Abstract
Induced pluripotent stem cell (iPSC) technology and advancements in three-dimensional (3D) bioprinting technology enable scientists to reprogram somatic cells to iPSCs and 3D print iPSC-derived organ constructs with native tissue architecture and function. iPSCs and iPSC-derived cells suspended in hydrogels (bioinks) allow to print tissues and organs for downstream medical applications. The bioprinted human tissues and organs are extremely valuable in regenerative medicine as bioprinting of autologous iPSC-derived organs eliminates the risk of immune rejection with organ transplants. Disease modeling and drug screening in bioprinted human tissues will give more precise information on disease mechanisms, drug efficacy, and drug toxicity than experimenting on animal models. Bioprinted iPSC-derived cancer tissues will aid in the study of early cancer development and precision oncology to discover patient-specific drugs. In this review, we present a brief summary of the combined use of two powerful technologies, iPSC technology, and 3D bioprinting in health-care applications.
Keywords: Induced pluripotent stem cells, Three-dimensional bioprinting, Regenerative medicine, Disease modeling, Cancer iPSCs, Drug screening
1 Introduction
The advent of induced pluripotent stem cell (iPSC) technology in 2006 paved the way for paradigm shifting changes in regenerative medicine, disease modeling, and drug discovery applications. The technology facilitates to de-differentiate an adult cell to its pluripotent stem cell state and then differentiate into defined cell lineages. iPSCs are phenotypically indistinguishable from embryonic stem cells and they can differentiate into specialized cells of the body in cell culture and in animal models. Initially, human iPSCs were derived using transduction of genes coding for four embryonic transcriptional regulators; Oct4, Sox2, Klf4, and c-Myc (OSKM), popularly known as the Yamanaka factors[1] or Oct4, Sox2, Lin28, and Nanog (OSLN)[2]. Each of the Yamanaka factor serve specific purposes, Sox2 interacts with Oct3/4 to control gene expression. This interaction is important in maintaining pluripotency[3]. C-Myc plays an important role in controlling growth and differentiation of cells[4], whereas klf4 is crucial for cell division and maintenance of pluripotency[5]. Later, different combinations of at least 24 embryonic transcription factors were identified to induce stemness in adult cells[6]. The Yamanaka factors are highly conserved and sufficient to induce pluripotency across species.
Reprogramming of somatic cells is orchestrated by cooperative binding of pioneer factors (Oct4, Sox2, and Klf4)[7], followed by epigenetic remodeling of entire genome and two waves of transcriptional events[8,9]. Each cell type in the body require different combinations of transcriptional factors to induce the stemness where Oct4 is considered as an indispensable core pluripotency gene in the reprogramming process[10]. Exogenous supply of Oct4 alone could convert adult neural stem cells into iPSCs. Recent work by An et al. showed that Sox2 and Klf4 were enough to prepare iPSCs from various types of somatic cells[7]. Small molecules that inhibit DNA or histone modifications were also used for generating iPSCs more efficiently along with the use of reprogramming transcription factors. The hematopoietic stem cells can be de-differentiated into iPSCs much more efficiently compared to the highly specialized cells such as B and T lymphocytes[11,12].
Fibroblasts are the most popular cell type used to generate iPSCs. However, well-differentiated adult cells such as keratinocytes, neural cells, fat cells, melanocytes, amniotic fluid cells, pancreatic beta cells, and peripheral blood derived cells had also been successfully reprogrammed to pluripotent stem cells. The capacity to induce pluripotency to somatic cells helps to generate pluripotent patient-specific cell lines that can help model human diseases and can aid in the reconstruction of damaged tissues and organs. The “disease in a dish” models derived from IPSCs provide insights into disease pathogenesis and can serve as a novel tool for drug evaluation in precision medicine field[13-15]. Human iPSCs reinforced with biocompatible scaffold materials are valuable in three-dimensional (3D) bioprinting applications[16]. Current bioprinting techniques allow to print undifferentiated iPSCs and iPSC-derived cells mixed with a suitable bioink[17,18]. Popular bioprinting techniques used to print iPSCs are extrusion, stereolithography (SLA), laser-assisted, and drop-on-demand bioprinting[19-22]. A single biomaterial or a mixture of several biomaterials in the bioink are used to suspend the desired cells for bioprinting[23,24]. The bioinks should be non-toxic, biocompatible and should provide structural support for the printed cells. The commonly used bioinks for printing iPSC derived cells are hydrogels derived from alginate, carboxymethyl chitosan, agarose, nano-fibrillated cellulose, hydroxypropyl chitin, gelatin methacryloyl (GelMA), and Matrigel. Most of these hydrogels need a crosslinker to give the final structure of the intended tissue constructs. Calcium chloride, ultraviolet (UV) radiation, photo crosslinking, and altered temperatures are used for crosslinking the bioink molecules[25-27].
Here, we review the applications of the 3D bioprinted iPSCs or iPSC-derived cellular products in healthcare, especially in regenerative medicine, disease modeling, and drug testing (Figure 1). The methods of reprograming of iPSCs were described. Glimpses of the technological advancement in organ bioprinting were discussed. The advantages, limitations, and future directions of using iPSCs in clinics were outlined.
Figure 1.
Bioprinting of induced pluripotent stem cell (Ipsc)-derived tissues for regenerative medicine, disease modeling, and drug testing. Adult somatic cells are collected from a donor or a patient; the cells are reprogrammed to iPSCs and differentiated to specialized cells.
2 Human iPSC reprogramming methods
For clinical application and disease modeling, the reprogramming method of choice should have adequate efficiency to produce iPSCs from less abundant samples[28]. Production of iPSCs using a combination of reprogramming methods can augment the efficiency of iPSCs generation even from the most difficult type of adult cells[29]. More than 10 years of extensive research on iPSC technology lead to the establishment of novel strategies for the production of iPSCs including the use of right cell type for reprogramming, use of non-integrative gene introduction methods, overexpression of gene enhancers of transcription factors, and the use of small molecules[30,31] (Figure 2).
Figure 2.
Reprogramming methods used to generate induced pluripotent stem cell.
2.1 Integrating viral vectors
Initial iPSC experiments used lentivirus and retrovirus vectors to deliver Yamanaka factors in adult fibroblasts[2,32]. These retroviral-vectors possess the risk of creating mutagenesis by integrating to the host cell genetic material[33]. Moreover, the reprogramming procedure is tedious, also, it can cause chromosomal instability and potential threat of tumorigenesis from the viral vectors[1,2,31-34]. It is essential to do the quality control of the iPSC lines created using viral vectors using whole-genome single nucleotide polymorphism array and karyotyping to verify the genomic integrity.
2.2 Non-integrating vectors
To reduce the risk of unwanted genetic perturbations, there was introduction of non-integrating methods for transcription factor delivery. Recent methods of reprogramming use Sendai Virus (SeV) particles and episomes. SeV particles are used to transduce the reprogramming genes: Oct4, Sox2, Klf4, and c-Myc[8]. SeV reprogramming offers the absence of viral sequences in the host cell lines and is an efficient method to induce pluripotency[35]. In 2009, Yu et al. reported the episomal reprogramming in human cells where Epstein-Barr virus–derived sequences were used for the non-integrated expression of the transcription genes that enable episomal plasmid DNA replication in dividing cells[36]. Polycistronic expression plasmids generates transgene-free and vector-free iPSCs with limited genomic integration, but this method requires multiple transfections[37,38].
2.3 mRNAs and miRNAs
Human primary fibroblasts were reprogramed by introducing synthetic modified mRNAs coding for reprogramming proteins. MicroRNAs like miRNA-367/302s are used to reprogram human primary fibroblasts into iPSCs[39]. The mRNA transfection is foot print free reprogramming. The capped mRNAs coding for a 6-factor modified-mRNA referred to as 5fM3O mod-mRNAs was used in fibroblasts to make iPSCs[40]. Yamanaka factors and the miRNA-367/302s act synergistically to increase the efficiency of transfection[41]. It has been reported that microRNAs such as miR-294, miR-291-3, and miR-295 can replace c- myc transcription factor and help to generate homogeneous populations of iPSC colonies[42]. Downregulation of let-7 miRNA upregulates the expression of target genes of c-myc and Lin-28 to promote cell reprogramming[43,44].
2.4 Reprogramming proteins
This method allows the direct introduction of the recombinantly expressed reprogramming factors to cells[49]. This method mitigates the risks associated with the introduction of viral and external DNA and harmful chemicals into the cells[46]. The reprogramming proteins Oct4, Sox2, Klf4, and c-Myc were successfully delivered into adult somatic cells with the help of cell penetrating peptides (CPP). The cationic amino acid rich CPPs are capable of penetrating the cell membrane barrier and deliver the exogene-free reprogramming proteins directly inside the cells[47]. This method enables the production of foot print-free iPSCs.
2.5 Small molecules
Reprogramming can be achieved using small molecules by epigenetic modifications[48]. Small molecules used for reprogramming fall under the category of epigenetic events regulators, mesenchymal-epithelial transition inhibitors, metabolic pathway modulators, wingless and integration site growth factor (WNT) signal pathway modulators, regulators of cell death, and senescence pathways[48]. These small molecules alone or in combination can substitute exogenous transcription factors. Using valproic acid, a Histone deacetylase inhibitor improved the reprogramming efficiency to 100-fold compared to the transcription factor mediated reprogramming method[49]. Another histone methyltransferase inhibitor compound, BIX-01294 activated calcium channels in the cell membrane, and improve reprogramming efficiency by increasing the expression of October 4 and Klf4[50,51]. In 2013, Hou et al. replaced all transcription factors and made chemical induced iPSCs of mouse embryo fibroblasts using the small molecules VC6TFZ and 2i but the reprogramming was incomplete[52]. Optimizing the use of small molecules to enhance reprogramming will definitely help to generate safer and higher quality iPSCs for clinical use without the risk of genomic integration and tumor induction.
The advantages and limitations of these reprogramming methods are summarized in Table 1.
Table 1.
Advantages and limitations of the reprogramming methods.
Delivery method | Advantages | Limitations | References |
---|---|---|---|
Integrated viral vectors | High efficiency, validated in many cell types | Create insertional mutations, tumor induction | [1,2,31-34] |
Integration free viral vectors. For example, Sendai virus, Episomes | Completely free of vector and transgene sequences and can use in clinical applications, GMP compatible | Low efficiency of reprogramming, reprogramming efficiency varies with cell types | [8,35-38] |
mRNAs and miRNAs | Faster, high efficiency, absence of integration, need low input of starting cells | Multiple transfection required, may elicit immune response, not evaluated in many cell types | [40-44] |
Reprogramming Proteins | Foot print-free, cGMP compliant | Not evaluated in many cell types, expensive | [45-47] |
Small molecules | Foot print-free, cGMP compliant, economical | Incomplete reprogramming, reprogramming efficiency varies from cell to cell | [48-52] |
3 Sources of iPSCs for bioprinting and cell differentiation strategies of iPSCs to different cell lineages
The iPSC technology allows the use of autologous cells derived from the patients to be used in regenerative medicine. The iPSC cell lines have been derived from a variety of cells namely neuronal progenitor cells, keratinocytes, hepatocytes, B cells, fibroblasts, hepatocytes, gastric epithelial cells, muscles, adipocytes, and adrenal glandular cells. The reprograming efficiency varies among the type of cells used depending on the developmental origins and the epigenetic status. Multiple studies showed that the efficiency of reprogramming of keratinocytes is better than fibroblasts[53-55]. The difference in the efficiency is attributed to the high-level endogenous expression of c-Myc and Klf4 in keratinocytes compared to other cell types. For therapeutic scenarios such as internal organ reconstruction (e.g., liver, and peripheral nerve), an ideal starting material to create iPSCs should be peripheral tissue. Peripheral tissue samples such as keratinocytes collected from patients can be reprogrammed to produce iPSCs and these iPSCs can serve as a valuable source for the cellular component in the tissue bioprinting[56]. Peripheral blood cells may represent a good source to derive iPSCs because taking blood samples from patients are a less invasive clinical procedure. Several research groups standardized the protocols for making iPSC lines from blood cells[57,58]. The iPSC-derived cells such as neurons, hepatocytes, osteoblasts, myocytes, skin cells, and pancreatic beta cells are in great demand in clinics for regenerative medicine applications[59-61]. Small molecules and growth factors such as recombinant proteins are used in the iPSC cultures to differentiate specific lineage cells from iPSCs[62,63].
4 Application of bioprinted iPSCs in healthcare
3D bioprinting using iPSCs hold high potential for several applications in the health-care sector. Current bioprinting techniques allow to print undifferentiated iPSCs and differentiated iPSC-derived cells embedded in a suitable bioink. Development of novel bioinks, iPSC-derived cells, and the technological advancement in devising new generation 3D bioprinters has created a whole new field of medical bioprinting that hold great promise for artificial tissue/organ printing for regenerative medicine, disease modeling, and drug testing[64]. Here, in section 4, we describe about the commonly used bioprinting techniques to print iPSCs and the application of bioprinting in health-care field.
4.1 Commonly used iPSC 3D bioprinting technologies
Bioprinting iPSCs could avoid the ethical and immunological bottle necks of organ printing. iPSCs and iPSC derived cells for developing into cartilage, bone, skin, heart, liver, and neural tissues have been successfully printed using 3D bioprinting technology. iPSC-derived chondrocytes along with irradiated chondrocytes were bioprinted to cartilage tissues using mixture of alginate and nanocellulose bioink. RegenHu 3D discovery bioprinter was used to print the chondrocytes. Extrusion bioprinting is the most commonly used method to print iPSCs and organs, followed by SLA, laser-assisted, drop-on demand, inkject, and microvalve based methods. The extrusion method applies a pneumatic (air) or mechanical (screw or piston) force to extrude iPSCs or its derivatives embedded in hydrogel bioinks through a nozzle orifice using seamless direct printing[65]. The printing is carried out in a spatially controlled layer-by-layer fashion precisely to biofabricate 3D tissue constructs. Extrusion printing permits printing of cell-dense high viscous hydrogels, but cells may experience high shear force stress during the printing process. All three germ layers[66], neural tissues[67], cortical neural constructs[68], chondrocytes[69], cardiac tissue,[70] and peripheral blood mononuclear cells have been bioprinted from iPSC or iPSC derived cells using extrusion method.
SLA and digital light projection (DLP) are popular nozzle-free bioprinting techniques work through photopolymerization. The liquid resin is solidified by UV laser beam in SLA. The DLP uses visible light for polymerization of the resin[71,72]. There are two types of SLA and DLP, i.e., bottom-up and top-down approach. The layer of resin on a support platform is cured by a light from above in bottom-up biofabrication, while the light source is located under a transparent platform in the top-down fabrication. iPSC-derived 3D liver models which mimic the native liver module architecture were printed using this technique[73]. The flipside of SLA is that, it can be detrimental to the living cells by damaging the genetic materials due to the use of UV. Moreover, the use of photo-initiator resins may be cytotoxic to the cells[74]. Vat polymerization (VP) based bioprinting is a novel and accurate bioprinting method tissue engineering applications[75]. Various types of photo-initiators are used for the cross-linking of the printed tissues in the VP based bioprinting. VP use light sensitive hydrogels such as polyethylene glycol–diacrylate (PEGDA) and gelatin-methylacryloyl (GelMA). Label-free diamagnetophoretic printing is another method for microtissue printing uses intrinsic diamagnetic forces to control positioning of cells in a paramagnetic medium. Magnetic bioprinting is a contactless technique which does not use nozzles and therefore promise less contamination of cell suspension. Whole blood cells were printed using this technique[76], this technique may be efficient to adopt to print iPSCs as there is fewer chemical manipulations that are involved.
Droplet-based bioprinting is simple, fast, and precisely controlled bioprinting method to deposit composites of cells, growth factors, biomolecules, drugs, and scaffolds. Droplet-based bioprinting is derived from inkjet printing technology. It has been noted as a prominent technique widely used in regenerative medicine to print cells due to its flexibility. The droplet-based bioprinting can be subdivided into inkjet, acoustic, and micro-valve bioprinting modalities. The drawback of using this technology is that the rage of biomaterials compatible for this method of bioprinting is limited[77]. A valve-based bioprinting method is used to print iPSCs differentiated post-printing into hepatocyte-like cells (HLC cells). A 40-layer thick alginate bioink containing HLC cells showed typical liver tissue structure and the construct secreted hepatic albumin throughout the differentiation protocol. The work proved that the valve-based printing process is safe to print human iPSCs by maintaining pluripotency and differentiation[19].
The laser-assisted bioprinting uses pulsed laser beam with a focusing device. It consists of an energy-absorbing layer coated with further layers of cell-encapsulated hydrogel. It is a nozzle-free bioprinting method that excludes clogging during printing[78]. Human iPSCs combined with bioinks were bioprinted with laser-assisted bioprinting method and the cells were evaluated for their efficacy, pluripotency, and differentiation capacity. The hyaluronic acid-based bioinks are ideal for laser-assisted bioprinting[16,79]. While designing a tissue structure for bioprinting, factors such as shape, size, resolution, scaffold materials, iPSCs, or iPSC derived cellular components, and post-processing tissue dynamics are to be considered. Bioprinting can be carried out as direct printing, crosslinking during the bioprinting, post-printing crosslinking, and hybrid methods, where more than one technique is used for printing the final tissue construct. The factors appear to affect cell survival after bioprinting are shear stress, laser exposure, duration, temperature, humidity, mechanical pressure, and vibration of the printing process. The selection of the printing techniques depends on the end use of the printed tissues, such as organ/tissue transplantation, disease modeling, or drug evaluation. Among, all the methods mentioned, extrusion bioprinting is the most commonly used technique in iPSC bioprinting[80,81] (Table 2). Extrusion method causes less damage to the cellular components while printing, as it uses adjustable mechanical forces with no harsh treatments for the deposition of the bioink to the platform.
Table 2.
Summary of iPSC-based Bioprinting works
Printing technique | Cell source | Cells/tissues printed | Bioink used | Reference |
---|---|---|---|---|
Extrusion | iPSCs, BJFF iPSCs | Cardiac | Collagen I, Matrigel, Gelatin | [70] |
Human iPSCs | Chondrocytes | Nano-fibrillated cellulose in alginate | [69] | |
Fibroblasts derived human iPSCs | Germ layers | Geltrex | [66] | |
Human iPSCs ((WT I line) | Neural construct | Matrigel/alginate mixture | [68] | |
Human iPSCs | Neural tissues | Alginate, carboxymethyl-chitosan, agarose | [67] | |
Human peripheral blood mononuclear cells derived iPSCs | Pluripotent cells | Hydroxypropyl chitin, Matrigel | [79] | |
Stereolithography | Human iPSCs | Hepatic progenitor cells | Gelatin methacrylate (GelMA), Glycidal methacrylate-hyaluronic acid (GMHA) | [73] |
Laser-assisted | Human iPSCs from cord blood | Germ layers | Matrigel, Collagen type I, Alginate, Hyaluronic acid | [16] |
Microvalve-based | Human iPSCs | Hepatocyte-like cells (HLCs) | Geltrex | [19] |
4.2 Regenerative medicine
Autologous iPSCs derived from individuals provide unlimited source of cells for tissue regeneration. The unspecialized iPSCs can differentiate and develop to organoids/spheroids with specific characteristics of organs in vivo[74,82-84]. These mini-organoids can serve as building blocks for bioprinting of whole organs. Bioengineers and surgeons are looking for novel methods to synthesize artificial skin substitutes that is readily available and easily implantable in burn injury patients[85,86]. Scaffold-free cellular spheroids obtained from a coculture of human iPSC-derived cardiomyocytes, fibroblasts, and endothelial cells were 3D printed and these cardiac cellular patches were tested successfully in rat models of myocardial infarction[87]. Bioprinted organ substitutes such as pancreas, ovary, liver, kidney, and nervous tissues also will be in high demand in the near future. Figure 3 shows the workflow of 3D bioprinting of peripheral nerve tissue[88,89] for the treatment of peripheral nerve injury.
Figure 3.
Workflow of three-dimensional bioprinting of peripheral nerve tissue for treatment of peripheral nerve injury. Sciatica is the pain due to the injury to the largest nerve in the body. Extreme case of sciatica needs surgical intervention and if possible, regenerative therapy. The autologous peripheral tissues can be collected from the patient, reprogram to induced pluripotent stem cells and differentiate to peripheral nerve tissue progenitors or peripheral neurons, then bioprint using a suitable scaffold material, allow the maturation of the bioprinted tissue in a bioreactor and transplant to the patient to repair and regenerate the injured nervous tissue.
Human iPSCs are capable of differentiation into many types of specialized cells and have high value in clinical use. These cells require specific cell culture media to keep their pluripotent characteristics intact. The isolation, expansion, and maintenance of human iPSCs intended for clinical use should be cultured in xeno-free conditions in compliance with the good manufacturing practice to avoid hypersensitivity reactions after transplantation in patients[90,91]. However, many conventional protocols of iPSC culture require to culture in feeder cells. The feeder cells are usually derived from mouse embryonic fibroblasts (MEFs). The cells are cultured on feeder cells to reduce the genetic instability of the cultured cells[89]. Culturing in MEF feeder cells or the usage of matrix coating substance (e.g., gelatin or Matrigel) made of animal components make the iPSCs xeno-positive. Recent introduction of synthetic polymers enables to maintain the iPSC cultures in xeno-free environment[92].
Yamanaka factor introduction techniques use different type of retroviral or plasmid vectors to integrate to the genome of the cell to make it pluripotent. For making clinical grade iPSCs and its products, the cells should be free from any genetic integration of foreign DNA materials in the iPSC genome. mRNA-based, episomal, and recombinant protein-based introduction of transcription factors eliminate the risk of genomic integrations or aberrations in the iPSCs, which is an important step toward using the iPSCs for cell-based therapies in patients[93-95].
4.2.1 Type 1 diabetes mellitus (T1DM) treatment by the replacement of pancreatic beta cells
T1DM is known to be associated with the immune-mediated destruction of insulin producing pancreatic β-cells[96,97]. Effective treatment of T1DM is a long pending requirement in diabetes care. Islet cell transplantation is a traditional method of managing T1DM, but the transplanted islets are rejected by the host immune system. Bioprinted islets encapsulated in a suitable biocompatible material have been emerged as a treatment method to tackle this immune rejection. Biofabrication of 3D constructs of patient-derived iPSCs differentiated to insulin-producing pancreatic islet cells can potentially be an allogeneic source of cells for T1DM treatment. In 2019, Kim et al. successfully developed a pancreatic tissue-derived (pdECM)-bioink to provide the pancreatic tissue-specific microenvironment to bioprint the human iPSC-derived pancreatic islet cells. The study used decellularized porcine pancreas to provide the extra cellular matrix support for the growth of the cells[98]. Biotechnology companies such as Cellheal and Celprogen are working toward to bioprint the functional pancreatic tissues for diabetic treatment[99].
4.2.2 Reproductive system disorders
Ovarian failure is a major cause of infertility worldwide[100,101]. Functional 3D printed ovarian tissues would be a boon for infertility treatment. In 2017, Laronda et al. reported that the 3D bioprinted ovary using porous gelatin scaffold material could support the growth and maturation of printed ovarian follicles in laboratory conditions. The lab-grown ovary is tested in a sterilized mouse model and found to be functional. The bioprosthetic ovary used scaffold materials such as gelatin and gelatin methacrylate for bioprinting[102]. The structural features of the scaffold material such as pore size, pore geometry, and the surface contact area are the deciding factors for successful growth of functional ovarian tissue. Stem cells isolated from the patient’s own ovarian tissue can serve as the starting material to bioprint the functional ovary.
4.2.3 Thyroid gland replacement in thyrectomized patients
Bioprinting of a functional vascularized mouse thyroid gland construct from embryonic tissue spheroids was reported by Bulenova et al. in 2017[103]. Self-assembling thyroid spheres, thyrocytes, and endothelial cells suspended in collagen gel were used for bioprinting the thyroid gland. The bioprinted construct was implanted in a hypothyroid mouse and it could normalize the blood thyroxine levels and body temperature in the tested mice. Bioprinting of functional mouse thyroid gland tissue represents a major advance in bioprinting technology and organ regeneration research. In March 2019, NASA announced a plan for bioprinting thyroid gland in international space station to study the effect of microgravity on organs[104].
4.3 Bioprinted iPSCs in disease modeling
The most advantageous aspect of using induced pluripotent cells in clinics is the ability of reprogramming of autologous cells taken directly from patients. At present, the majority of disease modeling studies makes use of the traditional 2D cultures. Any monogenic or polygenic disease conditions can be re-created in a cell culture system[83]. While 2D cultures are good to understand the molecular level interactions, they possess several limitations including lack of heterogenic cell environment and the cell to cell communication cues[105]. The disease progression hugely depends on the extracellular matrix (ECM) mechanics and the cell to cell interactions[106]. Cellular phenotypes and the non-cell autonomous disease pathogenesis require mimicking the disease conditions in a more realistic 3D environment. 3D disease models would help in understanding the disease mechanism in detail in the early stages of the disease[107,108]. A few examples of disease modeling in cardiac, neurodegenerative and neurodevelopmental diseases and oncology are briefly discussed in the following sections.
4.3.1 Cardiac diseases
Cardiovascular diseases remain the leading cause of death in the developed world, accounting for more than 30% of all deaths. Collection of cardiac tissue from patients with disease causing mutations for genetic studies and functional analyses is a highly invasive procedure. iPSCs derived from the peripheral tissues of patients with disease specific mutations are a valuable tool to study the cardiac pathophysiology and drug development. Cardiac tissues were biofabricated using hydrogels and supporting cells such as cardiomyocytes, endothelial cells, smooth muscle cells, and fibroblasts[109,110]. The cells were cocultured and engineered to resemble their natural physiological microenvironment and recapitulate coordinated contractile and electrophysiological interactions with the ECM and heterogeneous cell types that make up the myocardial tissue environment[111]. The iPSC-derived cardiac cells were successfully used to model cardiac diseases such as dilated cardiomyopathy and myocardial infarction[112]. These disease models help identifying the cellular phenotypes critical to cardiac pathology[113,114]. The microfluidic organ-on-chip methods were also developed to evaluate the vascular perfusion in cardiac tissue. The tissue composition and architecture of the in vitro 3D microdevice can be precisely defined using microfabrication methods[115-117]. The iPSC and embryonic stem cell derived organ-on-chip systems are being used for modeling a wide range of diseases, including dilated cardiomyopathy, kidney glomerular injury, and wound healing[118,119].
4.3.2 Neurodegenerative and neurodevelopmental diseases
4.3.2.1 Alzheimer’s disease (AD)
AD is a progressive neurodegenerative disorder characterized by loss of cognition and disruption of basic functions, such as swallowing, walking, attention, and memory[120]. All major nerve cell types can be differentiated from iPSCs and can be cultured in complex conditions, which mimic the AD conditions. Precise Genome editing techniques can be used to introduce or correct AD-linked mutations to examine phenotypes in isogenic backgrounds cells[14]. It has become increasingly clear in recent years that multiple different brain cell types can contribute to AD progression[121]. Thus, examining their interactions and impacts on each other are of critical importance. The iPSCs can be differentiated into neural crest or neural progenitor cells, which can subsequently be patterned to different neuron subtypes including glutamatergic, GABAergic, cholinergic, and dopaminergic neurons[122-124]. 3D bioprinted AD models will facilitate the development of effective therapeutics to combat AD-induced dementia. Moreover, bioprinted AD tissue models can serve as a more humanized model system for AD drug testing, as many drugs tried in experimental animals failed in clinical trials due to species variability[125,126].
4.3.2.2 Parkinson’s disease (PD)
PD is the second most common neurodegenerative disorder[127]. Studies using iPSC-derived dopaminergic neurons from patients with monogenic and sporadic PD have successfully illustrated key features of PD pathophysiology, including impaired mitochondrial function, increased oxidative stress, and accumulation of α-synuclein protein, namely, Lewy bodies[128]. Using iPSC-derived dopaminergic neurons from PD patients with mutations in the disease causing genes, many investigators have drawn mechanistic insights on how mutations of these genes are linked to PD. Coculturing glial cells and neurons both derived from iPSCs of PD patients should therefore be another platform to advance insights into the multifactorial pathogenesis of PD. Bioprinted dopaminergic neurons can be treated with neurotoxins such as 6-hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, paraquat, and rotenone to induce PD like syndrome to study PD pathogenesis[129-131].
4.3.2.3 Amyloid lateral sclerosis (ALS)
ALS is the most prevalent motor neuron (MN) disease characterized by the progressive loss of the upper and lower MNs, leading to muscular atrophy, paralysis, and death within 5 years after the first diagnosis[130]. The iPSC-derived disease models of ALS showed increased oxidative stress and DNA damage in neurons. ALS specific mutations and altered transcriptome profile were also noticed in the iPSC models. Osborne et al. reported the effectiveness of the small molecule vardenafil, in regaining the resilience of MNs by regulating the insulin-like growth factor-II signaling in an iPSC-derived ALS model[132]. Researchers created 3D bioprinted ALS disease model using iPSC-derived MNs from a patient with TDP-43 gene mutation. TDP-43 gene mutation causes sporadic ALS. Bioprinted iPSC derived autologous tissue models of ALS disease serve as a valuable tool for studying the disease pathology as well as aid in the screening of personalized drugs against the disease[133].
4.3.3 Bioprinted iPSCs in oncology
The iPSCs derived from cancer tissues present a range of new opportunities for the study of human cancer. If human cancer cells were converted to pluripotency and then allowed to differentiate back into specific cancer tissue, they might shed light on the early stages of cancer[134].
Although 3D printing was developed decades ago, recent times witnessing a huge jump in adapting this versatile technology to the field of cancer modeling by fabricating sophisticated biological structures typical to cancer tissues. The cancer cells could be isolated from surgically removed cancer tissues. The isolated tumor cells are reprogrammed to pluripotent state by introducing the four transcription factors Oct4, Sox2, Klf4, and c-myc using a suitable method. The colonies are grown for a period of 2 – 4 weeks and cells with pluripotent stem cell morphology and molecular characters are expanded to create specific cancer iPSC lines[135]. These lines are differentiated into the tumor cell type of origin. The iPSC-derived differentiated cells could be then bioprinted into tumor tissues which mimic the tumor microenvironment (TME). The commonly used cancer bioprinting methods are: Inkject printing, extrusion-based printing, lase-assisted printing, and SLA. The tissues can be 3D bioprinted in the format of spheroids, organoids, coculture with other tumor TME cells, and organ-on chip. Improvements in the 3D bioprinting technology enable to distribute the cancer derived iPSCs in a 3D space with high precision and reproducibility. The 3D bioprinted tissues can be used as cancer tissue-on-chip models or transplanted into animal models to study different stages of cancer pathogenesis.
The most important hurdle in establishing iPSC-derived cancer models are the variation of intrinsic transcription factors in the cancer cells. This variation can affect the reprogramming efficiency of tumor cells. Many published studies showed that cancer cells are generally difficult to reprogram than normal cells[136]. The differentiation of cancer iPSCs to its initial tumor cell of origin also appears tedious and inconsistent. The success rate of establishing a cancer iPSC depends on the type cancer. So far, the successful reprogramming of myeloid tumors is established[137-139]. Establishing protocols for generating cancer iPSCs can help to model cancer progression, to understand the complex cancer genetics, and contribution of TME in cancer progression, anti-cancer drug development, and precision oncology (Figure 4).
Figure 4.
Bioprinted cancer tissue with induced pluripotent stem cell (iPSC)-derived cells: Establishing protocols for generating cancer iPSCs can help to model cancer progression, to understand the complex cancer genetics, and contribution of tumor microenvironment in cancer progression, anti-cancer drug development, and precision oncology.
4.4 Bioprinted iPSCs for drug and cosmetic testing
4.4.1 Bioprinted iPSCs for drug testing
More than 90% of drug molecules under different phase of clinical trials fail to reach market because of unanticipated toxicity to vital organs or lack of efficacy. This failure rate is partly attributed to the use of overly simplistic 2D cell culture-based assays[140]. The spectrum of activity of most of the drug molecules varied across the species, so animal testing has limited predictive value[107]. 3D bioprinting and iPSC technology enable printing of any tissue in its native architecture by preserving the complexity of cellular pathways, cell-cell interactions, and cellular-microenvironment interactions. The tissue microenvironment is critical for the understanding of drug acting mechanisms in humans[141]. The use of 3D bioprinted tissue is predicted to be an integral part of future drug discovery research for improved in vitro assays with better predictive value.
4.4.2 Cosmetics testing
The European Union Cosmetics Directive was introduced in 1976 to enforce high safety standards for cosmetics across the EU member states. In 2009, the EU commission has introduced new directives to phasing out animal testing for cosmetic products. The directive bans the testing of the cosmetic products and cosmetic ingredients on animals. The law even prohibits the marketing of finished cosmetic products and ingredients tested on animals in the European Union[142]. Hence, the cosmetic industry is looking for methods to replace the animal testing. Bioprinted artificial skin tissue can substitute the use of animals for cosmetics testing (Figure 5). At present, the human skin models required for cosmetic testing are manufactured by layering fibroblasts in a collagen hydrogel, then adding keratinocytes on top. The cells are allowed to differentiate and mature into the different epidermal layers. This type of artificial skin models is simple and would not exactly reflect the complexity of the native skin. Advanced bioprinting technology and iPSCs as cell source allow fabrication of more realistic artificial skin models for drug testing as well as for regenerative medicine[143]. This would help develop skin models with different skin types with respect to race (Asian, Caucasian, etc.), character (dry, oily, etc.), or other specific skin types depending on the intended use of the cosmetic as these models incorporate iPSC-derived cells. Collagen-based skin construct reinforced with biocompatible materials such as polycaprolactone (PCL) mesh prevented the contraction of collagen during tissue maturation and enable manufacturing of biomimetic human skin models[144,145]. Other biomaterials such as polylactic acid, Pluronic, alginate, chitosan, hyaluronic acid, fibrin, and gelatin, or a combination of PEGDA and GelMA were used as the scaffold material for printing skin models[145-147].
Figure 5.
Bioprinting of human skin tissue models for cosmetic testing to replace the animal testing procedures. Use of induced pluripotent stem cell-derived skin cells would help develop skin models with different skin types (Asian, Caucasian, etc.), character (dry, oily, etc.), or other specific skin types depending on the intended use of the cosmetic.
5 Bioinks used in iPSC bioprinting
Native tissue has complex architecture consisting of different cell types, ECM materials, growth factors, and many signaling molecules. ECM is organized in a highly delicate manner in a tissue to serve the tissue specific functions such as shape, consistency, mechanical strength, and molecular miscibility. Bioinks are cell laden hydrogels containing ECM components of the tissue to be printed. The materials used in the bioinks should be biocompatible without eliciting any undesirable response inside the body, should allow seamless printing, and should have tissue compatible rheological properties[56]. The bioink components can be natural, synthetic, different types of cells, and soluble growth factors specific for the cell types used. Natural polymers such as collagen, gelatin, fibronectin, laminin, and silk fibroin have been widely used in bioinks to augment cell attachment and migration in the matrix material[148] Polysaccharides such as alginate, agarose, and chitosan are also widely using in bioinks. Alginate is obtained from a type of brown algae and is widely used in 3D bioprinting applications due to its biocompatibility, promotion of cell proliferation, low price, and the ability of crosslinking in calcium ion solutions. However, alginate lacks sufficient mechanical stiffness for 3D bioprinting. Agarose is another biocompatible polysaccharide but it liquifies above physiological temperatures. Chitin and chitosan obtained from crustaceous animal are widely used polysaccharide component of bioinks, but it is slow in solidification. The methacrylated form of gelatin (GelMA) is a popular bioink component to print iPSCs which possess easily tunable physiochemical properties to use in bioprinters[149].
Still, many of these polysaccharides are too fragile and lack sufficient mechanical strength to retain in the transplant tissue site and often suffer from low mechanical properties, and thus, other materials have been combined as additive elements in the bioinks[150]. PCL and Poly (propylene fumarate) are polymers utilized in bioprinting due to its superior viscoelastic and rheological properties, biodegradability, and biocompatibility compared to the natural compounds. Synthetic hydrogels such as PEGDA are used as resins in 3D bioprinting, where cells can be entrapped[147]. The major limitation of these kind of hydrogels is that the bioprinted structure tends to collapse because of low viscosity and low mechanical strength[151]. New generation bioprinters have enabled us to print combinatorial bioinks with spatial and nanoscale resolution in seamless swift ways, aiming to reproduce the complex architecture of the native tissues. There are different types of bioinks available depends up on the tissue structure. One example is that researchers bioprinted hepatic tissue constructs using iPSC derived hepatocytes, endothelial cells, and mesenchymal cells resuspended in two different bioinks; GelMA with stiffness similar to healthy liver tissues, and a mix of glycidyl methacrylate-hyaluronic acid/GelMA which supported vascularization[73,152]. New generation polymers responsive to light, thermal, magnetic, humidity, and pH stimuli would allow the 3D bioprinting to leap to the next level.
6 Challenges associated with the use of reprogrammed iPSCs and bioprinting:
The combinatorial application of 3D bioprinting and iPSC technologies would have a major impact on regenerative medicine research. However, how much have we achieved to take this technique to clinics and how far we have to go? Many obstacles still remain regarding the production of safer iPSCs that are to be resolved to take full advantage of this technology for therapeutic purpose[67]. One of the most important problems is the use of retroviral and lentiviral vectors to introduce the transcription factor genes into somatic cells for cell reprogramming, which can cause mutagenesis and tumor induction in the host cell. The iPSCs derived using viral vectors may be still suitable for the study of disease mechanisms or for drug testing but they lack the suitability for clinical bioprinting. The use of non-integrative methods and small molecules to activate the pluripotency program in somatic cells represents the safest approach to produce clinical grade iPSCs cells. High throughput screening to identify small molecules for cell reprogramming is ongoing in many laboratories, with a goal to establish iPSCs free of any exogenously introduced DNA fragments. Incompletely differentiated cells evoked immune response in transplanted animals[153]. Therefore, obtaining completely differentiated cells for therapeutic purpose are of prime importance. Futuristic technology should be focused on establishing safe strategies for genetic modification of iPSCs, devise efficient methods for differentiation and purification of iPSCs into required cell types in vitro for transplantation.
After production of iPSCs, there are limitations in the bioprinting process itself and associated challenges in the preparation of optimized bioinks suitable for each cell type. 3D bioprinting has the advantage of reconstructing complex structures from computed tomography or magnetic resonance imaging images and producing accurate structures from predetermined digital designs such as computer-aided design models. The support scaffold materials with suitable mechanical and biological properties can be designed and printed using advanced 3D printers. Directed differentiation of printed iPSCs to different cell lineages is required for organ printing. When we use undifferentiated iPSCs for bioprinting, the printing parameters should be adjusted to avoid any mechanical damage to the cells, as iPSCs are highly sensitive cells. Depending on the type of the bioprinting method used, the cells are exposed to high shear forces, radiation-induced damage, and electric or thermal stresses during the printing process[154,155].
Vascularization and innervation of the bioprinted tissue are a challenge to achieve. Bioprinted iPSC constructs are unable to form long-term viable and vascularized tissue. To resolve this problem, researchers at Wyss institute recently developed a method called sacrificial writing into functional tissue (SWIFT), which is a multistep biomanufacturing process that involves creating organ building blocks composed of hundreds of thousands of iPSCs and then rapidly 3D bioprinting vasculature into those building blocks[70]. The SWIFT method could create a perfusable cardiac tissue that fuses and beats synchronously for more than a week, taking the field of bioprinting vascularized functional tissues using iPSCs to the next level.
7 Future perspectives for iPSCs in bioprinting
Despite the challenges associated with the use of reprogrammed iPSCs and limitations of bioprinting, the potential of bioprinting iPSC-derived tissue is tremendous in the health-care field. Resolution of these challenges will have significant implications in the understanding of human diseases and will have major effects on the treatment of these diseases. Future perspectives of bioprinting iPSCs should focus on:
•Establishing xeno-free and footprint-free clinical-grade iPSC reprogramming protocols: The use of non-integrative methods and small molecules should be further explored. High throughput screening to identify small molecules for cell reprogramming to establish iPSCs free of any exogenously introduced DNA fragments would be potential area to focus on.
•Development of tissue-specific bioinks for bioprinting: New bioinks with tunable mechanical and rheological properties that mimic the native tissue ECM is to be developed and a deeper understanding of cell-bioink interactions must be sought as the mechanobiology and the molecular pathways would have a major effect on the differentiation of the bioprinted iPSCs.
•Improved bioprinting strategies to mitigate harmful effects on cells: Since iPSCs are sensitive cells (not as sturdy as cancer cell lines), the mechanical, thermal, or chemical stressors induced by the bioprinting process might result in cell-phenotype changes and functionality. Strategies to mitigate the exposure of cells to these process-induced stressors must be developed.
•Integrated bioreactor systems for tissue maturation: Bioprinting of functional tissues with iPSC-derived cells would be successful only if they can be matured and maintained over a long-term in physiologically-relevant environments. Hence, development of suitable post-processing strategies such as integrated perfusion bioreactor systems becomes necessary.
•Pathway for clinical translation: A coordinated effort between the clinicians, scientists, and bioengineers in solving the technological limitations and support from the government and policy-makers would go a long way in establishing a pathway for clinical translation of bioprinted iPSC-derived tissues for regenerative medicine, disease modeling and drug testing.
Conflicts of interest
The authors declare no conflict of interest.
References
- 1.Takahashi K, Yamanaka S. Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell. 2006;126:663–76. doi: 10.1016/j.cell.2006.07.024. DOI:10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
- 2.Yu J, Vodyanik MA, Smuga-Otto K, et al. Thomson, Induced Pluripotent Stem Cell Lines Derived from Human Somatic Cells. Science. 2007;318:1917–20. doi: 10.1126/science.1151526. DOI:10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
- 3.Zhang Y, Chen K, Sloan SA, et al. An RNA-sequencing Transcriptome and Splicing Database of Glia, Neurons, and Vascular Cells of the Cerebral Cortex. J Neurosci. 2004;34:11929–47. doi: 10.1523/JNEUROSCI.1860-14.2014. DOI:10.1523/jneurosci.1860-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chappell J, Dalton S. Roles for MYC in the Establishment and Maintenance of Pluripotency. Cold Spring Harb. Perspect Med. 2013;3:a014381. doi: 10.1101/cshperspect.a014381. DOI:10.1101/cshperspect.a014381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhang P, Zhang X, Brown J, et al. Global Healthcare Expenditure on Diabetes for 2010 and 2030. Diabetes Res Clin Pract. 2010;87:293–301. doi: 10.1016/j.diabres.2010.01.026. DOI:10.1016/j.diabres.2010.12.025. [DOI] [PubMed] [Google Scholar]
- 6.Han JW, Yoon YS. Induced Pluripotent Stem Cells:Emerging Techniques for Nuclear Reprogramming. Antioxidants Redox Signal. 2011;15:1799–820. doi: 10.1089/ars.2010.3814. DOI:10.1089/ars.2010.3814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.An Z, Liu P, Zheng J, et al. Sox2 and Klf4 as the Functional Core in Pluripotency Induction without Exogenous Oct4. Cell Rep. 2019;29:1986–2000. doi: 10.1016/j.celrep.2019.10.026. DOI:10.1016/j.celrep.2019.10.026. [DOI] [PubMed] [Google Scholar]
- 8.Chen IP, Fukuda K, Fusaki N, et al. Induced Pluripotent Stem Cell Reprogramming by Integration-free Sendai Virus Vectors from Peripheral Blood of Patients with Craniometaphyseal Dysplasia. Cell Reprogram. 2013;15:503–13. doi: 10.1089/cell.2013.0037. DOI:10.1089/cell.2013.0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sridharan R, Tchieu J, Mason MJ, et al. Role of the Murine Reprogramming Factors in the Induction of Pluripotency. Cell. 2009;136:364–77. doi: 10.1016/j.cell.2009.01.001. DOI:10.1016/j.cell.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kim JB, Greber B, Arazo-Bravo MJ, et al. Direct Reprogramming of Human Neural Stem Cells by OCT4. Nature. 2009;461:649–53. doi: 10.1038/nature08436. DOI:10.1038/nature08436. [DOI] [PubMed] [Google Scholar]
- 11.Stadtfeld M, Hochedlinger K. Induced Pluripotency:History, Mechanisms, and Applications. Genes Dev. 2010;24:2239–63. doi: 10.1101/gad.1963910. DOI:10.1101/gad.1963910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Yee JK. Turning Somatic Cells into Pluripotent Stem Cells. Nat Educ. 2010;3:25. [Google Scholar]
- 13.Mertens J, Reid D, Lau S, et al. Aging in a Dish:iPSC-Derived and Directly Induced Neurons for Studying Brain Aging and Age-Related Neurodegenerative Diseases. Annu Rev Genet. 2018;52:271–93. doi: 10.1146/annurev-genet-120417-031534. DOI:10.1146/annurev-genet-120417-031534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Penney J, Ralvenius WT, Tsai LH. Modeling Alzheimer's Disease with iPSC-Derived Brain Cells. Mol Psychiatry. 2019;25:148–67. doi: 10.1038/s41380-019-0468-3. DOI:10.1038/s41380-019-0468-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Chen IY, Matsa E, Wu JC. Induced Pluripotent Stem Cells:At the Heart of Cardiovascular Precision Medicine. Nat Rev Cardiol. 2016;13:333–49. doi: 10.1038/nrcardio.2016.36. DOI:10.1038/nrcardio.2016.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Koch L, Deiwick A, Franke A, et al. Laser Bioprinting of Human Induced Pluripotent Stem Cells the Effect of Printing and Biomaterials on Cell Survival, Pluripotency, and Differentiation. Biofabrication. 2018;10:035005. doi: 10.1088/1758-5090/aab981. DOI:10.1088/1758-5090/aab981. [DOI] [PubMed] [Google Scholar]
- 17.Maiullari F, Costantini M, Milan M, et al. A Multi-cellular 3D Bioprinting Approach for Vascularized Heart Tissue Engineering Based on HUVECs and iPSC-Derived Cardiomyocytes. Sci Rep. 2018;8:13532. doi: 10.1038/s41598-018-31848-x. DOI:10.1038/s41598-018-31848-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Yeung E, Fukunishi T, Bai Y, et al. Cardiac Regeneration Using Human-induced Pluripotent Stem Cell-derived Biomaterial-free 3D-bioprinted Cardiac Patch In Vivo. J Tissue Eng Regen Med. 2019;13:2031-9. doi: 10.1002/term.2954. DOI:10.1002/term.2954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Faulkner-Jones A, Fyfe C, Cornelissen DJ, et al. Bioprinting of Human Pluripotent Stem Cells and their Directed Differentiation into Hepatocyte-like Cells for the Generation of Mini-livers in 3D. Biofabrication. 2015;7:044102. doi: 10.1088/1758-5090/7/4/044102. doi.org/10.1088/1758-5090/7/4/044102. [DOI] [PubMed] [Google Scholar]
- 20.Nguyen HX, Hooshmand MJ, Saiwai H, et al. Systemic Neutrophil Depletion Modulates the Migration and Fate of Transplanted Human Neural Stem Cells to Rescue Functional Repair. J Neurosci. 2017;37:9269–87. doi: 10.1523/JNEUROSCI.2785-16.2017. DOI:10.1523/jneurosci.2785-16.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Li Y, Lu B, Sheng L, et al. Hexokinase 2-dependent Hyperglycolysis Driving Microglial Activation Contributes to Ischemic Brain Injury. J Neurochem. 2018;144:186–200. doi: 10.1111/jnc.14267. DOI:10.1111/jnc.14267. [DOI] [PubMed] [Google Scholar]
- 22.Koch SC, Acton D, Goulding M. Spinal Circuits for Touch, Pain, and Itch. Annu Rev Physiol. 2018;80:189–217. doi: 10.1146/annurev-physiol-022516-034303. DOI:10.1146/annurev-physiol-022516-034303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gungor-Ozkerim PS, Inci I, Zhang YS, et al. Bioinks for 3D Bioprinting:An Overview. Biomater Sci. 2018;6:915–46. doi: 10.1039/c7bm00765e. DOI:10.1039/c7bm00765e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Montero FE, Rezende RA, da Silva JV, et al. Development of a Smart Bioink for Bioprinting Applications. Front Mech Eng. 2019;5:56. DOI:10.3389/fmech.2019.00056. [Google Scholar]
- 25.Noor N, Shapira A, Edri R, et al. 3D Printing of Personalized Thick and Perfusable Cardiac Patches and Hearts. Adv Sci. 2019;6:3–10. doi: 10.1002/advs.201900344. DOI:10.1002/advs.201970066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Gopinathan J, Noh I. Recent Trends in Bioinks for 3D Printing. Biomater Res. 2018;22:11. doi: 10.1186/s40824-018-0122-1. DOI:110.1186/s40824-018-0122-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Gu J, Bradbury J, Xiong C, et al. Non-autoregressive Neural Machine Translation. 6thInternational Conference Learning Representations 2018 Conference Track Proceeding 2018 [Google Scholar]
- 28.Rao ZMS, Malik N. Assessing iPSC Reprogramming Methods for their Suitability in Translational Medicine. J Cell Biochem. 2012;113:3061–8. doi: 10.1002/jcb.24183. DOI:10.1002/jcb.24183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kamath A, Ternes A, McGowan S, et al. Efficient Method to Create Integration-free, Virus-free, Myc and Lin28-free Human Induced Pluripotent Stem Cells from Adherent Cells. Future Sci OA. 2017;3:FSO211. doi: 10.4155/fsoa-2017-0028. DOI:10.4155/fsoa-2017-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Malik N, Rao MS. A Review of the Methods for Human iPSC Derivation. Methods Mol Biol. 2013;997:23–33. doi: 10.1007/978-1-62703-348-0_3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Omole AE, Fakoya AO. Ten Years of Progress and Promise of Induced Pluripotent Stem Cells:Historical Origins, Characteristics, Mechanisms, Limitations, and Potential Applications. PeerJ. (e4370) 2018;6 doi: 10.7717/peerj.4370. DOI:10.7717/peerj.4370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Shi Y, Inoue H, Wu JC, et al. Induced Pluripotent Stem Cell Technology:A Decade of Progress. Nat Rev Drug Discov. 2017;16:115–30. doi: 10.1038/nrd.2016.245. DOI:10.1038/nrd.2016.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yamanaka S. Induced Pluripotent Stem Cells:Past, Present, and Future. Cell Stem Cell. 2012;10:678–84. doi: 10.1016/j.stem.2012.05.005. DOI:10.1016/j.stem.2012.05.005. [DOI] [PubMed] [Google Scholar]
- 34.Takahashi K, Tanabe K, Ohnuki M, et al. Yamanaka, Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell. 2007;131:861–72. doi: 10.1016/j.cell.2007.11.019. DOI:10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
- 35.Schlaeger TM, Daheron L, Brickler TR, et al. A Comparison of Non-integrating Reprogramming Methods. Nat Biotechnol. 2015;33:58–63. doi: 10.1038/nbt.3070. DOI:10.1038/nbt.3070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Junying Y, Kejin H, Kim SO, et al. Human Induced Pluripotent Stem Cells Free of Vector and Transgene Sequences. Science. 2009;324:797–801. doi: 10.1126/science.1172482. DOI:10.1126/science.1172482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Shao L, Fujii H, Colmegna I, et al. Deficiency of the DNA Repair Enzyme ATM in Rheumatoid Arthritis. J Exp Med. 2009;206:1435–49. doi: 10.1084/jem.20082251. DOI:10.1084/jem.20082251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Carey BW, Markoulaki A, Hanna J, et al. Reprogramming of Murine and Human Somatic Cells Using a Single Polycistronic Vector. Proc Natl Acad Sci USA. 2009;106:157–62. doi: 10.1073/pnas.0811426106. DOI:10.1073/pnas.0811426106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.McGrath PS, Diette N, Kogut I, et al. RNA-based Reprogramming of Human Primary Fibroblasts into Induced Pluripotent Stem Cells. J Vis Exp. 2018;141:58687. doi: 10.3791/58687. DOI:10.3791/58687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kogut I, McCarthy SM, Pavlova M, et al. High-efficiency RNA-based Reprogramming of Human Primary Fibroblasts. Nat Commun. 2018;9:745. doi: 10.1038/s41467-018-03190-3. DOI:10.1038/s41467-018-03190-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Qiao S, Deng Y, Li S, et al. Partially Reprogrammed Induced Pluripotent Stem Cells Using MicroRNA Cluster miR-302s in Guangxi Bama Minipig Fibroblasts. Cell Reprogram. 2019;21:229–37. doi: 10.1089/cell.2019.0035. DOI:10.1089/cell.2019.0035. [DOI] [PubMed] [Google Scholar]
- 42.Baker M. MicroRNAs Boost Reprogramming, Boot Out cMyc. Nat Reports Stem Cells. 2009;1:46. DOI:10.1038/stemcells.2009.62. [Google Scholar]
- 43.Hirschi KK, Li S, Roy K. Induced Pluripotent Stem Cells for Regenerative Medicine. Annu Rev Biomed Eng. 2014;16:277–94. doi: 10.1146/annurev-bioeng-071813-105108. DOI:10.1146/annurev-bioeng-071813-105108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Worringer KA, Rand TA, Hayashi Y, et al. The Let-7/LIN-41 Pathway Regulates Reprogramming to Human Induced Pluripotent Stem Cells by Controlling Expression of Prodifferentiation Genes. Cell Stem Cell. 2014;14:40–52. doi: 10.1016/j.stem.2013.11.001. DOI:10.1016/j.stem.2013.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kim D, Kim CH, J. Il Moon, et al. Generation of Human Induced Pluripotent Stem Cells by Direct Delivery of Reprogramming Proteins. Cell Stem Cell. 2009;4:472–6. doi: 10.1016/j.stem.2009.05.005. DOI:10.1016/j.stem.2009.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Zhou H, Wu S, Joo JY, et al. Generation of Induced Pluripotent Stem Cells Using Recombinant Proteins. Cell Stem Cell. 2009;4:381–4. doi: 10.1016/j.stem.2009.04.005. DOI:10.1016/j.stem.2009.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Seo BJ, Hong YJ, Do JT. Cellular Reprogramming Using Protein and Cell-Penetrating Peptides. Int J Mol Sci. 2017;18:552. doi: 10.3390/ijms18030552. DOI:10.3390/ijms18030552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lin T, Wu S. Reprogramming with Small Molecules Instead of Exogenous Transcription Factors. Stem Cells Int. 20152015:794632. doi: 10.1155/2015/794632. DOI:10.1155/2015/794632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Huangfu D, Maehr R, Guo W, et al. Induction of Pluripotent Stem Cells by Defined Factors is Greatly Improved by Small-molecule Compounds. Nat Biotechnol. 2008;26:795–7. doi: 10.1038/nbt1418. DOI:10.1038/nbt1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Shi Y, Desponts C, Do JT, et al. Induction of Pluripotent Stem Cells from Mouse Embryonic Fibroblasts by Oct4 and Klf4 with Small-Molecule Compounds. Cell Stem Cell. 2008;3:568–74. doi: 10.1016/j.stem.2008.10.004. DOI:10.1016/j.stem.2008.10.004. [DOI] [PubMed] [Google Scholar]
- 51.Vijayavenkataraman S, Kannan S, Cao T, et al. 3D-Printed PCL/PPy Conductive Scaffolds as Three-Dimensional Porous Nerve Guide Conduits (NGCs) for Peripheral Nerve Injury Repair. Front Bioeng Biotechnol. 2019;7:266. doi: 10.3389/fbioe.2019.00266. DOI:10.3389/fbioe.2019.00266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hou P, Li Y, Zhang X, et al. Pluripotent Stem Cells Induced from Mouse Somatic Cells by Small-molecule Compounds. Science. 2013;341:651–4. doi: 10.1126/science.1239278. DOI:10.1126/science.1239278. [DOI] [PubMed] [Google Scholar]
- 53.Zhang Y, Hu W, Ma K, et al. Reprogramming of Keratinocytes as Donor or Target Cells Holds Great Promise for Cell Therapy and Regenerative Medicine. Stem Cell Rev Reports. 2019;15:680–9. doi: 10.1007/s12015-019-09900-8. DOI:10.1007/s12015-019-09900-8. [DOI] [PubMed] [Google Scholar]
- 54.Aasen T, Raya A, Barrero MJ, et al. Efficient and Rapid Generation of Induced Pluripotent Stem Cells from Human Keratinocytes. Nat Biotechnol. 2008;26:1276–84. doi: 10.1038/nbt.1503. DOI:10.1038/nbt.1503. [DOI] [PubMed] [Google Scholar]
- 55.Sun N, Panetta NJ, Gupta DM, et al. Feeder-free Derivation of Induced Pluripotent Stem Cells from Adult Human Adipose Stem Cells. Proc Natl Acad Sci USA. 2009;106:15720–5. doi: 10.1073/pnas.0908450106. DOI:10.1073/pnas.0908450106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Medvedev SP, Shevchenko AI, Zakian SM. Induced Pluripotent Stem Cells:Problems and Advantages when Applying them in Regenerative Medicine. Acta Nat. 2010;2:18–27. DOI:10.32607/20758251-2010-2-2-18-27. [PMC free article] [PubMed] [Google Scholar]
- 57.Kim Y, Rim YA, Yi H, et al. The Generation of Human Induced Pluripotent Stem Cells from Blood Cells:An Efficient Protocol Using Serial Plating of Reprogrammed Cells by Centrifugation. Stem Cells Int. 20162016:1329459. doi: 10.1155/2016/1329459. DOI:410.1155/2016/1329459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Okumura T, Horie Y, Lai CY, et al. Robust and Highly Efficient hiPSC Generation from Patient Non-mobilized Peripheral Blood-derived CD34+Cells Using the Auto-erasable Sendai Virus Vector. Stem Cell Res Ther. 2019;10:185. doi: 10.1186/s13287-019-1273-2. DOI:10.1186/s13287-019-1273-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Corbett JL, Duncan SA. iPSC-Derived Hepatocytes as a Platform for Disease Modeling and Drug Discovery. Front Med. 2019;6:265. doi: 10.3389/fmed.2019.00265. DOI:10.3389/fmed.2019.00265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kwong G, Marquez HA, Yang C, et al. Generation of a Purified iPSC-Derived Smooth Muscle-like Population for Cell Sheet Engineering. Stem Cell Reports. 2019;13:499–514. doi: 10.1016/j.stemcr.2019.07.014. DOI:10.1016/j.stemcr.2019.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Abdul Manaph NP, Sivanathan KN, Nitschke J, et al. An Overview on Small Molecule-induced Differentiation of Mesenchymal Stem Cells Into Beta Cells for Diabetic Therapy. Stem Cell Res Ther. 2019;10:2194. doi: 10.1186/s13287-019-1396-5. DOI:10.1186/s13287-019-1396-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Tabar V, Studer L. Pluripotent Stem Cells in Regenerative Medicine:Challenges and Recent Progress. Nat Rev Genet. 2014;15:82–92. doi: 10.1038/nrg3563. DOI:10.1038/nrg3563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kang H, Shih YR, Nakasaki M, et al. Small Molecule Driven Direct Conversion of Human Pluripotent Stem Cells into Functional Osteoblasts. Sci Adv. 2:e1600691. doi: 10.1126/sciadv.1600691. DOI:10.1126/sciadv.1600691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ong CS, Yesantharao P, Huang CY, et al. 3D Bioprinting Using Stem Cells. Pediatr Res. 2018;83:223–31. doi: 10.1038/pr.2017.252. [DOI] [PubMed] [Google Scholar]
- 65.Ozbolat IT, Hospodiuk M. Current Advances and Future Perspectives in Extrusion-based Bioprinting. Biomaterials. 2016;76:321–43. doi: 10.1016/j.biomaterials.2015.10.076. DOI:10.1016/j.biomaterials.2015.10.076. [DOI] [PubMed] [Google Scholar]
- 66.Reid JA, Mollica PA, Johnson GD, et al. Accessible Bioprinting:Adaptation of a Low-cost 3D-Printer for Precise Cell Placement and Stem Cell Differentiation. Biofabrication. 2016;8:025017. doi: 10.1088/1758-5090/8/2/025017. DOI:10.1088/1758-5090/8/2/025017. [DOI] [PubMed] [Google Scholar]
- 67.Gu Q, Tomaskovic-Crook E, Wallace GG, et al. 3D Bioprinting Human Induced Pluripotent Stem Cell Constructs for In Situ Cell Proliferation and Successive Multilineage Differentiation. Adv Healthc Mater. 2017;6:1700175. doi: 10.1002/adhm.201700175. DOI:10.1002/adhm.201700175. [DOI] [PubMed] [Google Scholar]
- 68.Colosi S, Soloperto B, Benedetti T, et al. 3D Bioprinted Human Cortical Neural Constructs Derived from Induced Pluripotent Stem Cells. J Clin Med. 2019;8:1595. doi: 10.3390/jcm8101595. DOI:10.3390/jcm8101595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Nguyen D, Hägg DA, Forsman A, et al. Cartilage Tissue Engineering by the 3D Bioprinting of iPS Cells in a Nanocellulose/Alginate Bioink. Sci Rep. 2017;7:6684. doi: 10.1038/s41598-017-00690-y. DOI:10.1038/s41598-017-00690-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Skylar-Scott MA, Uzel SG, Nam LL, et al. Biomanufacturing of Organ-Specific Tissues with High Cellular Density and Embedded Vascular Channels. Sci Adv. 2019;5:eaaw2459. doi: 10.1126/sciadv.aaw2459. DOI:10.1126/sciadv.aaw2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhang C, Yang F, Cornelia R, et al. Hypoxia-inducible Factor-1 is a Positive Regulator of Sox9 Activity in Femoral Head Osteonecrosis. Bone. 2011;48:507–13. doi: 10.1016/j.bone.2010.10.006. DOI:10.1016/j.bone.2010.10.006. [DOI] [PubMed] [Google Scholar]
- 72.Sun AX, Lin H, Beck AM, et al. Projection Stereolithographic Fabrication of Human Adipose Stem Cell-Incorporated Biodegradable Scaffolds for Cartilage Tissue Engineering. Front Bioeng Biotechnol. 2015;3:115. doi: 10.3389/fbioe.2015.00115. DOI:10.3389/fbioe.2015.00115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ma X, Qu X, Zhu W, et al. Deterministically Patterned Biomimetic Human iPSC-Derived Hepatic Model Via Rapid 3D Bioprinting. Proc Natl Acad Sci. 2016;113:2206–11. doi: 10.1073/pnas.1524510113. DOI:10.1073/pnas.1524510113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Fairbanks BD, Schwartz MP, Bowman CN, et al. Photoinitiated Polymerization of PEG-diacrylate with Lithium Phenyl-2,4,6-Trimethylbenzoylphosphinate:Polymerization Rate and Cytocompatibility. Biomaterials. 2009;30:6702–7. doi: 10.1016/j.biomaterials.2009.08.055. DOI:10.1016/j.biomaterials.2009.08.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ng WL, Lee JM, Zhou M, et al. Vat Polymerization-Based Bioprinting Process, Materials, Applications and Regulatory Challenges. Biofabrication. 2020;12:022001. doi: 10.1088/1758-5090/ab6034. DOI:410.1088/1758-5090/ab6034. [DOI] [PubMed] [Google Scholar]
- 76.Fattah AR, Meleca E, Mishriki S, et al. In Situ3D Label-Free Contactless Bioprinting of Cells through Diamagnetophoresis. ACS Biomater Sci Eng. 2016;2:2133–8. doi: 10.1021/acsbiomaterials.6b00614. DOI:10.1021/acsbiomaterials.6b00614. [DOI] [PubMed] [Google Scholar]
- 77.Gudapati H, Dey M, Ozbolat I. A Comprehensive Review on Droplet-based Bioprinting:Past, Present and Future. Biomaterials. 2016;102:20–42. doi: 10.1016/j.biomaterials.2016.06.012. DOI:10.1016/j.biomaterials.2016.06.012. [DOI] [PubMed] [Google Scholar]
- 78.Guillotin B, Souquet A, Catros S, et al. Laser Assisted Bioprinting of Engineered Tissue with High Cell Density and Microscale Organization. Biomaterials. 2010;31:7250–6. doi: 10.1016/j.biomaterials.2010.05.055. DOI:10.1016/j.biomaterials.2010.05.055. [DOI] [PubMed] [Google Scholar]
- 79.Li Y, Jiang X, Li L, et al. 3D Printing Human Induced Pluripotent Stem Cells with Novel Hydroxypropyl Chitin Bioink:Scalable Expansion and Uniform Aggregation. Biofabrication. 2018;10:044101. doi: 10.1088/1758-5090/aacfc3. DOI:10.1088/1758-5090/aacfc3. [DOI] [PubMed] [Google Scholar]
- 80.Ng WL, Chua CK, Shen YF. Print Me An Organ!Why We Are Not There Yet. Prog Polym Sci. 2019;97:101145. DOI:10.1016/j.progpolymsci.2019.101145. [Google Scholar]
- 81.Romanazzo S, Nemec S, Roohani I. iPSC Bioprinting:Where are we at? Materials (Basel) 2019;12:2453. doi: 10.3390/ma12152453. DOI:10.3390/ma12152453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.McCauley HA, Wells JM. Pluripotent Stem Cell-derived Organoids:Using Principles of Developmental Biology to Grow Human Tissues in a Dish. Development. 2017;144:958–62. doi: 10.1242/dev.140731. DOI:10.1242/dev.140731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Liu C, Oikonomopoulos A, Sayed N, et al. Modeling Human Diseases with Induced Pluripotent Stem Cells:From 2D to 3D and Beyond. Development. 2018;145:dev156166. doi: 10.1242/dev.156166. DOI:10.1242/dev.156166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zhang B, Gao L, Ma L, et al. 3D Bioprinting:A Novel Avenue for Manufacturing Tissues and Organs. Engineering. 2017;5:777–94. [Google Scholar]
- 85.Varkey M, Visscher DO, van Zuijlen PP, et al. Skin Bioprinting:The Future of Burn Wound Reconstruction? Burn Trauma. 2019;7:8171. doi: 10.1186/s41038-019-0142-7. DOI:10.1186/s41038-019-0142-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kamel RA, Ong JF, Eriksson E, et al. Tissue Engineering of Skin. J Am Coll Surg. 2013;217:533–55. doi: 10.1016/j.jamcollsurg.2013.03.027. [DOI] [PubMed] [Google Scholar]
- 87.Yang B, Lui C, Yeung E, et al. A Net Mold-Based Method of Biomaterial-Free Three-Dimensional Cardiac Tissue Creation. Tissue Eng Part C Methods. 2019;25:243–52. doi: 10.1089/ten.TEC.2019.0003. DOI:10.1089/ten.tec.2019.0003. [DOI] [PubMed] [Google Scholar]
- 88.Vijayavenkataraman S, Kannan S, Cao T, et al. 3D-Printed PCL/PPy Conductive Scaffolds as Three-Dimensional Porous Nerve Guide Conduits (NGCs) for Peripheral Nerve Injury Repair. Front Bioeng Biotechnol. 2019;7:266. doi: 10.3389/fbioe.2019.00266. DOI:10.3389/fbioe.2019.00266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chen YM, Chen LH, Li MP, et al. Xeno-free Culture of Human Pluripotent Stem Cells on Oligopeptide-Grafted Hydrogels with Various Molecular Designs. Sci Rep. 2017;7:45146. doi: 10.1038/srep45146. DOI:10.1038/srep45146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Wiley LA, Anfinson KR, Cranston CM, et al. Generation of Xeno-Free, cGMP-Compliant Patient-Specific iPSCs from Skin Biopsy. Curr Protoc Stem Cell Biol. 2017;42:4A.12.1–4A.12.4. doi: 10.1002/cpsc.30. DOI:10.1002/cpsc.30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Pruksananonda K, Rungsiwiwut R. Pluripotent Stem Cells:From the Bench to the Clinic. BoD-Books on Demand; Norderstedt, Germany: 2016. Moving toward xeno-free culture of human pluripotent stem cells. DOI:10.5772/62770. [Google Scholar]
- 92.Boreström C, Simonsson S, Enochson L, et al. Footprint-Free Human Induced Pluripotent Stem Cells From Articular Cartilage With Redifferentiation Capacity:A First Step Toward a Clinical-Grade Cell Source. Stem Cells Transl Med. 2014;3:433–47. doi: 10.5966/sctm.2013-0138. DOI:10.5966/sctm.2013-0138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Attwood S, Edel M. iPS-Cell Technology and the Problem of Genetic Instability can it ever be Safe for Clinical Use? J Clin Med. 2019;8:288. doi: 10.3390/jcm8030288. DOI:10.3390/jcm8030288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Matz P, Wruck W, Fauler B, et al. Footprint-free Human Fetal Foreskin Derived iPSCs:A Tool for Modeling Hepatogenesis Associated Gene Regulatory Networks. Sci Rep. 2017;7:310. doi: 10.1038/s41598-017-06546-9. DOI:10.1038/s41598-017-06546-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Atkinson MA, Eisenbarth GS, Michels AW. Type 1 Diabetes. Lancet. 2014;383:69–82. doi: 10.1016/S0140-6736(13)60591-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Katsarou A, Gudbjörnsdottir S, Rawshani A, et al. Type 1 Diabetes Mellitus. Nat Rev Dis Prim. 2017;3:17016. doi: 10.1038/nrdp.2017.16. [DOI] [PubMed] [Google Scholar]
- 97.Kim J, Shim IK, Hwang DG, et al. 3D Cell Printing of Islet-laden Pancreatic Tissue-derived Extracellular Matrix Bioink Constructs for Enhancing Pancreatic Functions. J Mater Chem B. 2019;7:1773–81. doi: 10.1039/c8tb02787k. DOI:10.1039/c8tb02787k. [DOI] [PubMed] [Google Scholar]
- 98.Taalesen H, Ketner MA. Milestone in Development of 3D Bioprinting of Biomimetic Pancreas to Treat Diabetes. [Last accessed on 2020 Jun 20];University of Oslo, Hybrid Technology Hub, Oslo. 2018 Available from:https://www.med.uio.no/hth/english/news-and-events/news/3dbioprinting.html .
- 99.Davies S. Mag Des Innovations. Celprogen Inc; Torrance: 2016. Successfully 3D Print Functioning Pancreas Model. [Google Scholar]
- 100.Ebrahimi M, Asbagh FA. Pathogenesis and Causes of Premature Ovarian Failure:An Update. Int J Fertil Steril. 2011;5:54–65. [PMC free article] [PubMed] [Google Scholar]
- 101.Laronda MM, Rutz AL, Xiao S, et al. A Bioprosthetic Ovary Created Using 3D Printed Microporous Scaffolds Restores Ovarian Function in Sterilized Mice. Nat Commun. 2017;8:15261. doi: 10.1038/ncomms15261. DOI:10.1038/ncomms15261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Irvine SA, Agrawal A, Lee BH, et al. Printing cell-laden gelatin constructs by free-form fabrication and enzymatic protein crosslinking. Biomed Microdevices. 2015;17:16. doi: 10.1007/s10544-014-9915-8. DOI:10.1007/s10544-014-9915-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bulanova EA, Koudan EV, Degosserie J, et al. Bioprinting of a Functional Vascularized Mouse Thyroid Gland Construct. Biofabrication. 2017;9:034105. doi: 10.1088/1758-5090/aa7fdd. DOI:10.1088/1758-5090/aa7fdd. [DOI] [PubMed] [Google Scholar]
- 104.International Space Station Program Science Forum. International Space Station Benefits for Humanity. [Last accessed on 2020 Jun 20];NASA. (3rd ed). 2018 Available from:https://www.nasa.gov/sites/default/files/atoms/files/benefits-for-humanity_third.pdf .
- 105.Hamazaki T, El Rouby N, Fredette NC, et al. Concise Review:Induced Pluripotent Stem Cell Research in the Era of Precision Medicine. Stem Cells. 2017;35:545–50. doi: 10.1002/stem.2570. DOI:10.1002/stem.2570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Walker C, Mojares E, Del Río Hernández A. Role of Extracellular Matrix in Development and Cancer Progression. Int J Mol Sci. 2018;19:3028. doi: 10.3390/ijms19103028. DOI:10.3390/ijms19103028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ma X, Liu J, Zhu W, et al. 3D Bioprinting of Functional Tissue Models for Personalized Drug Screening and In Vitro Disease Modeling. Adv Drug Deliv Rev. 2018;132:235–51. doi: 10.1016/j.addr.2018.06.011. DOI:10.1016/j.addr.2018.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Antill-O'Brien N, Bourke J, O'Connell CD. Layer-by-layer:The case for 3D bioprinting neurons to create patient-specific epilepsy models. Materials (Basel) 2019;12:3218. doi: 10.3390/ma12193218. DOI:10.3390/ma12193218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Alonzo M, AnilKumar S, Roman B, et al. 3D Bioprinting of Cardiac Tissue and Cardiac Stem Cell Therapy. Transl Res. 2019;211:64–83. doi: 10.1016/j.trsl.2019.04.004. DOI:10.1016/j.trsl.2019.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zimmermann WH, Cesnjevar R. Cardiac Tissue Engineering:Implications for Pediatric Heart Surgery. Pediatr Cardiol. 2009;30:716–23. doi: 10.1007/s00246-009-9405-6. DOI:10.1007/s00246-009-9405-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tzatzalos E, Abilez OJ, Shukla P, et al. Engineered Heart Tissues and Induced Pluripotent Stem Cells:Macro and Microstructures for Disease Modeling, Drug Screening, and Translational Studies. Adv Drug Deliv Rev. 2016;96:234–44. doi: 10.1016/j.addr.2015.09.010. DOI:10.1016/j.addr.2015.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Yoshida Y, Yamanaka S. Induced Pluripotent Stem Cells 10 Years Later. Circ Res. 2017;120:1958–68. doi: 10.1161/CIRCRESAHA.117.311080. DOI:10.1161/circresaha.117.311080. [DOI] [PubMed] [Google Scholar]
- 113.Hinson JT, Chopra A, Nafissi N, et al. Titin Mutations in iPS Cells Define Sarcomere Insufficiency as a Cause of Dilated Cardiomyopathy. Science. 2015;349:982–6. doi: 10.1126/science.aaa5458. DOI:10.1126/science.aaa5458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Tiburcy M, Hudson JE, Balfanz P, et al. Defined Engineered Human Myocardium with Advanced Maturation for Applications in Heart Failure Modeling and Repair. Circulation. 2017;135:1832–47. doi: 10.1161/CIRCULATIONAHA.116.024145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Takebe T, Zhang B, Radisic M. Synergistic Engineering:Organoids Meet Organs-on-a-Chip. Cell Stem Cell. 2017;21:297–300. doi: 10.1016/j.stem.2017.08.016. DOI:10.1016/j.stem.2017.08.016. [DOI] [PubMed] [Google Scholar]
- 116.Bhatia SN, Ingber DE. Microfluidic Organs-on-chips. Nat Biotechnol. 2014;32:760–72. doi: 10.1038/nbt.2989. DOI:10.1038/nbt.2989. [DOI] [PubMed] [Google Scholar]
- 117.Huh D, Matthews BD, Mammoto A, et al. Ingber, Reconstituting Organ-level Lung Functions on a Chip. Science. 2010;328:1662–8. doi: 10.1126/science.1188302. DOI:10.1126/science.1188302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Low LA, Tagle DA. Tissue Chips-innovative Tools for Drug Development and Disease Modeling. Lab Chip. 2017;17:3026–36. doi: 10.1039/c7lc00462a. DOI:10.1039/c7lc00462a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Eiraku M, Watanabe K, Matsuo-Takasaki M, et al. Self-Organized Formation of Polarized Cortical Tissues from ESCs and its Active Manipulation by Extrinsic Signals. Cell Stem Cell. 2008;3:519–32. doi: 10.1016/j.stem.2008.09.002. DOI:10.1016/j.stem.2008.09.002. [DOI] [PubMed] [Google Scholar]
- 120.Montine TJ, Phelps CH, Beach TG, et al. National Institute on Aging-Alzheimer's Association Guidelines for the Neuropathologic Assessment of Alzheimer's Disease:A Practical Approach. Acta Neuropathol. 2012;123:1–11. doi: 10.1007/s00401-011-0910-3. DOI:10.1007/s00401-011-0910-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.van Giau V, Lee H, Shim KH, et al. Genome-editing Applications of CRISPR Cas9 to Promote In Vitro Studies of Alzheimer's Disease. Clin Interv Aging. 2018;13:221–33. doi: 10.2147/CIA.S155145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.De Strooper B, Karran E. The Cellular Phase of Alzheimer's Disease. Cell. 2016;164:603–15. doi: 10.1016/j.cell.2015.12.056. DOI:10.1016/j.cell.2015.12.056. [DOI] [PubMed] [Google Scholar]
- 123.Vijayavenkataraman S, Thaharah S, Zhang S, et al. 3D-Printed PCL/rGO Conductive Scaffolds for Peripheral Nerve Injury Repair. Artif Organs. 2019;43:515–23. doi: 10.1111/aor.13360. DOI:10.1111/aor.13360. [DOI] [PubMed] [Google Scholar]
- 124.Chambers SM, Fasano CA, Papapetrou EP, et al. Highly Efficient Neural Conversion of Human ES and iPS Cells by Dual Inhibition of SMAD Signaling. Nat Biotechnol. 2009;27:275–80. doi: 10.1038/nbt.1529. DOI:10.1038/nbt.1529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Ortiz-Virumbrales M, Moreno CL, Kruglikov I, et al. CRISPR/Cas9-Correctable Mutation-related Molecular and Physiological Phenotypes in iPSC-Derived Alzheimer's PSEN2N141I Neurons. Acta Neuropathol Commun. 2017;5:77. doi: 10.1186/s40478-017-0475-z. DOI:10.1186/s40478-017-0475-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Drummond E, Wisniewski T. Alzheimer's Disease:Experimental Models and Reality. Acta Neuropathol. 2017;133:155–75. doi: 10.1007/s00401-016-1662-x. DOI:10.1007/s00401-016-1662-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Li H, Jiang H, Zhang B, et al. Modeling Parkinson's Disease Using Patient-specific Induced Pluripotent Stem Cells. J Parkinsons Dis. 2018;8:479–93. doi: 10.3233/JPD-181353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Torrent R, De Angelis Rigotti F, Dell'Era P, et al. Using iPS Cells Toward the Understanding of Parkinson's Disease. J Clin Med. 2015;4:548–66. doi: 10.3390/jcm4040548. DOI:10.3390/jcm4040548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Zeng XS, Geng WS, Jia JJ. Neurotoxin-Induced Animal Models of Parkinson Disease:Pathogenic Mechanism and Assessment. ASN Neuro. 2018;10:175909141–438. doi: 10.1177/1759091418777438. DOI:10.1177/175909141≉438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Bordoni M, Rey F, Fantini V, et al. From Neuronal Differentiation of iPSCs to 3D Neuro-organoids:Modelling and Therapy of Neurodegenerative Diseases. Int J Mol Sci. 2018;19:3972. doi: 10.3390/ijms19123972. DOI:10.3390/ijms19123972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Thomas M, Willerth SM. 3-D Bioprinting of Neural Tissue for Applications in Cell Therapy and Drug Screening. Front Bioeng Biotechnol. 2017;5:69. doi: 10.3389/fbioe.2017.00069. DOI:10.3389/fbioe.2017.00069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Osborn TM, Beagan J, Isacson O. Increased Motor Neuron Resilience by Small Molecule Compounds that Regulate IGF-II Expression. Neurobiol Dis. 2018;110:218–30. doi: 10.1016/j.nbd.2017.11.002. DOI:10.1016/j.nbd.2017.11.002. [DOI] [PubMed] [Google Scholar]
- 133.Osaki T, Uzel SG, Kamm RD. Microphysiological 3D Model of Amyotrophic Lateral Sclerosis (ALS) from Human iPS-derived Muscle Cells and Optogenetic Motor Neurons. Sci Adv. 2018;4:eaat5847. doi: 10.1126/sciadv.aat5847. DOI:10.1126/sciadv.aat5847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Câmara DA, Mambelli LI, Porcacchia AS, et al. Advances and Challenges on Cancer Cells Reprogramming Using Induced Pluripotent Stem Cells Technologies. J Cancer. 2016;7:2296–303. doi: 10.7150/jca.16629. DOI:10.7150/jca.16629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Papapetrou EP. Patient-derived Induced Pluripotent Stem Cells in Cancer Research and Precision Oncology. Nat Med. 2016;22:1392–401. doi: 10.1038/nm.4238. DOI:10.1038/nm.4238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kotini AG, Chang CJ, Chow A, et al. Stage-Specific Human Induced Pluripotent Stem Cells Map the Progression of Myeloid Transformation to Transplantable Leukemia. Cell Stem Cell. 2017;20:315–28. doi: 10.1016/j.stem.2017.01.009. DOI:10.1016/j.stem.2017.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hu K, Yu J, Suknuntha K, et al. Efficient Generation of Transgene-free induced Pluripotent Stem Cells from Normal and Neoplastic Bone Marrow and Cord Blood Mononuclear Cells. Blood. 2011;117:e109–19. doi: 10.1182/blood-2010-07-298331. DOI:10.1182/blood-2010-07-298331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Gandre-Babbe S, Paluru P, Aribeana C, et al. Patient-derived Induced Pluripotent Stem Cells Recapitulate Hematopoietic Abnormalities of Juvenile Myelomonocytic Leukemia. Blood. 2013;121:4925–9. doi: 10.1182/blood-2013-01-478412. DOI:10.1182/blood-2013-01-478412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Kotini AG, Chang CJ, Boussaad I, et al. Functional Analysis of a Chromosomal Deletion Associated with Myelodysplastic Syndromes using Isogenic Human Induced Pluripotent Stem Cells. Nat Biotechnol. 2015;33:646–55. doi: 10.1038/nbt.3178. DOI:10.1038/nbt.3178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Fang Y, Eglen RM. Three-Dimensional Cell Cultures in Drug Discovery and Development. SLAS Discov. 2017;22:456–72. doi: 10.1177/1087057117696795. DOI:10.1177/10∁7117696795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Senthebane DA, Rowe A, Thomford NE, et al. The Role of Tumor Microenvironment in Chemoresistance:To Survive, Keep your Enemies Closer. Int J Mol Sci. 2017;18:1586. doi: 10.3390/ijms18071586. DOI:10.3390/ijms18071586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Abbott A. The Lowdown on Animal Testing for Cosmetics. Nature. 2019;2:496. [Google Scholar]
- 143.Skardal A, Mack D, Kapetanovic E, et al. Bioprinted Amniotic Fluid-Derived Stem Cells Accelerate Healing of Large Skin Wounds. Stem Cells Transl Med. 2012;1:792–802. doi: 10.5966/sctm.2012-0088. DOI:10.5966/sctm.2012-0088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Teoh JH, Thamizhchelvan AM, Davoodi P, et al. Investigation of the Application of a Taylor-Couette Bioreactor in the Post-processing of Bioprinted Human Dermal Tissue. Biochem Eng J. 2019;151:107317. DOI:10.1016/j.bej.2019.107317. [Google Scholar]
- 145.Kim BS, Lee JS, Gao G, et al. Direct 3D Cell-printing of Human Skin with Functional Transwell System. Biofabrication. 2017;9:025034. doi: 10.1088/1758-5090/aa71c8. DOI:10.1088/1758-5090/aa71c8. [DOI] [PubMed] [Google Scholar]
- 146.Gentile P, McColgan-Bannon K, Gianone NC, et al. Biosynthetic PCL-Graft-collagen Bulk Material for Tissue Engineering Applications. Materials (Basel) 2017;10:693. doi: 10.3390/ma10070693. DOI:10.3390/ma10070693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Ashammakhi N, Ahadian S, Xu C, et al. Bioinks and Bioprinting Technologies to Make Heterogeneous and Biomimetic tissue Constructs. Mater Today Bio. 2019;1:100008. doi: 10.1016/j.mtbio.2019.100008. DOI:10.1016/j.mtbio.2019.100008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Mosadegh B, Xiong G, Dunham S, et al. Current Progress in 3D Printing for Cardiovascular Tissue Engineering. Biomed Mater (Bristol) 2015;10:034002. doi: 10.1088/1748-6041/10/3/034002. DOI:10.1088/1748-6041/10/3/034002. [DOI] [PubMed] [Google Scholar]
- 149.Mandrycky C, Wang Z, Kim K, et al. 3D Bioprinting for Engineering Complex Tissues. Biotechnol Adv. 2016;34:422–34. doi: 10.1016/j.biotechadv.2015.12.011. DOI:10.1016/j.biotechadv.2015.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Zhang XF, Huang Y, Gao G, et al. Current Progress in Bioprinting. In: Tripathi A, Melo JS, editors. Advances in Biomaterials for Biomedical Applications. Springer; Singapore: 2017. pp. 227–59. DOI:10.1016/j.mtbio.2019.100008. [Google Scholar]
- 151.Skardal A, Atala A. Biomaterials for Integration with 3-D Bioprinting. Annals of Biomedical Engineering. 2015;43:730–46. doi: 10.1007/s10439-014-1207-1. DOI:10.1007/s10439-014-1207-1. [DOI] [PubMed] [Google Scholar]
- 152.Billiet T, Vandenhaute M, Schelfhout J, et al. A Review of Trends and Limitations in Hydrogel-rapid Prototyping for Tissue Engineering. Biomaterials. 2012;33:6020-41. doi: 10.1016/j.biomaterials.2012.04.050. DOI:10.1016/j.biomaterials.2012.04.050. [DOI] [PubMed] [Google Scholar]
- 153.Zhao T, Zhang ZN, Rong Z, et al. Immunogenicity of Induced Pluripotent Stem Cells. Nature. 2011;474:212–6. doi: 10.1038/nature10135. DOI:10.1038/nature10135. [DOI] [PubMed] [Google Scholar]
- 154.Vijayavenkataraman S, Yan WC, Lu WF, et al. 3D Bioprinting of Tissues and Organs for Regenerative Medicine. Adv Drug Deliv Rev. 2018;132:296–332. doi: 10.1016/j.addr.2018.07.004. DOI:10.1016/j.addr.2018.07.004. [DOI] [PubMed] [Google Scholar]
- 155.Zhang S, Vijayavenkataraman S, Lu WF, et al. A Review on the Use of Computational Methods to Characterize, Design, and Optimize Tissue Engineering Scaffolds, with a Potential in 3D Printing Fabrication. J Biomed Mater Res Part B Appl Biomater. 2019;107:1329–51. doi: 10.1002/jbm.b.34226. DOI:10.1002/jbm.b.34226. [DOI] [PubMed] [Google Scholar]