hiPSCs into lung epithelial cells in 2D [84, 85, 86, 87, 88]. However, 3D lung organoid differentiation has become the trend. Briana et al. [24] reported a breakthrough in the stepwise differentiation of human lung organoids from hiPSCs that consist of both epithelial and mesenchymal components. These lung organoids possess upper airway-like epithelium with basal cells and immature ciliated cells surrounded by smooth muscle and myofibroblasts as well as an alveolar-like domain with appropriate cell types. Later, Chen et al. reported the generation of lung bud organoids (LBOs) that contain mesoderm and pulmonary endoderm and develop into branching airway and early alveolar structures after xenotransplantation and Matrigel 3D culture [89].

The application of acellular lung matrices has been reported in 3D lung tissue reconstruction. Decellularized lung matrix supports the culture and lineage commitment of hiPSC-derived lung progenitor cells [90]. The rotating bioreactor was also used to provide an air-liquid interface, which is a potent inducer of type I epithelial differentiation for both hiPSC-Alveolar epithelial type (AT) II and ATI cells [91]. The bioreactor system provides a method for large-scale production of alveolar epithelium for tissue engineering and drug discovery. Another improvement for lung regeneration from hiPSCs is the use of biomaterials [25].

Advertisement

4. Application of iPSC-based tissue regeneration

hiPSC-derived cells or organoids are becoming promising resources for disease modeling and therapeutical applications. In general, somatic cells from patients can be reprogrammed to hiPSCs. In turn, patient-specific hiPSCs can be converted into target organs using established protocols. These in vitro derived organs can be used for multiple purposes, including patient-specific disease modeling, drug testing, therapy screening, and transplantation.

4.1. Personalized disease modeling

The biggest advantage of the hiPSC technology lies in its patient-specific feature. hiPSC-derived 3D organoid models have recently emerged as a powerful tool to recapitulate and investigate the physiologically-relevant process of disease onset and progression in vitro. This model system leverages the self-renewal and multi-lineage differentiation capability of multipotent stem cells and their intrinsic self-organization regenerative ability to form 3D tissue architecture. Importantly, hiPSCs can be derived from patients with known hereditary genetic mutations that are associated with a higher risk of a particular disease. This provides a valuable approach to determine whether additional genetic alterations are needed to interact with the known mutations, thereby contributing to disease susceptibility, initiation, and progression [92].

Several hiPSC-derived, inherited human disease models have been used to reproduce cancers associated with those high-risk patients [93, 94]. A hiPSC-derived osteosarcoma model for Li-Fraumeni syndrome has yielded promising results in displaying disease pathogenesis and carcinogenesis events commonly found in relevant human cells [95]. Cystic Fibrosis (CF) is an inherit disease of secretory glands. Among all the organs, pancreas is the earliest and most severely affected organs impacted by CF. hiPSC-derived pancreatic epithelial cells can be used to study personalized CF development [96]. Kyle et al. [81] used hiPSC-derived gastric organoids to model the pathophysiological response of the human stomach to H. pylori infection. In addition, Miguel et al. reported using hiPSC-derived colonic organoids to model family APC mutation-associated colon cancer initiation [83]. More and more hiPSCs-based disease models will be established.

4.2. Therapeutic applications

4.2.1. Drug screening

Organoids differentiated from patient-derived hiPSCs can be used to build a screening platform to develop and validate therapeutic approaches. hiPSC-derived organoids have a line of features that make them suitable models. Using a defined protocol, hiPSC-derived organoids become an unlimited resource for a specific patient. The in vitro direction of organ differentiation allows the rapid and robust generation of organoids with identical features. Most importantly, the organoids are 3D based mini-tissues that consist of multiple cell types, and that recapitulate the tissue structures in vivo. Thus, the drug screening results are more applicable in vivo. As an example, hiPSC-based drug screening for Huntington’s disease has been established [97] developed. The applications of hiPSCs that have been reprogramed from patients of heritable, genetic diseases has been summarized by Wonhee Suh in a review paper [98].

Biomimetic tissues on a chip have been developed for drug discovery [99]. Organ-on-a-chip is based on microfluidic technology and has been proposed as a novel cell-based assay tool in pre-clinical studies. Furthermore, the concept of body-on-a-chip, which is stands for multiple organs connected through microfluid devices, can mimic multiple interactions between organs [100]. Applying hiPSC research to the concept of organ-on-a-chip has provided a promising future for the development of the patient-specific body-on-a-chip [101]. Drug screening is no longer a process that is limited by the responses of targeted organ, it can also provide an evaluation of systemic responses.

4.2.2. Gene therapy

The nature of the disease and desired genetic modification, efficiency and accuracy of gene repair methodology, as well as cell state will determine the success of gene therapy [102]. In theory, monogenic diseases dictated by a dysfunctional copy of the causative gene would be reversed by introducing a wild-type copy of the gene into cells [103, 104]. Over 80% of rare diseases are considered to have a genetic origin [105], which means the precise gene editing technologies can be practically used to correct these genetic factors. The application of genome editing technologies in therapeutic trials have been reported in many diseases, such as retinal diseases [106], lysosomal storage diseases [107], arthritis [108], and neurological disorders [109, 110]. In contrast, polygenic diseases that require simultaneous multiple alterations of the genome are more challenging to treat with gene therapy [111].

Gang et al. presented a highly efficient and reproducible protocol to edit the genome of hiPSCs through the combined use of the CRISPR/Cas9 RNA-guided nuclease and piggyBac (bacterial artificial chromosome) transposase [112]. Their method can result in efficient, targeted genome editing and concurrent “scarless” transgene excision. Satoru et al. reported using gene editing with engineered site-specific endonuclease technology to treat dominant-negative disorders by targeting only the mutant allele while leaving the normal allele intact [113]. Using precise gene editing technology to correct gene mutations from hiPSCs generated from patients combined with hiPSC differentiation into target cells/organs for transplantation provides an immense promise for the future of gene therapy (Figure 10).

Figure 10.

The summary of gene therapy applying precise genome editing technology in hiPSCs.

4.2.3. Transplantation

Given that hiPSCs are pluripotent stem cells which can be propagated unlimitedly and protocols for their differentiation into different cells/organoids have been established, hiPSC-derived micro-tissues are a potentially innovative material source for transplantation. In addition, immune rejection will be minimized when essentially returning the hiPSC-derived tissue to the original patient. For mature cells that have no or limited regenerative ability, such as cardiomyocytes, neurons, and pancreatic cells, hiPSC-derived cell/organoids are especially valuable for tissue repair. There are a series of clinical studies evaluating hiPSC-cells/organoids for treatment of neural degeneration, diabetes, heart failure, and retinal cells [114]. Although research on the application of hiPSCs in therapy have shown encouraging progress, there are some concerns involving the safety of hiPSC-based cell transplantation. Tumor risk and acquired gene mutations are major concerns.

Advertisement

5. Future and challenges

The original protocol to generate hiPSCs involves four transcriptional factors, but this method is not suitable because of its effect on genome integrity via the introduction of additional plasmids with exogenous genes. To make hiPSCs and their derivatives applicable for clinical uses, many improvements have been made to optimize the method for iPSC generation. The integration-free and chemical reprogramming protocols have been developed to minimize the risk of jeopardizing genome integrity [115, 116].

In general, the genetic nature of a disease, the molecular editing platform used, the delivery method, and the targeted cells and organs are all factors that influence the efficacy of treatment and determine the likelihood of clinical benefits [117]. The CRISPR/Cas9 molecular scissor system has been used to edit the genomes of a diverse array of mammalian cell types and organisms with high efficiency and precision. Determining and overcoming the actual frequency of off-target activities is challenging, yet critical to the application of the technology in gene therapy. CRISPR/Cas9 technology allows the study of complex genetic diseases, including human cancer, in which multiple mutations and chromosomal translocations are present in the genome [118, 119].

The potential application of hiPSC technology in cancer studies has been proposed, based on the idea of reprogramming cancer cells via hiPSC technology to cancer stem cell (CSC) state. CSCs are well-known as the origin of tumor development, the seeds for distant metastasis, and are critical in therapeutic resistance. Reprogramming the malignant cells back to their original state before the oncogenic transformation occurs [120], may provide tools for exploring the mechanisms of tumor initiation and progression in vitro, for studying the heterogeneity and origin of CSCs, and for producing cancer type-specific drug discovery. However, these reprogramming methods remain a challenge because of the cancer-specific epigenetic state and chromosomal aberrations of cancer cells.

Advertisement

Acknowledgments

This work was supported by the National Institutes of Health (2R01CA151610), the Fashion Footwear Charitable Foundation of New York, Inc., the Margie and Robert E. Petersen Foundation, and the Linda and Jim Lippman Research Fund.

Advertisement

Conflict of interest

All the authors declare no conflict of interest.

References

  1. 1. Shi Y, Inoue H, Wu JC, Yamanaka S. Induced pluripotent stem cell technology: A decade of progress. Nature Reviews. Drug Discovery. 2016;16(2):115-130
  2. 2. Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Ishige N. Transcription factors and medium suitable for initiating the differentiation of human induced pluripotent stem cells to the hepatocyte lineage. Journal of Cellular Biochemistry. 2016;117(9):2001-2009
  3. 3. Sareen D, Gowing G, Sahabian A, Staggenborg K, Paradis R, Avalos P, et al. Human induced pluripotent stem cells are a novel source of neural progenitor cells (iNPCs) that migrate and integrate in the rodent spinal cord. The Journal of Comparative Neurology. 2014;522:2707-2728
  4. 4. Kawamura T, Miyagawa S, Fukushima S, Maeda A, Kashiyama N, Kawamura A, et al. Cardiomyocytes Derived from MHC-homozygous induced pluripotent stem cells exhibit reduced allogeneic immunogenicity in MHC-matched non-human primates. Stem Cell Reports. 2016;6:312-320
  5. 5. Sayed N, Liu C, Wu JC. Translation of human-induced pluripotent stem cells: From clinical trial in a dish to precision medicine. Journal of the American College of Cardiology. 2016;67:2161-2176
  6. 6. 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;131:861-872
  7. 7. Hayes M, Zavazava N. Strategies to generate induced pluripotent stem cells. Methods in Molecular Biology. 2013;1029:77-92
  8. 8. Zhou H, Martinez H, Sun B, Li A, Zimmer M, Katsanis N, et al. Rapid and efficient generation of transgene-free iPSC from a small volume of cryopreserved blood. Stem Cell Reviews. 2015;11:652-665
  9. 9. Pandian GN, Sato S, Anandhakumar C, Taniguchi J, Takashima K, Syed J, et al. Identification of a small molecule that turns ON the pluripotency gene circuitry in human fibroblasts. ACS Chemical Biology. 2014;9:2729-2736
  10. 10. Petkov S, Hyttel P, Niemann H. The small molecule inhibitors PD0325091 and CHIR99021 reduce expression of pluripotency-related genes in putative porcine induced pluripotent stem cells. Cellular Reprogramming. 2014;16:235-240
  11. 11. Jung DW, Kim WH, Williams DR. Reprogram or reboot: Small molecule approaches for the production of induced pluripotent stem cells and direct cell reprogramming. ACS Chemical Biology. 2014;9:80-95
  12. 12. Haase A, Gohring G, Martin U. Generation of non-transgenic iPS cells from human cord blood CD34(+) cells under animal component-free conditions. Stem Cell Research. 2017;21:71-73
  13. 13. Artero Castro A, Leon M, Del Buey Furio V, Erceg S, Lukovic D. Generation of a human iPSC line by mRNA reprogramming. Stem Cell Research. 2018;28:157-160
  14. 14. Mariani J, Coppola G, Zhang P, Abyzov A, Provini L, Tomasini L, et al. FOXG1-dependent dysregulation of GABA/glutamate neuron differentiation in autism Spectrum disorders. Cell. 2015;162:375-390
  15. 15. Sances S, Bruijn LI, Chandran S, Eggan K, Ho R, Klim JR, et al. Modeling ALS with motor neurons derived from human induced pluripotent stem cells. Nature Neuroscience. 2016;19:542-553
  16. 16. Miller JD, Ganat YM, Kishinevsky S, Bowman RL, Liu B, Tu EY, et al. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell. 2013;13:691-705
  17. 17. Garcez PP, Loiola EC, Madeiro da Costa R, Higa LM, Trindade P, Delvecchio R, et al. Zika virus impairs growth in human neurospheres and brain organoids. Science. 2016;352:816-818
  18. 18. Gabriel E, Wason A, Ramani A, Gooi LM, Keller P, Pozniakovsky A, et al. CPAP promotes timely cilium disassembly to maintain neural progenitor pool. The EMBO Journal. 2016;35:803-819
  19. 19. Qian X, Nguyen HN, Song MM, Hadiono C, Ogden SC, Hammack C, et al. Brain-region-specific Organoids using mini-bioreactors for modeling ZIKV exposure. Cell. 2016;165:1238-1254
  20. 20. Qu Y, Han B, Gao B, Bose S, Gong Y, Wawrowsky K, et al. Differentiation of human induced pluripotent stem cells to mammary-like organoids. Stem Cell Reports. 2017;8:205-215
  21. 21. Sampaziotis F, de Brito MC, Madrigal P, Bertero A, Saeb-Parsy K, Soares FAC, et al. Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation. Nature Biotechnology 2015;33:845-852
  22. 22. Yucer N, Holzapfel M, Jenkins Vogel T, Lenaeus L, Ornelas L, Laury A, et al. Directed differentiation of human induced pluripotent stem cells into fallopian tube epithelium. Scientific Reports. 2017;7:10741
  23. 23. 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;499:481-484
  24. 24. Dye BR, Hill DR, Ferguson MA, Tsai YH, Nagy MS, Dyal R, et al. In vitro generation of human pluripotent stem cell derived lung organoids. eLife. 2015:4
  25. 25. Dye BR, Dedhia PH, Miller AJ, Nagy MS, White ES, Shea LD, et al. A bioengineered niche promotes in vivo engraftment and maturation of pluripotent stem cell derived human lung organoids. eLife. 2016;5
  26. 26. Hohwieler M, Illing A, Hermann PC, Mayer T, Stockmann M, Perkhofer L, et al. Human pluripotent stem cell-derived acinar/ductal organoids generate human pancreas upon orthotopic transplantation and allow disease modelling. Gut. 2017;66:473-486
  27. 27. Tucker BA, Solivan-Timpe F, Roos BR, Anfinson KR, Robin AL, Wiley LA, et al. Duplication of TBK1 stimulates autophagy in iPSC-derived retinal cells from a patient with normal tension Glaucoma. Journal of Stem Cell Research Therapy. 2014;3:161
  28. 28. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, et al. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501:373-379
  29. 29. Di Lullo E, Kriegstein AR. The use of brain organoids to investigate neural development and disease. Nature Reviews. Neuroscience. 2017;18:573-584
  30. 30. Pasca AM, Sloan SA, Clarke LE, Tian Y, Makinson CD, Huber N, et al. Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nature Methods. 2015;12:671-678
  31. 31. Kadoshima T, Sakaguchi H, Nakano T, Soen M, Ando S, Eiraku M, et al. Self-organization of axial polarity, inside-out layer pattern, and species-specific progenitor dynamics in human ES cell-derived neocortex. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:20284-20289
  32. 32. Lindborg BA, Brekke JH, Vegoe AL, Ulrich CB, Haider KT, Subramaniam S, et al. Rapid induction of cerebral organoids from human induced pluripotent stem cells using a chemically defined hydrogel and defined cell culture medium. Stem Cells Translational Medicine. 2016;5:970-979
  33. 33. Ranga A, Girgin M, Meinhardt A, Eberle D, Caiazzo M, Tanaka EM, et al. Neural tube morphogenesis in synthetic 3D microenvironments. Proceedings of the National Academy of Sciences of the United States of America. 2016;113:E6831-E68E9
  34. 34. Kawada J, Kaneda S, Kirihara T, Maroof A, Levi T, Eggan K, et al. Generation of a motor nerve organoid with human stem cell-derived neurons. Stem Cell Reports. 2017;9:1441-1449
  35. 35. Itoh M, Kiuru M, Cairo MS, Christiano AM. Generation of keratinocytes from normal and recessive dystrophic epidermolysis bullosa-induced pluripotent stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:8797-8802
  36. 36. Itoh M, Umegaki-Arao N, Guo Z, Liu L, Higgins CA, Christiano AM. Generation of 3D skin equivalents fully reconstituted from human induced pluripotent stem cells (iPSCs). PLoS One. 2013;8:e77673
  37. 37. Kawakami T, Okano T, Takeuchi S, Osumi K, Soma Y, Itoh M, et al. Approach for the derivation of melanocytes from induced pluripotent stem cells. The Journal of Investigative Dermatology. 2018;138:150-158
  38. 38. 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:e16182
  39. 39. Propper AY, Howard BA, Veltmaat JM. Prenatal morphogenesis of mammary glands in mouse and rabbit. Journal of Mammary Gland Biology and Neoplasia. 2013;18:93-104
  40. 40. Mikkola ML. Genetic basis of skin appendage development. Seminars in Cell and Developmental Biology. 2007;18:225-236
  41. 41. Kaminski MM, Tosic J, Pichler R, Arnold SJ, Lienkamp SS. Engineering kidney cells: Reprogramming and directed differentiation to renal tissues. Cell and Tissue Research. 2017;369:185-197
  42. 42. Krause M, Rak-Raszewska A, Pietila I, Quaggin SE, Vainio S. Signaling during kidney development. Cell. 2015;4:112-132
  43. 43. Takasato M, Er PX, Becroft M, Vanslambrouck JM, Stanley EG, Elefanty AG, et al. Directing human embryonic stem cell differentiation towards a renal lineage generates a self-organizing kidney. Nature Cell Biology. 2014;16:118-126
  44. 44. 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;526:564-568
  45. 45. Morizane R, Lam AQ, Freedman BS, Kishi S, Valerius MT, Bonventre JV. Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nature Biotechnology. 2015;33:1193-1200
  46. 46. Freedman BS, Brooks CR, Lam AQ, Fu H, Morizane R, Agrawal V, et al. Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nature Communications. 2015;6:8715
  47. 47. Chuah JKC, Zink D. Stem cell-derived kidney cells and organoids: Recent breakthroughs and emerging applications. Biotechnology Advances. 2017;35:150-167
  48. 48. Harvey PA, Leinwand LA. The cell biology of disease: Cellular mechanisms of cardiomyopathy. The Journal of Cell Biology. 2011;194:355-365
  49. 49. Navarrete EG, Liang P, Lan F, Sanchez-Freire V, Simmons C, Gong T, et al. Screening drug-induced arrhythmia [corrected] using human induced pluripotent stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circulation. 2013;128:S3-S13
  50. 50. Matsa E, Burridge PW, Wu JC. Human stem cells for modeling heart disease and for drug discovery. Science Translational Medicine. 2014;6:239ps6
  51. 51. Mordwinkin NM, Lee AS, Wu JC. Patient-specific stem cells and cardiovascular drug discovery. Journal of the American Medical Association. 2013;310:2039-2040
  52. 52. Noseda M, Peterkin T, Simoes FC, Patient R, Schneider MD. Cardiopoietic factors: Extracellular signals for cardiac lineage commitment. Circulation Research. 2011;108:129-152
  53. 53. Spater D, Hansson EM, Zangi L, Chien KR. How to make a cardiomyocyte. Development. 2014;141:4418-4431
  54. 54. Smith AS, Macadangdang J, Leung W, Laflamme MA, Kim DH. Human iPSC-derived cardiomyocytes and tissue engineering strategies for disease modeling and drug screening. Biotechnology Advances. 2017;35:77-94
  55. 55. Burridge PW, Keller G, Gold JD, Wu JC. Production of de novo cardiomyocytes: Human pluripotent stem cell differentiation and direct reprogramming. Cell Stem Cell. 2012;10:16-28
  56. 56. Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M, et al. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature. 2008;453:524-528
  57. 57. Kattman SJ, Witty AD, Gagliardi M, Dubois NC, Niapour M, Hotta A, et al. Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines. Cell Stem Cell. 2011;8:228-240
  58. 58. Willems E, Spiering S, Davidovics H, Lanier M, Xia Z, Dawson M, et al. Small-molecule inhibitors of the Wnt pathway potently promote cardiomyocytes from human embryonic stem cell-derived mesoderm. Circulation Research. 2011;109(4):360
  59. 59. Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nature Biotechnology. 2007;25:1015-1024
  60. 60. Zhang Q, Jiang J, Han P, Yuan Q, Zhang J, Zhang X, et al. Direct differentiation of atrial and ventricular myocytes from human embryonic stem cells by alternating retinoid signals. Cell Research. 2011;21:579-587
  61. 61. Uosaki H, Fukushima H, Takeuchi A, Matsuoka S, Nakatsuji N, Yamanaka S, et al. Efficient and scalable purification of cardiomyocytes from human embryonic and induced pluripotent stem cells by VCAM1 surface expression. PLoS One. 2011;6:e23657
  62. 62. Elliott DA, Braam SR, Koutsis K, Ng ES, Jenny R, Lagerqvist EL, et al. NKX2-5(eGFP/w) hESCs for isolation of human cardiac progenitors and cardiomyocytes. Nature Methods. 2011;8:1037-1040
  63. 63. Hudson J, Titmarsh D, Hidalgo A, Wolvetang E, Cooper-White J. Primitive cardiac cells from human embryonic stem cells. Stem Cells and Development. 2012;21:1513-1523
  64. 64. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proceedings of the National Academy of Sciences of the United States of America. 2012;109:E1848-E1857
  65. 65. Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM, Ebert AD, et al. Chemically defined generation of human cardiomyocytes. Nature Methods. 2014;11:855-860
  66. 66. Lian X, Bao X, Zilberter M, Westman M, Fisahn A, Hsiao C, et al. Chemically defined, albumin-free human cardiomyocyte generation. Nature Methods. 2015;12:595-596
  67. 67. Li S, Chen G, Li RA. Calcium signalling of human pluripotent stem cell-derived cardiomyocytes. The Journal of Physiology. 2013;591:5279-5290
  68. 68. Yang X, Rodriguez M, Pabon L, Fischer KA, Reinecke H, Regnier M, et al. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. Journal of Molecular and Cellular Cardiology. 2014;72:296-304
  69. 69. Lundy SD, Zhu WZ, Regnier M, Laflamme MA. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells and Development. 2013;22:1991-2002
  70. 70. Nunes SS, Miklas JW, Liu J, Aschar-Sobbi R, Xiao Y, Zhang B, et al. Biowire: A platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nature Methods. 2013;10:781-787
  71. 71. Yang X, Pabon L, Murry CE. Engineering adolescence: Maturation of human pluripotent stem cell-derived cardiomyocytes. Circulation Research. 2014;114:511-523
  72. 72. Foldes G, Mioulane M, Wright JS, Liu AQ, Novak P, Merkely B, et al. Modulation of human embryonic stem cell-derived cardiomyocyte growth: A testbed for studying human cardiac hypertrophy? Journal of Molecular and Cellular Cardiology. 2011;50:367-376
  73. 73. Moretti A, Bellin M, Welling A, Jung CB, Lam JT, Bott-Flugel L, et al. Patient-specific induced pluripotent stem-cell models for long-QT syndrome. The New England Journal of Medicine. 2010;363:1397-1409
  74. 74. Fatima A, Xu G, Shao K, Papadopoulos S, Lehmann M, Arnaiz-Cot JJ, et al. In vitro modeling of ryanodine receptor 2 dysfunction using human induced pluripotent stem cells. Cellular Physiology and Biochemistry. 2011;28:579-592
  75. 75. Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK, Wang L, et al. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell. 2013;12:101-113
  76. 76. Kurman RJ. Origin and molecular pathogenesis of ovarian high-grade serous carcinoma. Annals of Oncology. 2013;24(Suppl 10):x16-x21
  77. 77. Jones PM, Drapkin R. Modeling high-grade serous carcinoma: How converging insights into pathogenesis and genetics are driving better experimental platforms. Frontiers in Oncology. 2013;3:217
  78. 78. Finkbeiner SR, Freeman JJ, Wieck MM, El-Nachef W, Altheim CH, Tsai YH, et al. Generation of tissue-engineered small intestine using embryonic stem cell-derived human intestinal organoids. Biology Open. 2015;4:1462-1472
  79. 79. 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;470:105-109
  80. 80. Takahashi Y, Sato S, Kurashima Y, Yamamoto T, Kurokawa S, Yuki Y, et al. A refined culture system for human induced pluripotent stem cell-derived intestinal epithelial Organoids. Stem Cell Reports. 2018;10:314-328
  81. 81. McCracken KW, Cata EM, Crawford CM, Sinagoga KL, Schumacher M, Rockich BE, et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature. 2014;516:400-404
  82. 82. Munera JO, Sundaram N, Rankin SA, Hill D, Watson C, Mahe M, et al. Differentiation of human pluripotent stem cells into colonic Organoids via transient activation of BMP signaling. Cell Stem Cell. 2017;21:51-64e6
  83. 83. Crespo M, Vilar E, Tsai SY, Chang K, Amin S, Srinivasan T, et al. Colonic organoids derived from human induced pluripotent stem cells for modeling colorectal cancer and drug testing. Nature Medicine. 2017;23:878-884
  84. 84. Kadzik RS, Morrisey EE. Directing lung endoderm differentiation in pluripotent stem cells. Cell Stem Cell. 2012;10:355-361
  85. 85. Huang SX, Islam MN, O’Neill J, Hu Z, Yang YG, Chen YW, et al. Efficient generation of lung and airway epithelial cells from human pluripotent stem cells. Nature Biotechnology. 2014;32:84-91
  86. 86. Firth AL, Dargitz CT, Qualls SJ, Menon T, Wright R, Singer O, et al. Generation of multiciliated cells in functional airway epithelia from human induced pluripotent stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E1723-E1730
  87. 87. 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;123:4950-4962
  88. 88. Longmire TA, Ikonomou L, Hawkins F, Christodoulou C, Cao Y, Jean JC, et al. Efficient derivation of purified lung and thyroid progenitors from embryonic stem cells. Cell Stem Cell. 2012;10:398-411
  89. 89. Chen YW, Huang SX, de Carvalho A, Ho SH, Islam MN, Volpi S, et al. A three-dimensional model of human lung development and disease from pluripotent stem cells. Nature Cell Biology 2017;19:542-549
  90. 90. Gilpin SE, Ren X, Okamoto T, Guyette JP, Mou H, Rajagopal J, et al. Enhanced lung epithelial specification of human induced pluripotent stem cells on decellularized lung matrix. The Annals of Thoracic Surgery. 2014;98:1721-1729; discussion 9
  91. 91. Ghaedi M, Mendez JJ, Bove PF, Sivarapatna A, Raredon MS, Niklason LE. Alveolar epithelial differentiation of human induced pluripotent stem cells in a rotating bioreactor. Biomaterials. 2014;35:699-710
  92. 92. Curry EL, Moad M, Robson CN, Heer R. Using induced pluripotent stem cells as a tool for modelling carcinogenesis. World Journal of Stem Cells. 2015;7:461-469
  93. 93. Drost J, Karthaus WR, Gao D, Driehuis E, Sawyers CL, Chen Y, et al. Organoid culture systems for prostate epithelial and cancer tissue. Nature Protocols. 2016;11:347-358
  94. 94. Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159:176-187
  95. 95. Lee DF, Su J, Kim HS, Chang B, Papatsenko D, Zhao R, et al. Modeling familial cancer with induced pluripotent stem cells. Cell. 2015;161:240-254
  96. 96. Simsek S, Zhou T, Robinson CL, Tsai SY, Crespo M, Amin S, et al. Modeling cystic fibrosis using pluripotent stem cell-derived human pancreatic ductal epithelial cells. Stem Cells Translational Medicine. 2016;5:572-579
  97. 97. Zhang N, Bailus BJ, Ring KL, Ellerby LM. iPSC-based drug screening for Huntington’s disease. Brain Research. 1638;2016:42-56
  98. 98. Suh W. A new era of disease modeling and drug discovery using induced pluripotent stem cells. Archives of Pharmacal Research. 2017;40:1-12
  99. 99. Ghaemmaghami AM, Hancock MJ, Harrington H, Kaji H, Khademhosseini A. Biomimetic tissues on a chip for drug discovery. Drug Discovery Today. 2012;17:173-181
  100. 100. Kimura H, Sakai Y, Fujii T. Organ/body-on-a-chip based on microfluidic technology for drug discovery. Drug Metabolism and Pharmacokinetics. 2018;33:43-48
  101. 101. Williamson A, Singh S, Fernekorn U, Schober A. The future of the patient-specific body-on-a-chip. Lab on a Chip. 2013;13:3471-3480
  102. 102. Kotterman MA, Schaffer DV. Engineering adeno-associated viruses for clinical gene therapy. Nature Reviews. Genetics. 2014;15:445-451
  103. 103. Aiuti A, Biasco L, Scaramuzza S, Ferrua F, Cicalese MP, Baricordi C, et al. Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science. 2013;341:1233151
  104. 104. Howe SJ, Mansour MR, Schwarzwaelder K, Bartholomae C, Hubank M, Kempski H,et al. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. The Journal of Clinical Investigation. 2008;118:3143-3150
  105. 105. Melnikova I. Rare diseases and orphan drugs. Nature Reviews. Drug Discovery. 2012;11:267-268
  106. 106. Chuang K, Fields MA, Del Priore LV. Potential of gene editing and induced pluripotent stem cells (iPSCs) in treatment of retinal diseases. The Yale Journal of Biology and Medicine. 2017;90:635-642
  107. 107. Christensen CL, Choy FYM. A prospective treatment option for lysosomal storage diseases: CRISPR/Cas9 gene editing technology for mutation correction in induced pluripotent stem cells. Diseases. 2017;5
  108. 108. Adkar SS, Brunger JM, Willard VP, Wu CL, Gersbach CA, Guilak F. Genome engineering for personalized arthritis therapeutics. Trends in Molecular Medicine. 2017;23:917-931
  109. 109. Russo FB, Cugola FR, Fernandes IR, Pignatari GC, Beltrao-Braga PC. Induced pluripotent stem cells for modeling neurological disorders. World Journal of Transplantation. 2015;5:209-221
  110. 110. Okano H, Yamanaka S. iPS cell technologies: Significance and applications to CNS regeneration and disease. Molecular Brain. 2014;7:22
  111. 111. Tg, Hdl working Group of the Exome Sequencing Project NHL, Blood I, Crosby J, Peloso GM, Auer PL, et al. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. The New England Journal of Medicine. 2014;371:22-31
  112. 112. Wang G, Yang L, Grishin D, Rios X, Ye LY, Hu Y, et al. Efficient, footprint-free human iPSC genome editing by consolidation of Cas9/CRISPR and piggyBac technologies. Nature Protocols. 2017;12:88-103
  113. 113. Shinkuma S, Guo Z, Christiano AM. Site-specific genome editing for correction of induced pluripotent stem cells derived from dominant dystrophic epidermolysis bullosa. Proceedings of the National Academy of Sciences of the United States of America. 2016;113:5676-5681
  114. 114. Martin U. Therapeutic application of pluripotent stem cells: Challenges and risks. Front Med (Lausanne). 2017;4:229
  115. 115. Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, et al. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324:797-801
  116. 116. Yusa K, Rashid ST, Strick-Marchand H, Varela I, Liu PQ, Paschon DE, et al. Targeted gene correction of alpha1-antitrypsin deficiency in induced pluripotent stem cells. Nature. 2011;478:391-394
  117. 117. Eid A, Mahfouz MM. Genome editing: The road of CRISPR/Cas9 from bench to clinic. Experimental and Molecular Medicine. 2016;48:e265
  118. 118. Frock RL, Hu J, Meyers RM, Ho YJ, Kii E, Alt FW. Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nature Biotechnology. 2015;33:179-186
  119. 119. Weber J, Ollinger R, Friedrich M, Ehmer U, Barenboim M, Steiger K, et al. CRISPR/Cas9 somatic multiplex-mutagenesis for high-throughput functional cancer genomics in mice. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:13982-13987
  120. 120. Yue F, Hirashima K, Tomotsune D, Takizawa-Shirasawa S, Yokoyama T, Sasaki K. Reprogramming of retinoblastoma cancer cells into cancer stem cells. Biochemical and Biophysical Research Communications. 2017;482:549-555

Written By

Ying Qu, Nur Yucer, Veronica J. Garcia, Armando E. Giuliano and Xiaojiang Cui

Submitted: 27 March 2018 Reviewed: 04 April 2018 Published: 06 June 2018