Skip to main content
Journal of Cellular and Molecular Medicine logoLink to Journal of Cellular and Molecular Medicine
. 2016 Apr 21;20(8):1571–1588. doi: 10.1111/jcmm.12839

hiPSC‐derived iMSCs: NextGen MSCs as an advanced therapeutically active cell resource for regenerative medicine

Vikram Sabapathy 1, Sanjay Kumar 1,
PMCID: PMC4956943  PMID: 27097531

Abstract

Mesenchymal stem cells (MSCs) are being assessed for ameliorating the severity of graft‐versus‐host disease, autoimmune conditions, musculoskeletal injuries and cardiovascular diseases. While most of these clinical therapeutic applications require substantial cell quantities, the number of MSCs that can be obtained initially from a single donor remains limited. The utility of MSCs derived from human‐induced pluripotent stem cells (hiPSCs) has been shown in recent pre‐clinical studies. Since adult MSCs have limited capability regarding proliferation, the quantum of bioactive factor secretion and immunomodulation ability may be constrained. Hence, the alternate source of MSCs is being considered to replace the commonly used adult tissue‐derived MSCs. The MSCs have been obtained from various adult and foetal tissues. The hiPSC‐derived MSCs (iMSCs) are transpiring as an attractive source of MSCs because during reprogramming process, cells undergo rejuvination, exhibiting better cellular vitality such as survival, proliferation and differentiations potentials. The autologous iMSCs could be considered as an inexhaustible source of MSCs that could be used to meet the unmet clinical needs. Human‐induced PSC‐derived MSCs are reported to be superior when compared to the adult MSCs regarding cell proliferation, immunomodulation, cytokines profiles, microenvironment modulating exosomes and bioactive paracrine factors secretion. Strategies such as derivation and propagation of iMSCs in chemically defined culture conditions and use of footprint‐free safer reprogramming strategies have contributed towards the development of clinically relevant cell types. In this review, the role of iPSC‐derived mesenchymal stromal cells (iMSCs) as an alternate source of therapeutically active MSCs has been described. Additionally, we also describe the role of iMSCs in regenerative medical applications, the necessary strategies, and the regulatory policies that have to be enforced to render iMSC's effectiveness in translational medicine.

Keywords: mesenchymal stem cells, MSCs, induced pluripotent stem cells, hiPSCs, iMSCs

Introduction

Mesenchymal stromal cells (MSCs) are assorted cell preparations and only a rare subpopulation often referred to as ‘mesenchymal stem cells’ retains clonogenic proliferation ability & multilineage differentiation potential 1. Mesenchymal stem cell preparations are significantly affected by starting cell source/material, such as bone marrow (BM), adipose tissue (AT) or other adult/perinatal tissue source; cell culture surface, media composition and other in vitro tissue culture conditions 1, 2, 9. Furthermore, they acquire phenotypic, biochemical, molecular as well as functional changes during long‐term in vitro culture expansion ending in replicative senescence 7, 8. So far, MSCs are occasionally defined by their plastic adherent growth displaying fibroblast‐like cellular colonies, a panel of positive (CD73, CD90, CD105) and negative cell surface markers (CD11b/CD14, CD34, CD45, CD79α/CD19) for phenotypic characterization and their capacity to differentiate towards at least trilineage differentiations such as adipogenic, osteogenic and chondrogenic lineages 2, 4. Many researchers indicate that plasticity and immunomodulatory capabilities of the MSCs contribute towards unique therapeutic potentials of the MSCs 1, 2. The bone marrow MSCs (BMMSCs) are considered to be the gold standard in the field of MSCs. However, their invasive accessibility and lower proliferation potential significantly undermine their ability to be considered for mainstream therapeutic applications 3. The therapeutic potency of MSCs is often limited because of age or pathologically related impairments regarding cell survival, proliferation and differentiations potential of BMMSCs 4, 5, 6. Before adult MSCs can exert its therapeutic potential in vivo; we must determine reasons behind their limited proliferation capability, quick down‐gradation of their differentiation potentials and secrete minimal protective factors during their expansion ex vivo? 7, 8 The adult MSCs unveil time‐limited functions under both in vivo and in vitro conditions 9, 10. Exploration for an alternate source of MSCs resulted in several groups reporting successful isolation of MSCs like cells from foetal, neonatal 11, 12, 13, 14, 15 and embryonic stem cells (ESCs) 16, 17, 18. As a result of the current deficit in adult MSCs regarding inadequacy in MSCs passages, cell numbers and consistencies in cellular behaviour; alternative, easily accessible, safe and healthy populations of MSCs are being considered for clinical applications 3. The iPSC‐derived MSCs (iMSCs) are emerging as an attractive option for obtaining a substantial population of stem cells in a sustained manner for regenerative medical applications 3. The achievement of cell‐based therapy of MSCs in preclinical trials has precipitated success in human translational applications 19.

Therapeutically active MSCs derived from human bone marrow

In the field of regenerative medicine, human mesenchymal stem cells (hMSCs) have transpired to be a promising candidate. Bone marrow‐derived MSCs (BMMSCs) have been used as a predominant source of MSCs. Bone marrow‐derived MSCs have been successfully used in a significant number of clinical and pre‐clinical applications 20, 21. In the early 20th century, Maximow and Friedenstein were the first to investigate the role of bone marrow fibroblast‐like subset cells in maintaining the haematopoiesis 22. The BMMSCs were first isolated and propagated under in vitro culture conditions in 1970s by Friedenstein et al. 21, 22. In 1991, Arnold Caplan termed the cells MSCs based on the ability of the cells to give rise to distinct tissue lineages 23. Maureen Owen further characterized the MSCs and observed the heterogeneity in its population 21, 24. The in vivo administration of the MSCs in animal and humans has shown to be safe without triggering adverse immune reaction or any tumour formation 25. Subsequently, MSCs have been shown to modulate the immune response and prevent graft‐versus‐host disease (GVHD) 26, 27. Above all MSCs has been demonstrated to be effective in both pre‐clinical and clinical stages in orthopaedic applications, cardiovascular therapies, burns, wounds, ulcers, neurodegenerative disorders, spinal cord injury, autoimmune disorders, etc., 28. Mesenchymal stem cells exert their biological functions through cellular migration, local engraftment, self‐renewal, plasticity, and secretion of various bioactive compounds. These intrinsic characteristics render the MSCs ideal for regenerative medical applications 29. Moreover, MSCs can be engineered to secrete various bioactive factors through viral or non‐viral‐based methods, which enhance the capabilities of MSCs in therapeutic applications 30, 31. The low proliferation potential of the adult BMMSCs renders the BMMSCs unsuitable for clinical applications 32. Further, the limited accessibility, difficulty in obtaining patient consent and invasive procedure contribute towards un‐usability of BMMSCs for routine clinical treatment. During specific times extraction of autologous MSCs from the patients might be counter‐productive for the management of the patients 3. The BMMSCs are short‐lived and hence cannot ensure consistent, long‐lasting immune regulatory functions both in vivo and in vitro 33. Moreover, the adult MSCs undergoes replicative senescence at a very early stage of proliferative cycle rendering it disadvantageous to use the cells for transplantation 3. Hence, for mainstream therapeutic applications alternate source of MSCs must be considered.

In this regard, human‐induced pluripotent stem cells (hiPSCs) reprogrammed from human adult somatic cells, converge to a better‐defined ground state of pluripotency. Human‐induced pluripotent stem cells can be differentiated into all three germ layer cell types (Ectodermal, Mesodermal and Endodermal) of the organism and – while in the pluripotent state – can be cultured virtually indefinitely without significant signs of replicative senescence. A recent breakthrough in the generation of hiPSCs from human somatic cells by using defined factors, 34, 35 could facilitate generation of patient‐specific iMSCs derived from hiPSCs. The iMSCs have the capabilities for utilization in a broad range of regenerative medical applications. Hence, they are often considered as readily accessible promising source of stem cells for future clinical therapies 3. The iMSCs shared the similar properties compared to the ESC‐derived MSCs 33. Recent studies have also revealed that biomimetic surface results in the rapid and efficient derivation of iMSCs from hiPSCs 36. However, several challenges need to be effectively tackled before iMSCs could be favourably used for translational applications.

Human pluripotent stem cells (hESCs & hiPSCs) as a novel cell resource for generating clinical‐grade products

The PSCs could serve as an alternate source for the generation of MSCs. Embryonic stem cells can be used as an efficient source to generate the MSCs almost indefinitely, attributed to tremendous proliferation potential of ESCs 37. Nonetheless, ethical concerns, allogenicity and immune reactivity proffer ESC‐derived MSCs unsuitable for clinical applications 30. On the other hand, efficient hiPSCs reprogramming methods could be successfully used to obtain patient‐specific iPSCs. Takahashi and Yamanaka were the first groups to demonstrate that mouse 32 and human 34 somatic cells could be successfully converted to iPSCs through the retroviral delivery of Oct4, Sox2, Klf4 and C‐Myc. Further characterization of iPSCs indicated human iPSCs are similar to human ESCs in their morphology, gene expression profile, in vitro differentiation potential and teratoma formation 30. Different types of human somatic cells have been successfully shown to reprogram into hiPSCs (Table 1).

Table 1.

Different types of human somatic cells that have been reprogrammed to induced pluripotent stem cells (hiPSCs)

Cell source References
Bone marrow MSCs 38
Adipose tissue‐derived stem cells 39
Cord blood cell 40
Keratinocytes 41
Skin fibroblasts 41
Mammary epithelial cells 42
Renal epithelial cells 43
Corneal epithelial cells 44
Peripheral blood cells 45
Umbilical cord MSCs 46
Placental MSCs 47
Amniotic membrane MSCs 46
Amniotic fluid‐derived cells 48

The discovery of hiPSCs has accelerated the regenerative medical research 19. Human‐induced PSCs are cells that have the capability of differentiating into all somatic cell derivatives (all three germ layers, for example, ectoderm, mesoderm and endoderm) and also, make a contribution to the germline (Table 2 shows a catalogue of different cell types derived from iPSCs); this unique ability of contribution to chimera and indefinite self‐renewal provides a unique opportunity for autologous personalized cell‐based therapy 34, 49, 50. Towards future studies, hiPSCs are considered as the driving force for personalized cell replacement therapy 51.

Table 2.

List of different cell types including iMSCs derived from hiPSCs

Cell types References
Ectoderm
Neural 52
Retinal pigment epithelial cells 53
Corneal epithelial cells 44
Mesoderm
Cardiomyocytes 54
Adipocytes 55
Osteocytes 56
Chondrocytes 57
iMSCs 58
Haematopoietic stem cells (HSCs) 59
Erythrocytes 60
Platelets 61
Endothelial cells 62
Neutrophils 63
Endoderm
Lung and airway epithelial cells 64
Nephrogenic intermediates 65
Follicular epithelial cells 66
Hepatocytes 67
Kidney progenitor cells 68, 69
Pancreatic beta cells 70
Germ cells 71, 72

Establishment of reliable and standardized source of functional MSCs for regenerative applications

The establishment of a reliable source of autologous, transgene‐free progenitor cells have enormous potential in the field of cell‐based regenerative medicine 3, 4, 30, 33, 34, 35, for example, in preparing a therapeutic strategy for infants born with devastating birth defects 73, 74, 75, 76, 77, 78, 79, 80, 81, 82. However, standardization of MSCs remains a major obstacle to the therapeutic usage in regenerative medicine 30, 31, 83. Comparison of experimental data with different studies becomes difficult when starting materials and culture conditions affects cell preparations 6, 8, 10, 16, 17, 18.

Recent studies using RNA‐based technology 84, pluripotency‐associated protein transfection 85, non‐integrating methods of the pluripotent gene containing plasmid usage 86 and a pluripotent gene containing Sendai viral vectors 87, 88 are hinting towards safe clinical usage of footprint‐free hiPSC‐derived cellular products, such as iMSCs, since these directed differentiated cells will not have any risk of undesired genomic modifications associated with reprogramming protocol.

iMSCs as a novel source of therapeutically active MSCs

The adult MSCs does not exhibit long‐lasting immunoregulatory functions in vitro and in vivo 10. The primary source of MSCs with high‐proliferation potential has been reported as a suitable alternative to the adult MSCs sources 33. The development of hiPSCs has, in turn, led to the culmination of the unique ability to generate iMSCs by directed differentiation (Table 3). Recent data suggest that iMSCs are emerging as a strong contender for the new sources of MSCs that could be suitable to replace the adult MSCs. Particularly, of late many studies have reported successful derivation of functional MSCs from iPSCs (iMSCs) 33, 36, 50, 58, 89, 90, 91, 92. The iMSCs are a novel class of stem cells that augments effective and reliable regeneration than contemporary methods. The iMSCs can be obtained from the readily accessible adult tissues and exhibit greater proliferation potential than the traditional sources of MSCs 58. Because of the promising pre‐clinical and clinical therapeutic potential of MSCs, the iMSCs derived from iPSCs may serve as an alternate and inexhaustible source 93. Additionally, the synthetic coating has been shown to assist in the derivation of iMSCs. The derivation of iPSCs into iMSCs on synthetic polymer coating, PMEDSAH [Poly [2‐(methacryloyloxy) ethyl dimethyl‐(3‐sulfopropyl) ammonium hydroxide] resulted in high differentiation efficiency, tri‐lineage differentiation potential and expression of characteristic MSCs markers (CD73+, CD90+, CD105+, CD166+, CD31, CD34 and CD45) 91. Similarly, Liu et al. has shown that iMSCs could be orderly derived in a single step from iPSCs on fibrillar collagen coating 36. In a recent study, Chen et al. has shown that treatment of iPSCs with SB431542, a transforming growth factor β pathway inhibitor to generate epithelial monolayer‐like cells in two‐dimensional (2D) culture system followed by induction of epithelial–mesenchymal transition lead to rapid and reliable differentiation into iMSCs 50. Overexpression of Oct4 along with the combination of GSK3 inhibitor has been demonstrated to reprogram CD34+ peripheral blood or cord blood into functional mesenchymal stromal cells 94.

Table 3.

Cell culture supplements that promote in vitro derivation of iMSCs from hiPSCs

Materials/Additives References
Synthetic polymer, PMEDSAH 91
Fibrillar collagen 36
SB431542, a TGF‐β pathway inhibitor 50
RGD (Arg‐Gly‐Asp) peptides 95
Fibronectin (Fn) 95
Fibronectin‐like engineered polymer protein (FEPP) 95
Extracellular matrix, Geltrex 95
Platelet concentrate 95
Oct4 94
CHIR99021, GSK inhibitor 94

Phenotypic features of iMSCs

The specific cell surface marker on the human MSCs remains to be properly elucidated. Currently, a panel of markers is used to characterize the MSCs isolated from different tissue sources, since there is no specific marker for identifying the bonafide MSCs. The iMSCs satisfies the essential criteria's such as plastic adherence, expression of key MSC surface markers and tri‐lineage differentiation capability properties as laid down by the International Society of Cellular Therapy 3. Himeno et al. have demonstrated that iMSCs from mice exhibited characteristic mice MSC surface marker such as CD105, CD140a, Sca‐1 and CD44 as previously described 19, 96. The immunosuppressive, cytoprotection and tissue regeneration properties are exerted by the paracrine factors secreted by the MSCs 97, 98, 99. The iMSCs and ESC‐derived MSCs displayed attenuation of proliferation and cytolytic activity of NK cells in a similar way to BMMSCs. The iMSCs offer vast superiority than traditional sources of MSCs, as they can be generated from any tissue source from the body and theoretically iPSCs pose unlimited growth potential. Thus, iMSCs should serve as an inexhaustible source of MSCs 3. The human MSCs from various tissue sources are typically identified by the expression CD29, CD44, CD73, CD90, CD105, CD146 and CD166. Newer studies have reported that human iMSCs exhibited the above indicated typical characteristics of adult MSCs 33.

Biological characteristics of iMSCs:

The iMSCs and ESC‐derived MSCs displayed similar strong immunosuppressive characteristics 33, also iMSCs display a wide range of cytokine profiles, microenvironment modulatory paracrine factors and exert different functions on the local cellular niche components via secretion of suitable bioactive molecules (Fig. 1). Giuliani et al. further reported that there was no marked functional variability between iMSCs and ESCs–MSCs 33. Unlike BMMSCs, iMSCs and ESCs–MSCs that could be subjected to long‐term culture without resulting in explicative senescence 33. Studies by Lian et al. have shown that iMSCs display typical MSC characteristics and there were no differences between human iMSCs and human ESC‐derived MSCs 58, 83. More robust proliferation was observed in iMSCs than BMMSCs. The iMSCs could be easily scaled up to more than 40 passages while stably maintaining normal diploid karyotype, and consistent gene expression and surface antigen profile 58. Human iMSCs apart from typical MSCs characteristic markers such as CD29, CD44 and CD73 also expressed a higher level of endogenous pluripotency markers such as Oct4 58. Liu has proposed that iMSCs derived from blood cells could be used as a novel and patient‐specific source for usage in disc repair 100. Comparative study of DNA methylation profiles of iMSCs with normal MSCs and PSCs suggested that iMSCs maintained donor‐derived epigenetic differences 101. In a recent study, published by Frobel et al. iMSCs are starter MSCs and subjected to epigenetic analysis. The study indicated that morphology, immunophenotype, in vitro differentiation and gene expression of iMSCs were consistent with the initial donor MSCs population. Except iMSCs were impaired in suppressing T‐cell proliferation. The iMSCs retained donor‐derived DNA methylation (DNAm) profile. However, tissue‐specific and age‐related DNAm profiles of iMSCs were completely erased. Further, the iMSCs reacquired senescence‐associated DNAm. The study also contrastingly highlights that iMSCs reacquire incomplete immunomodulatory functions 102.

Figure 1.

Figure 1

Intrinsic features of iMSCs, which may allow them to have better biological effectiveness compared to adult hMSCs. Multilineage differentiations may obtain a variety of specialized cells for cell replacement therapy (Table 2 lists different types of cells obtained from hiPSCs). Various cytokines in their secretome profile also help immunomodulation, antifibrotic, anti‐apoptotic activities. The microenvironment modulatory paracrine factors may exert a wide range of cellular functions on local cellular niche components via the release of the suitable bioactive compound.

Directed Differentiation of iMSCs

The use of stem cells and biologically suitable scaffolds offer the full potential for tissue regeneration. Transplantation of lineage‐committed cells can obviate in vivo teratoma formation that is caused by the rapid proliferation and uncontrolled spontaneous differentiation of PSCs. Thus, controlled differentiation of hiPSCs into cells that resemble adult MSCs is an attractive approach to obtain a readily available source of progenitor cells for tissue engineering. Unlike previously reported methods that typically rely on the addition of soluble factors to affect PSC differentiation, a recent study by Liu et al. reports an alternative approach using a biomaterial coating on a cell culture plate made of fibrillar collagen Type I to promote the derivation of MSC‐like cells. This study has reported a collagen matrix that could potentially play a positive role in regulating the differentiation of hESCs and hiPSCs towards a multipotent mesenchymal progenitor cell 36. Activation of epithelial‐to‐mesenchymal transition (EMT) of epithelial cells has been used successfully by others for generation of MSC‐like cells from hESCs 103. A study by ThienHan et al. to generate MSCs from human iPSCs, and investigate the osteogenic differentiation of iMSCs seeded on biofunctionalized CPCs containing RGD (Arg‐Gly‐Asp) peptides, fibronectin (Fn), Fibronectin‐like engineered polymer protein (FEPP), Geltrex and platelet concentrate has also been reported 80. A significant part of the study dedicated to the investigation of iMSCs proliferation and osteogenic differentiation atop calcium phosphate cement (CPC) containing biofunctional agents was also evaluated 80.

Therapeutic applications of iMSCs

The iMSCs could be effectively used for diseases modelling, drug screening and therapeutic applications (Fig. 1). The immunological concerns on cell therapy can be effectively bypassed by iMSCs 51. Nevertheless, the long‐term studies on the immunosuppressive activity of the iMSCs remain to be explored 33. Mesenchymal stem cells are considered as the first line of prophylactic treatment for GVHD and organ transplantation owing to their immunoregulatory properties 27, 104, 105, 106. During allogeneic transplantation, the circulating NK cells target and destroy the graft 107, 108. On the other hand cotransplantation of MSCs prevent GVHD by attenuating the cytotoxic activity of NK Cells 27, 105, 106. Under in vitro culture conditions Giuliani et al. has shown that human MSCs derived from the iPSCs considerably down‐regulated NK cell cytolytic capabilities 33. The iMSCs were more potent than the BMMSCs. Thus, iMSCs can be graded as a useful therapeutic option to prevent allograft rejection 33. The study from Himeno et al. showed MSCs from iPSCs ameliorated diabetic polyneuropathy (DNP) in mice 19. The results suggest that effects of DNP by MSCs might be because of the secretion of angiogenic/neurotrophic factors and differentiation into Schwann type cells. Mesenchymal stem cells have also been reportedly considered as a potential treatment option for periodontal defects arising from periodontitis. A report by Hynes et al. indicated that iMSCs facilitated the periodontal regeneration coupled with newly formed mineralized tissue in periodontitis rat models 3. Recently, Yang et al. demonstrated that tumour necrosis factor alpha‐stimulated gene‐6 (TSG6) expressing iMSCs were capable of decreasing the inflammation in experimental periodontitis model and inhibiting alveolar bone resorption 109. Human MSCs have emerged as a promising therapeutic source for treating myocardial and limb ischaemia 110, 111. An investigation by Lian et al. revealed that human iMSCs attenuate limb ischaemia in mice 58. Further analysis showed that transplantation of iMSCs into mice exhibited better attenuation in hindlimb ischaemia than adult BM‐MSCs. The greater therapeutic efficacy can be attributed to their ability to survive for a longer time after transplantation. Tracking of transplanted iMSCs divulged, iMSCs could engraft and survive for more than 5 weeks following transplantation 58. Wei et al. indicated human iMSCs could continuously proliferate for more than 32 passages without undergoing cellular senescence and displayed superior wound healing and pro‐angiogenic properties 92. The iMSCs derived on a synthetic polymeric coating; PMEDSAH resulted in novel bone formation when transplanted into the mice with calvarial defects 91. Zang et al. have shown that iMSCs derived from Hutchinson–Gilford Progeria syndrome (HGPS) were helpful in studying the molecular pathology of HGPS 89. In a recent study, Liu et al. has successfully utilized iMSCs for modelling Fanconi anaemia. The Fanconi anaemia iPSC‐derived MSCs displayed premature senescence 112. Also, hiPSC‐derived cells have been successfully used to model various other diseases. We have provided the comprehensive details below in Table 4, a list of disease modelling using hiPSCs (Fig. 2).

Table 4.

Disease modelling using hiPSCs

Disease modelling References
Neurological
Development Fragile X/ataxia syndrome (FXA) 74, 113
Rett syndrome (RS) 75
Angleman syndrome 76
Prader–Willi syndrome 76
Timothy syndrome (TS) 77
Microcephaly (MC) 78
Hereditary spastic paraplegias (HSP) 79
Olivopontocerebellar atrophy (OPCA) 80
Pelizaeus–Merzbacher disorder (PMD) 81
Mitochondrial encephalopathy with lactic acidosis and stroke‐like episodes (MELAS) 82
Glioblastoma iPSCs 114
Childhood cerebral adrenoleukodystrophy (CCALD) 115
Multiple sclerosis 116
Autism spectrum disorder (ASD) 117
Cernunnos deficiency syndrome (XLF) 118
William–Beuren syndrome (WBS) 119
William–Beuren region duplication syndrome (WBDS) 119
Degenerative Alzheimer's (AD) 120, 121, 122
Schizophrenia (SCZD) 123
Spinal muscular atrophy (SMA) 124
Parkinson disease (PD) 125, 126, 127
Huntington disease (HD) 125, 128
Amyotrophic lateral sclerosis (ALS) 129
Familial dysautonomia (FD) 130
X‐linked adrenoleukodystrophy (X‐ALD) 131
Machado–Joseph disease (MJD) 132
Friedreich's Ataxia (FRDA) 133
Familial transthyretin amyloidosis (ATTR) 134
Tauopathies (TAP) 135
Diabetic polyneuropathy (DPN) 19
Gaint axonal neuropathy (GAN) 136
Menkes disease (MD) 137
Frontotemporal dementia (FTD) 138, 139
Spinal cerebral ataxia type2 (SCA2) 140
Ataxia telangiectasia (AT) 141
Dravet syndrome (DVS) 142
Hematological Swachman–Bodian–Diamond syndrome (SBD) 125
Adenosine deaminase deficiency (ADA) severe combined immunodeficiency (SCID) 125
Fanconi anemia (FA) 143
Sickle cell anaemia (SCA) 144
Beta‐thalassaemia (BT) 145
Polycythaemia vera (PV) 146
Congenital amegakaryocytic thrombocytopenia (CAMT) 147
Paroxysmal nocturnal haemoglobinuria (PNH) 148
Dyskeratosis congenita (DC) 149
α‐Thalassaemia (AT) 150
Aplastic anaemia (AA) 151
Myeloproliferative disorder (MPN) 152
Chronic myeloid leukaemia (CML) 153
Juveline myelomonocytic leukaemia (JMML) 154
Chronic infantile neurological, cutaneous and articular syndrome (CINCA) 155
X‐linked chronic granulomatous disease (X‐CGD) 156
Severe congenital neutropaenia (SCN) 157
Wiskott–Aldrich syndrome (WAS) 158
Metabolic Gaucher disease type III (GD) 125
Juvenile diabetes mellitus (JDM) 125
Lesch–Nyhan syndrome (LNS) 125
Aplha1‐Antitrypsin deficiency (A1ATD) 159
Pompe disease (PomD) 160
Familial hypercholesterolaemia (FH) 161
Tyrosinaemia (TYS) 162
Glycogen storage disease type1 (GSD) 162
Progressive familial cholestasis (PFD) 162
Crigler–Najjar syndrome (CN) 162
Hurler syndrome (HS) 163
Neuronal ceroid lipofuscinosis (NCL) 164
Wilson's disease (WD) 165
Mitochondrial diabetes (MT) 166
Fabry disease (FD) 87
Mucopolysaccharidosis type IIIB disease (MPS) 167
Cardiovascular LEOPARD syndrome (LS) 78
Long QT syndrome type 1 (LQTS1) 168
Long QT syndrome type 2 (LQTS2) 169
Long QT syndrome type 3 (LQTS3) 170
Supervascular aortic stenosis (SVAS) 171
Hypertrophic cardiomyopathy (HCM) 172
Diabetic cardiomyopathy (DCM) 173
Hypoplastic left heart syndrome (HLHS) 174
Moyamoya disease (MMD) 175
Catecholaminergic polymorphic ventricular tachycardia (CPVT) 176
Familial dilated cardiomyopathy (DCM) 177
Familial hypertrophic cardiomyopathy (HCM) 178
Primary immunodeficiency SCID/Leaky SCID 179
Omenn syndrome (OS) 179
Cartilage–hair hypoplasia (CHH) 179
Herpes simplex encephalitis (HSE) 179
Musculoskeletal disorder Craniometaphyseal dysplasia (CMD) 88
Duchenne muscular dystrophy (DMD) 125
Becker muscular dystrophy (BMD) 125
Osteogenesis imperfect (OI) 180
Thanatophoric dysplasia (THD) 181
Achondroplasia (ACH) 181
Hutchinson–Gilford progeria syndrome (HGPS) 182
Werner syndrome (WS) 183
Facioscapulohumeral muscular dystrophy (FSHD) 184
Limb‐girdle muscular dystrophy (LGMD) 185
Myotonic dystrophy type 1 (MyD1) 186
Marfan syndrome (MFS) 187
Fibrodysplsia ossificans progressiva (FOP) 188
Lung disorder Cystic fibrosis (CF) 189
Pulmonary alveolar proteinosis (PAP) 190
Emphysema (EP) 191
Dermatological Disorder Recessive dystrophic epidermolysis bullosa (RDEB) 192, 193
Scleroderma (SC) 191
Focal dermal hypoplasia (FDH) 194
Hermansky–Pudlak syndrome (HPS) 195
Chediak–Higashi syndrome (CHS) 195
Cancer Breast cancer (BC) 196
Opthalmological disorder Retinitis pigmentosa (RP) 53, 197, 198
Gyrate atrophy (GA) 199
Best disease (BD) 200
Cataract (Cat) 201
Ectrodactyly‐ectodermal dysplasia‐cleft syndrome (EEC) 202
Nephrology End stage renal disease (ESRD) 203
Aneuploidy Turner syndrome (TS) 204
Warkany syndrome (WKS) 204
Patau syndrome (PS) 204
Emanuel syndrome (ES) 204
Klinefelter's syndrome (KS) 205
Down's syndrome 125

Figure 2.

Figure 2

Potential application overview of iMSCs derived from hiPSCs.

Recent developments in safe clinical products

Challenges & strategy to overcome them for Clinically Relevant iMSCs

Safety and Efficacy of iMSCs are of paramount importance to succeed in the field of translational regenerative medicine. The viral vector‐based strategy for reprogramming might result in tumour formation as a result of insertional mutagenesis of the transgene. C‐Myc is a proto‐oncogene that has been shown to increase the efficiency of reprogramming by suppressing the tumour suppressor p‐53 gene. The overexpression of proto‐oncogene and moderating of tumour suppressor genes render hiPSCs beneficial results regarding higher proliferative advantage for downstream translational applications 206. Consequently, many studies have unveiled several different strategies for generation of safer iPSCs. In 2010, Yamanaka's group suggested using L‐Myc as an alternative to C‐Myc for reprogramming based on the result that L‐Myc maintained in the reprogramming efficiency without inducing any tumorigenesis 206. Fang et al., reported generated iPSCs devoid of C‐Myc enervated retinal ischaemia and reperfusion injury following transplantation in rat models 207. The starter cell type has an enormous impact on reprogramming, differentiation and in vivo functionality because of epigenetic memory. Until now, there are no data on the best starter cell type for a particular clinical application. Hence, more research needs to be conducted to determine suitable starter cell type based on the type of clinical application 30. While iPSC‐derived cell source are emerging as a replacement cell source, their traits of self‐renewal and pluripotency after in vivo transplantation often leads to tumorigenicity and genomic instability might result in low clinical utility 3. The nature of pluripotency transgene elements present in the iMSCs is arduous to predict. Hence, iMSCs have been thoroughly characterized for silencing of the transgene expression and safe transgene integration 33. Presently, only initial studies are reported on preclinical applications of iMSCs. Hence, long‐term, multicentric, pre‐clinical and clinical studies are required for accurate prediction of iMSCs for the translational purpose 33. The recent development of non‐viral‐based generation of iPSCs might pave the way for considering iPSCs as a suitable candidate for biotherapeutics 86, 208, 209, 210. Newer technologies without viral transgene such as chemicals, plasmids and recombinant protein‐based approaches might augment the clinical utilization of these safe iPSCs 85, 211, 212. The low efficiency of iPSCs generation might be a serve‐debilitating factor to consider iPSCs/iMSCs for translational applications. Hence, more research needs to be focused on scaling and optimizing the quality of iPSCs 58.

Regulatory issues for future safe therapies using hiPSCs

The iPSCs present unique sets of technical and regulatory hurdles when compared to even ESCs for translational applications 213. The issues regarding the cell and gene therapy in every country are governed by its sovereign regulatory body. In the United States, the human iPSC products are regulated by Centre for Biologics Evaluation and Research at the United States Food and Drug Administration (USFDA) 214. Before proceeding with the clinical trials, the iPSC‐derived products are subjected to preclinical testing that requires extensive examination of safety, feasibility and efficacy 215. Pre‐clinical studies involve a comparative analysis of the various parameters between healthy animals and disease models. According to the FDA guidelines, the same cells used in preclinical trials should be used during clinical trials 215. Small animal models, such as rodents are used in preclinical studies. However, rodent models, although could be used for basic biological studies have a poor predictive outcome in term of clinical efficacy 215. Consequently, pre‐clinical studies consisting of large animal models such as swine, primates, etc., are favourable as they have relatively longer life span and displays physiological similarities to humans albeit a limited number of disease models and inability to modify the genome with ease constitute major road block in the usage of large animal models 215. For a particular disease condition, a single satisfactory model is not present. Hence, pre‐clinical testing must be carried out using suitable alternative models to highlight potential limitations and assist in finding suitable alternative avenues for handling the disorder 215. Necessary precautions must be undertaken before extrapolating the results from animal models to clinical trials 215.

Efficacy of the transplanted cells in vivo is not well documented. A few studies have demonstrated that transplantation of PSCs and differentiated cells resulted in poor survival of the cells 216, 217, 218, 219. The fate of the transplanted cells must be evaluated to ascertain the tangible effectiveness of the cells in vivo following transplantation. Hence, suitable surgical/imaging techniques should be developed for in vivo fate mapping of the cells 215.

Current good manufacturing protocol guidelines must be followed to generate and characterize iPSC‐based products 215 for any future clinical applications. The quality of cell products and homogeneity of the cell population will determine the effect, risk and potency of the iPSC‐based therapy 215. Method and duration of storage, viability, cell line contamination and risks of transmissible infections are some of the other possible confounding factors that can affect the cell therapy 215. Before scaling up towards clinical trials questions such as ideal cell source, efficient reprogramming and differentiation protocols, demonstration of safety and functionality have to be addressed 215.

Generation of iPSCs from somatic cells requires a significant amount of molecular manipulations 213 either by viral vectors containing reprogramming genes 34 or transfection of reprogramming mRNAs 84 or purified reprogramming factors 85 or transfection by non‐viral vectors containing reprogramming gene methods 86. The viral‐based reprogramming strategies form the basis of added concerns because of random integration into the host genome 34. On the contrary use of retroviral‐based genetically modified cells is technically permitted for human clinical trials under the existing National Institutes of Health (NIH), guidelines 213. Recent strategies for using small molecule‐based reprogramming & differentiation must be explored to develop and differentiate into clinical relevant cell types 220. Generation of iPSCs require a significant amount of manipulations 213. The viral‐based reprogramming strategies form the basis of added concerns because of random integration into the host genome 34. On the contrary, use of retroviral‐based genetically modified cells is technically permitted for human clinical trials under the existing NIH, guidelines 213. Recent strategies for using small molecule‐based reprogramming & differentiation must be explored to develop and differentiate into clinical relevant cell types 220. Every iPSC line would exhibit unique genetic and epigenetic constitution. Hence, each and every cell line has to be subjected to independent characterization to determine its precise characteristic features 213. It is necessary to determine to what extent iPSCs are similar to ESCs. Besides safety, efficacy, stability, heritability and absence of biased lineage differentiation have to examine and documented 213.

Immune response against transplanted cells presents a critical challenge that can detrimentally affect the outcome of therapy. Some of the important questions as to why the donor cells pose a risk of immune response or genetic diseases, cell efficiency, cells exhibiting risk of contamination, effectiveness and safety of transplanted cells has to be answered, if they are perceived to develop into potential therapeutic agents 221, 222, 223. Tumour formation remains one of the most important concerns while using the pluripotent cells or PSC‐derived products. It has been well documented that the reminiscent PSCs present in differentiated cells could effectively give rise to tumour formation 217, 219, 224, 225, 226, 227, 228.

One of the biggest advantages of iPSCs is the possibility of generation of patient‐specific autologous cell lines. Hence, the cumbersome procedure of screening against different cell lines for a proper match is excluded 34, 213, 229. The method of selection and characterization criteria needs good manufacturing protocol 213. The combination of a proven gene therapy with proven PSC‐derived products might hold a great potential for therapeutic application albeit certain technical and regulatory hurdles. Hence, suitable regulatory guidelines should be established for the application of genetically modified stem cells 213.

Conclusion

The invention of cellular reprogramming of adult cells from the terminally differentiated state of PSCs state with the help of transcription factors, biological factors and small molecules open up a large window of opportunity in the field of regenerative medicine. By incorporating the advantages of both iPSCs and MSCs, the resulting iMSCs are emerging as a novel stem cell population 3. The iMSCs generated from iPSCs successfully exhibited all the fundamental criteria for defining the MSC population based on the existing knowledge 3. Data indicate that iMSCs can be used as a promising alternative strategy for treatment of various immune‐mediated diseases 33. Although, the concept of iMSCs is at its nascent stage, recent studies nevertheless provide the proof of concept that functional iMSCs could be successfully generated from iPSCs that exhibit robust proliferation and differentiation potential, which could be used for tissue repair and engineering applications 58. The development of iMSCs offers promise of patient‐specific, cost‐efficient and batch to batch consistency 58. Presently, the scope of iMSCs is limited to the pre‐clinical utility for tissue engineering‐based treatment approaches. Further pre‐clinical and clinical studies are required before scaling it towards routine clinical utility.

Conflicts of interest

None.

Acknowledgements

Dr. Kumar contributed to the conception, design, data collection, assembly, analysis, interpretation, manuscript writing and final approval of the manuscript. Vikram Sabapathy contributed to the data collection, analysis, manuscript writing and final approval of the manuscript. We would like to thank Department of Biotechnology (DBT), India for Ramalingaswami Fellowship and research support grant (DBT Grant nos’. BT/PR8527/MED/31/234/2013, BT/PR8742/AGR/36/773/2013 and BT/PR15420/MED/31/122/2011) to Sanjay Kumar.

References

  • 1. English K, French A, Wood KJ. Mesenchymal stromal cells: facilitators of successful transplantation? Cell Stem Cell. 2010; 7: 431–42. [DOI] [PubMed] [Google Scholar]
  • 2. Sabapathy V, Sundaram B, Vm S, et al Human Wharton's Jelly Mesenchymal Stem Cells plasticity augments scar‐free skin wound healing with hair growth. PLoS ONE. 2014; 9: e93726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Hynes K, Menicanin D, Han J, et al Mesenchymal stem cells from iPS cells facilitate periodontal regeneration. J Dent Res. 2013; 92: 833–9. [DOI] [PubMed] [Google Scholar]
  • 4. Roobrouck VD, Ulloa‐Montoya F, Verfaillie CM. Self‐renewal and differentiation capacity of young and aged stem cells. Exp Cell Res. 2008; 314: 1937–44. [DOI] [PubMed] [Google Scholar]
  • 5. Xin Y, Wang Y, Zhang H, et al Aging adversely impacts biological properties of human bone marrow‐derived mesenchymal stem cells: implications for tissue engineering heart valve construction. Artif Organs. 2010; 34: 215–22. [DOI] [PubMed] [Google Scholar]
  • 6. Heeschen C, Lehmann R, Honold J, et al Profoundly reduced neovascularization capacity of bone marrow mononuclear cells derived from patients with chronic ischemic heart disease. Circulation. 2004; 109: 1615–22. [DOI] [PubMed] [Google Scholar]
  • 7. Kretlow JD, Jin Y‐Q, Liu W, et al Donor age and cell passage affects differentiation potential of murine bone marrow‐derived stem cells. BMC Cell Biol. 2008; 9: 60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Wagner W, Bork S, Horn P, et al Aging and replicative senescence have related effects on human stem and progenitor cells. PLoS ONE. 2009; 4: e5846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Pittenger MF, Mackay AM, Beck SC, et al Multilineage potential of adult human mesenchymal stem cells. Science. 1999; 284: 143–7. [DOI] [PubMed] [Google Scholar]
  • 10. Uccelli A, Moretta L, Pistoia V. Mesenchymal stem cells in health and disease. Nat Rev Immunol. 2008; 8: 726–36. [DOI] [PubMed] [Google Scholar]
  • 11. Campagnoli C, Roberts IA, Kumar S, et al Identification of mesenchymal stem/progenitor cells in human first‐trimester fetal blood, liver, and bone marrow. Blood. 2001; 98: 2396–402. [DOI] [PubMed] [Google Scholar]
  • 12. Götherström C, Ringdén O, Westgren M, et al Immunomodulatory effects of human foetal liver‐derived mesenchymal stem cells. Bone Marrow Transplant. 2003; 32: 265–72. [DOI] [PubMed] [Google Scholar]
  • 13. In ‘t Anker PS, Noort WA, Scherjon SA, et al Mesenchymal stem cells in human second‐trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential. Haematologica. 2003; 88: 845–52. [PubMed] [Google Scholar]
  • 14. In ‘t Anker PS, Scherjon SA, Kleijburg‐van der Keur C, et al Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004; 22: 1338–45. [DOI] [PubMed] [Google Scholar]
  • 15. Li CD, Zhang WY, Li HL, et al Mesenchymal stem cells derived from human placenta suppress allogeneic umbilical cord blood lymphocyte proliferation. Cell Res. 2005; 15: 539–47. [DOI] [PubMed] [Google Scholar]
  • 16. Barberi T, Willis LM, Socci ND, et al Derivation of multipotent mesenchymal precursors from human embryonic stem cells. PLoS Med. 2005; 2: e161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Choo A, Lim SK. Derivation of mesenchymal stem cells from human embryonic stem cells. Methods Mol Biol. 2011; 690: 175–82. [DOI] [PubMed] [Google Scholar]
  • 18. Olivier EN, Rybicki AC, Bouhassira EE. Differentiation of human embryonic stem cells into bipotent mesenchymal stem cells. Stem Cells. 2006; 24: 1914–22. [DOI] [PubMed] [Google Scholar]
  • 19. Himeno T, Kamiya H, Naruse K, et al Mesenchymal stem cell‐like cells derived from mouse induced pluripotent stem cells ameliorate diabetic polyneuropathy in mice. Biomed Res. Int. 2013; 2013: 259187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Pacini S. Deterministic and stochastic approaches in the clinical application of mesenchymal stromal cells (MSCs). Front. Cell Dev. Biol. 2014; 2: 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Parekkadan B, Milwid JM. Mesenchymal stem cells as therapeutics. Annu Rev Biomed Eng. 2010; 12: 87–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Friedenstein AJ, Chailakhyan RK, Latsinik NV, et al Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo . Transplantation. 1974; 17: 331–40. [DOI] [PubMed] [Google Scholar]
  • 23. Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991; 9: 641–5. [DOI] [PubMed] [Google Scholar]
  • 24. Owen M. Marrow stromal stem cells. J Cell Sci Suppl. 1988; 10: 63–76. [DOI] [PubMed] [Google Scholar]
  • 25. Bauer G, Dao MA, Case SS, et al In vivo biosafety model to assess the risk of adverse events from retroviral and lentiviral vectors. Mol Ther. 2008; 16: 1308–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Aggarwal S, Pittenger MF. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 2005; 105: 1815–22. [DOI] [PubMed] [Google Scholar]
  • 27. Le Blanc K, Frassoni F, Ball L, et al Mesenchymal stem cells for treatment of steroid‐resistant, severe, acute graft‐versus‐host disease: a phase II study. Lancet. 2008; 371: 1579–86. [DOI] [PubMed] [Google Scholar]
  • 28. Murphy MB, Moncivais K, Caplan AI. Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med. 2013; 45: e54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Wang S, Qu X, Zhao RC. Clinical applications of mesenchymal stem cells. J. Hematol. Oncol. 2012; 5: 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Jung Y, Bauer G, Nolta JA. Concise review: induced pluripotent stem cell‐derived mesenchymal stem cells: progress toward safe clinical products. Stem Cells. 2012; 30: 42–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Meyerrose T, Olson S, Pontow S, et al Mesenchymal stem cells for the sustained in vivo delivery of bioactive factors. Adv Drug Deliv Rev. 2010; 62: 1167–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. 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] [PubMed] [Google Scholar]
  • 33. Giuliani M, Oudrhiri N, Noman ZM, et al Human mesenchymal stem cells derived from induced pluripotent stem cells down‐regulate NK‐cell cytolytic machinery. Blood. 2011; 118: 3254–62. [DOI] [PubMed] [Google Scholar]
  • 34. Takahashi K, Tanabe K, Ohnuki M, et al Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007; 131: 861–72. [DOI] [PubMed] [Google Scholar]
  • 35. Yu J, Vodyanik MA, Smuga‐Otto K, et al Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007; 318: 1917–20. [DOI] [PubMed] [Google Scholar]
  • 36. Liu Y, Goldberg AJ, Dennis JE, et al One‐step derivation of mesenchymal stem cell (MSC)‐like cells from human pluripotent stem cells on a fibrillar collagen coating. PLoS ONE. 2012; 7: e33225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Thomson JA, Itskovitz‐Eldor J, Shapiro SS, et al Embryonic stem cell lines derived from human blastocysts. Science. 1998; 282: 1145–7. [DOI] [PubMed] [Google Scholar]
  • 38. Zhao Q, Gregory CA, Lee RH, et al MSCs derived from iPSCs with a modified protocol are tumor‐tropic but have much less potential to promote tumors than bone marrow MSCs. Proc Natl Acad Sci USA. 2015; 112: 530–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Aoki T, Ohnishi H, Oda Y, et al Generation of induced pluripotent stem cells from human adipose‐derived stem cells without c‐MYC. Tissue Eng Part A. 2010; 16: 2197–206. [DOI] [PubMed] [Google Scholar]
  • 40. Meng X, Neises A, Su R‐J, et al Efficient reprogramming of human cord blood CD34+ cells into induced pluripotent stem cells with OCT4 and SOX2 alone. Mol Ther. 2012; 20: 408–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Streckfuss‐Bömeke K, Wolf F, Azizian A, et al Comparative study of human‐induced pluripotent stem cells derived from bone marrow cells, hair keratinocytes, and skin fibroblasts. Eur Heart J. 2013; 34: 2618–29. [DOI] [PubMed] [Google Scholar]
  • 42. Li Y, Hong N, Zhang A, et al Enhancing mammary differentiation by overcoming lineage‐specific epigenetic modification and signature gene expression of fibroblast‐derived iPSCs. Cell Death Dis. 2014; 5: e1550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Zhou T, Benda C, Dunzinger S, et al Generation of human induced pluripotent stem cells from urine samples. Nat Protoc. 2012; 7: 2080–9. [DOI] [PubMed] [Google Scholar]
  • 44. Mikhailova A, Ilmarinen T, Uusitalo H, et al Small‐molecule induction promotes corneal epithelial cell differentiation from human induced pluripotent stem cells. Stem Cell Reports. 2014; 2: 219–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Sommer AG, Rozelle SS, Sullivan S, et al Generation of human induced pluripotent stem cells from peripheral blood using the STEMCCA lentiviral vector. J Vis Exp. 2012; 68: e4327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Cai J, Li W, Su H, et al Generation of human induced pluripotent stem cells from umbilical cord matrix and amniotic membrane mesenchymal cells. J Biol Chem. 2010; 285: 11227–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Jiang G, Di Bernardo J, Delong CJ, et al Induced pluripotent stem cells from human placental chorion for perinatal tissue engineering applications. Tissue Eng Part C Methods. 2014; 20: 731–40. [DOI] [PubMed] [Google Scholar]
  • 48. Li C, Zhou J, Shi G, et al Pluripotency can be rapidly and efficiently induced in human amniotic fluid‐derived cells. Hum Mol Genet. 2009; 18: 4340–9. [DOI] [PubMed] [Google Scholar]
  • 49. Awaya T, Kato T, Mizuno Y, et al Selective development of myogenic mesenchymal cells from human embryonic and induced pluripotent stem cells. PLoS ONE. 2012; 7: e51638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Chen YS, Pelekanos RA, Ellis RL, et al Small molecule mesengenic induction of human induced pluripotent stem cells to generate mesenchymal stem/stromal cells. Stem Cells Transl Med. 2012; 1: 83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Amabile G, Meissner A. Induced pluripotent stem cells: current progress and potential for regenerative medicine. Trends Mol Med. 2009; 15: 59–68. [DOI] [PubMed] [Google Scholar]
  • 52. Hu B‐Y, Weick JP, Yu J, et al Neural differentiation of human induced pluripotent stem cells follows developmental principles but with variable potency. Proc Natl Acad Sci USA. 2010; 107: 4335–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Okamoto S, Takahashi M. Induction of retinal pigment epithelial cells from monkey iPS cells. Invest Ophthalmol Vis Sci. 2011; 52: 8785–90. [DOI] [PubMed] [Google Scholar]
  • 54. Hoekstra M, Mummery CL, Wilde AAM, et al Induced pluripotent stem cell derived cardiomyocytes as models for cardiac arrhythmias. Front Physiol. 2012; 3: 346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Elefanty AG, Stanley EG. Efficient generation of adipocytes in a dish. Nat Cell Biol. 2012; 14: 126–7. [DOI] [PubMed] [Google Scholar]
  • 56. Phillips MD, Kuznetsov SA, Cherman N, et al Directed differentiation of human induced pluripotent stem cells toward bone and cartilage: in vitro versus in vivo assays. Stem Cells Transl Med. 2014; 3: 867–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Wei Y, Zeng W, Wan R, et al Chondrogenic differentiation of induced pluripotent stem cells from osteoarthritic chondrocytes in alginate matrix. Eur. Cell. Mater. 2012; 23: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Lian Q, Zhang Y, Zhang J, et al Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation. 2010; 121: 1113–23. [DOI] [PubMed] [Google Scholar]
  • 59. Suzuki N, Yamazaki S, Yamaguchi T, et al Generation of engraftable hematopoietic stem cells from induced pluripotent stem cells by way of teratoma formation. Mol Ther. 2013; 21: 1424–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Chang C‐J, Mitra K, Koya M, et al Production of embryonic and fetal‐like red blood cells from human induced pluripotent stem cells. PLoS ONE. 2011; 6: e25761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Takayama N, Nishimura S, Nakamura S, et al Transient activation of c‐MYC expression is critical for efficient platelet generation from human induced pluripotent stem cells. J Exp Med. 2010; 207: 2817–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Adams WJ, Zhang Y, Cloutier J, et al Functional vascular endothelium derived from human induced pluripotent stem cells. Stem Cell Reports. 2013; 1: 105–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Sweeney CL, Merling RK, Choi U, et al Generation of functionally mature neutrophils from induced pluripotent stem cells. Methods Mol Biol. 2014; 1124: 189–206. [DOI] [PubMed] [Google Scholar]
  • 64. Huang SXL, Islam MN, O'Neill J, et al Efficient generation of lung and airway epithelial cells from human pluripotent stem cells. Nat Biotechnol. 2014; 32: 84–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Araoka T, Mae S‐I, Kurose Y, et al Efficient and rapid induction of human iPSCs/ESCs into nephrogenic intermediate mesoderm using small molecule‐based differentiation methods. PLoS ONE. 2014; 9: e84881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Yang R, Zheng Y, Burrows M, et al Generation of folliculogenic human epithelial stem cells from induced pluripotent stem cells. Nat Commun. 2014; 5: 3071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Chen Y‐F, Tseng C‐Y, Wang H‐W, et al Rapid generation of mature hepatocyte‐like cells from human induced pluripotent stem cells by an efficient three‐step protocol. Hepatology. 2012; 55: 1193–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Xia Y, Nivet E, Sancho‐Martinez I, et al Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor‐like cells. Nat Cell Biol. 2013; 15: 1507–15. [DOI] [PubMed] [Google Scholar]
  • 69. Takasato M, Er PX, Becroft M, et al Directing human embryonic stem cell differentiation towards a renal lineage generates a self‐organizing kidney. Nat Cell Biol. 2014; 16: 118–26. [DOI] [PubMed] [Google Scholar]
  • 70. Hua H, Shang L, Martinez H, et al iPSC‐derived β cells model diabetes due to glucokinase deficiency. J. Clin. Invest. 2013; 123: 3146–53. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 71. Panula S, Medrano JV, Kee K, et al Human germ cell differentiation from fetal‐ and adult‐derived induced pluripotent stem cells. Hum Mol Genet. 2011; 20: 752–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Irie N, Weinberger L, Tang WWC, et al SOX17 is a critical specifier of human primordial germ cell fate. Cell. 2015; 160: 253–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Urbach A, Bar‐Nur O, Daley GQ, et al Differential modeling of fragile X syndrome by human embryonic stem cells and induced pluripotent stem cells. Cell Stem Cell. 2010; 6: 407–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Sheridan SD, Theriault KM, Reis SA, et al Epigenetic characterization of the FMR1 gene and aberrant neurodevelopment in human induced pluripotent stem cell models of fragile X syndrome. PLoS ONE. 2011; 6: e26203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Marchetto MCN, Carromeu C, Acab A, et al A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell. 2010; 143: 527–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Chamberlain SJ, Chen P‐F, Ng KY, et al Induced pluripotent stem cell models of the genomic imprinting disorders Angelman and Prader‐Willi syndromes. Proc Natl Acad Sci USA. 2010; 107: 17668–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Paşca SP, Portmann T, Voineagu I, et al Using iPSC‐derived neurons to uncover cellular phenotypes associated with Timothy syndrome. Nat Med. 2011; 17: 1657–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Lancaster MA, Renner M, Martin C‐A, et al Cerebral organoids model human brain development and microcephaly. Nature. 2013; 501: 373–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Havlicek S, Kohl Z, Mishra HK, et al Gene dosage dependent rescue of HSP neurite defects in SPG4 patients’ neurons. Hum Mol Genet. 2014; 23: 2527–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Luo Y, Fan Y, Zhou B, et al Generation of induced pluripotent stem cells from skin fibroblasts of a patient with olivopontocerebellar atrophy. Tohoku J Exp Med. 2012; 226: 151–9. [DOI] [PubMed] [Google Scholar]
  • 81. Shimojima K, Inoue T, Imai Y, et al Reduced PLP1 expression in induced pluripotent stem cells derived from a Pelizaeus‐Merzbacher disease patient with a partial PLP1 duplication. J Hum Genet. 2012; 57: 580–6. [DOI] [PubMed] [Google Scholar]
  • 82. Folmes CDL, Martinez‐Fernandez A, Perales‐Clemente E, et al Disease‐causing mitochondrial heteroplasmy segregated within induced pluripotent stem cell clones derived from a patient with MELAS. Stem Cells. 2013; 31: 1298–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Lian Q, Lye E, Suan Yeo K, et al Derivation of clinically compliant MSCs from CD105+, CD24 differentiated human ESCs. Stem Cells. 2007; 25: 425–36. [DOI] [PubMed] [Google Scholar]
  • 84. Warren L, Manos PD, Ahfeldt T, et al Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 2010; 7: 618–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Kim D, Kim C‐H, Moon J‐I, et al Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell. 2009; 4: 472–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Si‐Tayeb K, Noto FK, Sepac A, et al Generation of human induced pluripotent stem cells by simple transient transfection of plasmid DNA encoding reprogramming factors. BMC Dev Biol. 2010; 10: 81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Kawagoe S, Higuchi T, Otaka M, et al Morphological features of iPS cells generated from Fabry disease skin fibroblasts using Sendai virus vector (SeVdp). Mol Genet Metab. 2013; 109: 386–9. [DOI] [PubMed] [Google Scholar]
  • 88. Chen I‐P, 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] [PMC free article] [PubMed] [Google Scholar]
  • 89. Zhang J, Lian Q, Zhu G, et al A human iPSC model of Hutchinson Gilford Progeria reveals vascular smooth muscle and mesenchymal stem cell defects. Cell Stem Cell. 2011; 8: 31–45. [DOI] [PubMed] [Google Scholar]
  • 90. Guzzo RM, Gibson J, Xu R‐H, et al Efficient differentiation of human iPSC‐derived mesenchymal stem cells to chondroprogenitor cells. J Cell Biochem. 2013; 114: 480–90. [DOI] [PubMed] [Google Scholar]
  • 91. Villa‐Diaz LG, Brown SE, Liu Y, et al Derivation of mesenchymal stem cells from human induced pluripotent stem cells cultured on synthetic substrates. Stem Cells. 2012; 30: 1174–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Wei H, Tan G, Manasi, et al One‐step derivation of cardiomyocytes and mesenchymal stem cells from human pluripotent stem cells. Stem Cell Res. 2012; 9: 87–100. [DOI] [PubMed] [Google Scholar]
  • 93. Eberle I, Moslem M, Henschler R, et al Engineered MSCs from patient‐specific iPS cells. Adv Biochem Eng Biotechnol. 2013; 130: 1–17. [DOI] [PubMed] [Google Scholar]
  • 94. Meng X, Su R‐J, Baylink DJ, et al Rapid and efficient reprogramming of human fetal and adult blood CD34+ cells into mesenchymal stem cells with a single factor. Cell Res. 2013; 23: 658–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. TheinHan W, Liu J, Tang M, et al Induced pluripotent stem cell‐derived mesenchymal stem cell seeding on biofunctionalized calcium phosphate cements. Bone Res. 2013; 4: 371–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Houlihan DD, Mabuchi Y, Morikawa S, et al Isolation of mouse mesenchymal stem cells on the basis of expression of Sca‐1 and PDGFR‐α. Nat Protoc. 2012; 7: 2103–11. [DOI] [PubMed] [Google Scholar]
  • 97. Hare JM, Fishman JE, Gerstenblith G, et al Comparison of allogeneic vs autologous bone marrow–derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012; 308: 2369–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Chou S‐H, Lin S‐Z, Day CH, et al Mesenchymal stem cell insights: prospects in hematological transplantation. Cell Transplant. 2013; 22: 711–21. [DOI] [PubMed] [Google Scholar]
  • 99. Zhang Y, Liao S, Yang M, et al Improved cell survival and paracrine capacity of human embryonic stem cell‐derived mesenchymal stem cells promote therapeutic potential for pulmonary arterial hypertension. Cell Transplant. 2012; 21: 2225–39. [DOI] [PubMed] [Google Scholar]
  • 100. Liu T. iPSC‐derived mesenchymal stem cells from blood cells as a new source for disc repair. Glob. Spine J. 2014; 04: po.073. [Google Scholar]
  • 101. Shao K, Koch C, Gupta MK, et al Induced pluripotent mesenchymal stromal cell clones retain donor‐derived differences in DNA methylation profiles. Mol Ther. 2013; 21: 240–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Frobel J, Hemeda H, Lenz M, et al Epigenetic rejuvenation of mesenchymal stromal cells derived from induced pluripotent stem cells. Stem Cell Reports. 2014; 3: 414–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Boyd NL, Robbins KR, Dhara SK, et al Human embryonic stem cell‐derived mesoderm‐like epithelium transitions to mesenchymal progenitor cells. Tissue Eng Part A. 2009; 15: 1897–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Bartholomew A, Sturgeon C, Siatskas M, et al Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo . Exp Hematol. 2002; 30: 42–8. [DOI] [PubMed] [Google Scholar]
  • 105. Koç ON, Gerson SL, Cooper BW, et al Rapid hematopoietic recovery after coinfusion of autologous‐blood stem cells and culture‐expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high‐dose chemotherapy. J Clin Oncol. 2000; 18: 307–16. [DOI] [PubMed] [Google Scholar]
  • 106. Ringdén O, Uzunel M, Rasmusson I, et al Mesenchymal stem cells for treatment of therapy‐resistant graft‐versus‐host disease. Transplantation. 2006; 81: 1390–7. [DOI] [PubMed] [Google Scholar]
  • 107. Kroemer A, Edtinger K, Li XC. The innate natural killer cells in transplant rejection and tolerance induction. Curr. Opin. Organ Transplant. 2008; 13: 339–43. [DOI] [PubMed] [Google Scholar]
  • 108. Ljunggren HG, Kärre K. In search of the “missing self”: MHC molecules and NK cell recognition. Immunol Today. 1990; 11: 237–44. [DOI] [PubMed] [Google Scholar]
  • 109. Yang H, Aprecio RM, Zhou X, et al Therapeutic effect of TSG‐6 engineered iPSC‐derived MSCs on experimental periodontitis in rats: a pilot study. PLoS ONE. 2014; 9: e100285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Kim S‐W, Han H, Chae G‐T, et al Successful stem cell therapy using umbilical cord blood‐derived multipotent stem cells for Buerger's disease and ischemic limb disease animal model. Stem Cells. 2006; 24: 1620–6. [DOI] [PubMed] [Google Scholar]
  • 111. Trivedi P, Tray N, Nguyen T, et al Mesenchymal stem cell therapy for treatment of cardiovascular disease: helping people sooner or later. Stem Cells Dev. 2010; 19: 1109–20. [DOI] [PubMed] [Google Scholar]
  • 112. Liu G‐H, Suzuki K, Li M, et al Modelling Fanconi anemia pathogenesis and therapeutics using integration‐free patient‐derived iPSCs. Nat Commun. 2014; 5: 4330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Urbach A, Bar‐Nur O, Daley GQ, et al Differential modeling of fragile X syndrome by human embryonic stem cells and induced pluripotent stem cells. Cell Stem Cell. 2010; 6: 407–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Stricker SH, Feber A, Engström PG, et al Widespread resetting of DNA methylation in glioblastoma‐initiating cells suppresses malignant cellular behavior in a lineage‐dependent manner. Genes Dev. 2013; 27: 654–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Wang X‐M, Yik WY, Zhang P, et al The gene expression profiles of induced pluripotent stem cells from individuals with childhood cerebral adrenoleukodystrophy are consistent with proposed mechanisms of pathogenesis. Stem Cell Res Ther. 2012; 3: 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Song B, Sun G, Herszfeld D, et al Neural differentiation of patient specific iPS cells as a novel approach to study the pathophysiology of multiple sclerosis. Stem Cell Res. 2012; 8: 259–73. [DOI] [PubMed] [Google Scholar]
  • 117. DeRosa BA, Van Baaren JM, Dubey GK, et al Derivation of autism spectrum disorder‐specific induced pluripotent stem cells from peripheral blood mononuclear cells. Neurosci Lett. 2012; 516: 9–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Tilgner K, Neganova I, Singhapol C, et al Brief report: a human induced pluripotent stem cell model of cernunnos deficiency reveals an important role for XLF in the survival of the primitive hematopoietic progenitors. Stem Cells. 2013; 31: 2015–23. [DOI] [PubMed] [Google Scholar]
  • 119. Adamo A, Atashpaz S, Germain P‐L, et al 7q11.23 dosage‐dependent dysregulation in human pluripotent stem cells affects transcriptional programs in disease‐relevant lineages. Nat Genet. 2014; 47: 132–41. [DOI] [PubMed] [Google Scholar]
  • 120. Israel MA, Yuan SH, Bardy C, et al Probing sporadic and familial Alzheimer's disease using induced pluripotent stem cells. Nature. 2012; 482: 216–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Yagi T, Ito D, Okada Y, et al Modeling familial Alzheimer's disease with induced pluripotent stem cells. Hum Mol Genet. 2011; 20: 4530–9. [DOI] [PubMed] [Google Scholar]
  • 122. Yahata N, Asai M, Kitaoka S, et al Anti‐Aβ drug screening platform using human iPS cell‐derived neurons for the treatment of Alzheimer's disease. PLoS ONE. 2011; 6: e25788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Brennand KJ, Simone A, Jou J, et al Modelling schizophrenia using human induced pluripotent stem cells. Nature. 2011; 473: 221–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Ebert AD, Yu J, Rose FF, et al Induced pluripotent stem cells from a spinal muscular atrophy patient. Nature. 2009; 457: 277–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Park I‐H, Arora N, Huo H, et al Disease‐specific induced pluripotent stem cells. Cell. 2008; 134: 877–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Devine MJ, Ryten M, Vodicka P, et al Parkinson's disease induced pluripotent stem cells with triplication of the α‐synuclein locus. Nat Commun. 2011; 2: 440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Soldner F, Hockemeyer D, Beard C, et al Parkinson's disease patient‐derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009; 136: 964–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. An MC, Zhang N, Scott G, et al Genetic correction of Huntington's disease phenotypes in induced pluripotent stem cells. Cell Stem Cell. 2012; 11: 253–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Dimos JT, Rodolfa KT, Niakan KK, et al Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science. 2008; 321: 1218–21. [DOI] [PubMed] [Google Scholar]
  • 130. Lee G, Papapetrou EP, Kim H, et al Modelling pathogenesis and treatment of familial dysautonomia using patient‐specific iPSCs. Nature. 2009; 461: 402–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Jang J, Kang H‐C, Kim H‐S, et al Induced pluripotent stem cell models from X‐linked adrenoleukodystrophy patients. Ann Neurol. 2011; 70: 402–9. [DOI] [PubMed] [Google Scholar]
  • 132. Koch P, Breuer P, Peitz M, et al Excitation‐induced ataxin‐3 aggregation in neurons from patients with Machado‐Joseph disease. Nature. 2011; 480: 543–6. [DOI] [PubMed] [Google Scholar]
  • 133. Ku S, Soragni E, Campau E, et al Friedreich's ataxia induced pluripotent stem cells model intergenerational GAA·TTC triplet repeat instability. Cell Stem Cell. 2010; 7: 631–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Leung A, Nah SK, Reid W, et al Induced pluripotent stem cell modeling of multisystemic, hereditary transthyretin amyloidosis. Stem Cell Reports. 2013; 1: 451–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Fong H, Wang C, Knoferle J, et al Genetic correction of tauopathy phenotypes in neurons derived from human induced pluripotent stem cells. Stem Cell Reports. 2013; 1: 226–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Johnson‐Kerner BL, Ahmad FS, Diaz AG, et al Intermediate filament protein accumulation in motor neurons derived from giant axonal neuropathy iPSCs rescued by restoration of gigaxonin. Hum Mol Genet. 2015; 24: 1420–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Suh J‐H, Kim D, Kim H, et al Modeling of Menkes disease via human induced pluripotent stem cells. Biochem Biophys Res Commun. 2014; 444: 311–8. [DOI] [PubMed] [Google Scholar]
  • 138. Almeida S, Zhang Z, Coppola G, et al Induced pluripotent stem cell models of progranulin‐deficient frontotemporal dementia uncover specific reversible neuronal defects. Cell Rep. 2012; 2: 789–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Almeida S, Gascon E, Tran H, et al Modeling key pathological features of frontotemporal dementia with C9ORF72 repeat expansion in iPSC‐derived human neurons. Acta Neuropathol. 2013; 126: 385–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Xia G, Santostefano K, Hamazaki T, et al Generation of human‐induced pluripotent stem cells to model spinocerebellar ataxia type 2 in vitro . J Mol Neurosci. 2013; 51: 237–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Nayler S, Gatei M, Kozlov S, et al Induced pluripotent stem cells from ataxia‐telangiectasia recapitulate the cellular phenotype. Stem Cells Transl Med. 2012; 1: 523–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Higurashi N, Uchida T, Lossin C, et al A human Dravet syndrome model from patient induced pluripotent stem cells. Mol. Brain. 2013; 6: 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Müller LUW, Milsom MD, Harris CE, et al Overcoming reprogramming resistance of Fanconi anemia cells. Blood. 2012; 119: 5449–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Sebastiano V, Maeder ML, Angstman JF, et al In situ genetic correction of the sickle cell anemia mutation in human induced pluripotent stem cells using engineered zinc finger nucleases. Stem Cells. 2011; 29: 1717–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Wang Y, Zheng C‐G, Jiang Y, et al Genetic correction of β‐thalassemia patient‐specific iPS cells and its use in improving hemoglobin production in irradiated SCID mice. Cell Res. 2012; 22: 637–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Ye Z, Zhan H, Mali P, et al Human‐induced pluripotent stem cells from blood cells of healthy donors and patients with acquired blood disorders. Blood. 2009; 114: 5473–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Hirata S, Takayama N, Jono‐Ohnishi R, et al Congenital amegakaryocytic thrombocytopenia iPS cells exhibit defective MPL‐mediated signaling. J. Clin. Invest. 2013; 123: 3802–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Yuan X, Braunstein EM, Ye Z, et al Generation of glycosylphosphatidylinositol anchor protein‐deficient blood cells from human induced pluripotent stem cells. Stem Cells Transl Med. 2013; 2: 819–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Batista LFZ, Pech MF, Zhong FL, et al Telomere shortening and loss of self‐renewal in dyskeratosis congenita induced pluripotent stem cells. Nature. 2011; 474: 399–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Chang C‐J, Bouhassira EE. Zinc‐finger nuclease‐mediated correction of α‐thalassemia in iPS cells. Blood. 2012; 120: 3906–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Winkler T, Hong SG, Decker JE, et al Defective telomere elongation and hematopoiesis from telomerase‐mutant aplastic anemia iPSCs. J. Clin. Invest. 2013; 123: 1952–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Saliba J, Hamidi S, Lenglet G, et al Heterozygous and homozygous JAK2(V617F) states modeled by induced pluripotent stem cells from myeloproliferative neoplasm patients. PLoS ONE. 2013; 8: e74257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Kumano K, Arai S, Hosoi M, et al Generation of induced pluripotent stem cells from primary chronic myelogenous leukemia patient samples. Blood. 2012; 119: 6234–42. [DOI] [PubMed] [Google Scholar]
  • 154. 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] [PMC free article] [PubMed] [Google Scholar]
  • 155. Tanaka T, Takahashi K, Yamane M, et al Induced pluripotent stem cells from CINCA syndrome patients as a model for dissecting somatic mosaicism and drug discovery. Blood. 2012; 120: 1299–308. [DOI] [PubMed] [Google Scholar]
  • 156. Zou J, Sweeney CL, Chou B‐K, et al Oxidase‐deficient neutrophils from X‐linked chronic granulomatous disease iPS cells: functional correction by zinc finger nuclease‐mediated safe harbor targeting. Blood. 2011; 117: 5561–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Morishima T, Watanabe K, Niwa A, et al Genetic correction of HAX1 in induced pluripotent stem cells from a patient with severe congenital neutropenia improves defective granulopoiesis. Haematologica. 2014; 99: 19–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Ingrungruanglert P, Amarinthnukrowh P, Rungsiwiwut R, et al Wiskott‐Aldrich syndrome iPS cells produce megakaryocytes with defects in cytoskeletal rearrangement and proplatelet formation. Thromb Haemost. 2015; 113: 792–805. [DOI] [PubMed] [Google Scholar]
  • 159. Yusa K, Rashid ST, Strick‐Marchand H, et al Targeted gene correction of α1‐antitrypsin deficiency in induced pluripotent stem cells. Nature. 2011; 478: 391–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Huang H‐P, Chen P‐H, Hwu W‐L, et al Human Pompe disease‐induced pluripotent stem cells for pathogenesis modeling, drug testing and disease marker identification. Hum Mol Genet. 2011; 20: 4851–64. [DOI] [PubMed] [Google Scholar]
  • 161. Cayo MA, Cai J, DeLaForest A, et al JD induced pluripotent stem cell‐derived hepatocytes faithfully recapitulate the pathophysiology of familial hypercholesterolemia. Hepatology. 2012; 56: 2163–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Ghodsizadeh A, Taei A, Totonchi M, et al Generation of liver disease‐specific induced pluripotent stem cells along with efficient differentiation to functional hepatocyte‐like cells. Stem Cell Rev. 2010; 6: 622–32. [DOI] [PubMed] [Google Scholar]
  • 163. Tolar J, Park I‐H, Xia L, et al Hematopoietic differentiation of induced pluripotent stem cells from patients with mucopolysaccharidosis type I (Hurler syndrome). Blood. 2011; 117: 839–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Lojewski X, Staropoli JF, Biswas‐Legrand S, et al Human iPSC models of neuronal ceroid lipofuscinosis capture distinct effects of TPP1 and CLN3 mutations on the endocytic pathway. Hum Mol Genet. 2014; 23: 2005–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Zhang S, Chen S, Li W, et al Rescue of ATP7B function in hepatocyte‐like cells from Wilson's disease induced pluripotent stem cells using gene therapy or the chaperone drug curcumin. Hum Mol Genet. 2011; 20: 3176–87. [DOI] [PubMed] [Google Scholar]
  • 166. Fujikura J, Nakao K, Sone M, et al Induced pluripotent stem cells generated from diabetic patients with mitochondrial DNA A3243G mutation. Diabetologia. 2012; 55: 1689–98. [DOI] [PubMed] [Google Scholar]
  • 167. Lemonnier T, Blanchard S, Toli D, et al Modeling neuronal defects associated with a lysosomal disorder using patient‐derived induced pluripotent stem cells. Hum Mol Genet. 2011; 20: 3653–66. [DOI] [PubMed] [Google Scholar]
  • 168. Moretti A, Ph D, Bellin M, et al Patient‐specific induced pluripotent stem‐cell models for long‐QT syndrome. N Engl J Med. 2010; 363: 1397–409. [DOI] [PubMed] [Google Scholar]
  • 169. Itzhaki I, Maizels L, Huber I, et al Modelling the long QT syndrome with induced pluripotent stem cells. Nature. 2011; 471: 225–9. [DOI] [PubMed] [Google Scholar]
  • 170. Ma D, Wei H, Zhao Y, et al Modeling type 3 long QT syndrome with cardiomyocytes derived from patient‐specific induced pluripotent stem cells. Int J Cardiol. 2013; 168: 5277–86. [DOI] [PubMed] [Google Scholar]
  • 171. Ge X, Ren Y, Bartulos O, et al Modeling supravalvular aortic stenosis syndrome with human induced pluripotent stem cells. Circulation. 2012; 126: 1695–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Tanaka A, Yuasa S, Mearini G, et al Endothelin‐1 induces myofibrillar disarray and contractile vector variability in hypertrophic cardiomyopathy‐induced pluripotent stem cell‐derived cardiomyocytes. J Am Heart Assoc. 2014; 3: e001263–e001263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Drawnel FM, Boccardo S, Prummer M, et al Disease modeling and phenotypic drug screening for diabetic cardiomyopathy using human induced pluripotent stem cells. Cell Rep. 2014; 9: 810–20. [DOI] [PubMed] [Google Scholar]
  • 174. Jiang Y, Habibollah S, Tilgner K, et al An induced pluripotent stem cell model of hypoplastic left heart syndrome (HLHS) reveals multiple expression and functional differences in HLHS‐derived cardiac myocytes. Stem Cells Transl Med. 2014; 3: 416–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Hitomi T, Habu T, Kobayashi H, et al The moyamoya disease susceptibility variant RNF213 R4810K (rs112735431) induces genomic instability by mitotic abnormality. Biochem Biophys Res Commun. 2013; 439: 419–26. [DOI] [PubMed] [Google Scholar]
  • 176. Itzhaki I, Maizels L, Huber I, et al Modeling of catecholaminergic polymorphic ventricular tachycardia with patient‐specific human‐induced pluripotent stem cells. J Am Coll Cardiol. 2012; 60: 990–1000. [DOI] [PubMed] [Google Scholar]
  • 177. Sun N, Yazawa M, Liu J, et al Patient‐specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Sci Transl Med. 2012; 4: 130ra47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Lan F, Lee AS, Liang P, et al Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient‐specific induced pluripotent stem cells. Cell Stem Cell. 2013; 12: 101–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Pessach IM, Ordovas‐Montanes J, Zhang S‐Y, et al Induced pluripotent stem cells: a novel frontier in the study of human primary immunodeficiencies. J. Allergy Clin. Immunol. 2011; 127: 1400–7.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Deyle DR, Khan IF, Ren G, et al Normal collagen and bone production by gene‐targeted human osteogenesis imperfecta iPSCs. Mol Ther. 2012; 20: 204–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Yamashita A, Morioka M, Kishi H, et al Statin treatment rescues FGFR3 skeletal dysplasia phenotypes. Nature. 2014; 513: 507–11. [DOI] [PubMed] [Google Scholar]
  • 182. Liu G‐H, Barkho BZ, Ruiz S, et al Recapitulation of premature ageing with iPSCs from Hutchinson‐Gilford progeria syndrome. Nature. 2011; 472: 221–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Shimamoto A, Kagawa H, Zensho K, et al Reprogramming suppresses premature senescence phenotypes of werner syndrome cells and maintains chromosomal stability over long‐term culture. PLoS ONE. 2014; 9: e112900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Snider L, Geng LN, Lemmers RJLF, et al Facioscapulohumeral dystrophy: incomplete suppression of a retrotransposed gene. PLoS Genet. 2010; 6: e1001181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Tedesco FS, Gerli MFM, Perani L, et al Transplantation of genetically corrected human iPSC‐derived progenitors in mice with limb‐girdle muscular dystrophy. Sci Transl Med. 2012; 4: 140ra89. [DOI] [PubMed] [Google Scholar]
  • 186. Du J, Campau E, Soragni E, et al Length‐dependent CTG·CAG triplet‐repeat expansion in myotonic dystrophy patient‐derived induced pluripotent stem cells. Hum Mol Genet. 2013; 22: 5276–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Quarto N, Leonard B, Li S, et al Skeletogenic phenotype of human Marfan embryonic stem cells faithfully phenocopied by patient‐specific induced‐pluripotent stem cells. Proc Natl Acad Sci USA. 2012; 109: 215–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Matsumoto Y, Hayashi Y, Schlieve CR, et al Induced pluripotent stem cells from patients with human fibrodysplasia ossificans progressiva show increased mineralization and cartilage formation. Orphanet J. Rare Dis. 2013; 8: 190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Mou H, Zhao R, Sherwood R, et al Generation of multipotent lung and airway progenitors from mouse ESCs and patient‐specific cystic fibrosis iPSCs. Cell Stem Cell. 2012; 10: 385–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Lachmann N, Happle C, Ackermann M, et al Gene correction of human induced pluripotent stem cells repairs the cellular phenotype in pulmonary alveolar proteinosis. Am J Respir Crit Care Med. 2014; 189: 167–82. [DOI] [PubMed] [Google Scholar]
  • 191. Somers A, Jean J‐C, Sommer CA, et al Generation of transgene‐free lung disease‐specific human induced pluripotent stem cells using a single excisable lentiviral stem cell cassette. Stem Cells. 2010; 28: 1728–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Tolar J, Xia L, Riddle MJ, et al Induced pluripotent stem cells from individuals with recessive dystrophic epidermolysis bullosa. J. Invest. Dermatol. 2011; 131: 848–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Tolar J, McGrath JA, Xia L, et al Patient‐specific naturally gene‐reverted induced pluripotent stem cells in recessive dystrophic epidermolysis bullosa. J. Invest. Dermatol. 2013; 134: 1246–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Ross J, Busch J, Mintz E, et al A rare human syndrome provides genetic evidence that WNT signaling is required for reprogramming of fibroblasts to induced pluripotent stem cells. Cell Rep. 2014; 9: 1770–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Mica Y, Lee G, Chambers SM, et al Modeling neural crest induction, melanocyte specification, and disease‐related pigmentation defects in hESCs and patient‐specific iPSCs. Cell Rep. 2013; 3: 1140–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Soyombo AA, Wu Y, Kolski L, et al Analysis of induced pluripotent stem cells from a BRCA1 mutant family. Stem Cell Reports. 2013; 1: 336–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197. Jin Z‐B, Okamoto S, Osakada F, et al Modeling retinal degeneration using patient‐specific induced pluripotent stem cells. PLoS ONE. 2011; 6: e17084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Li Y, Wu W‐H, Hsu C‐W, et al Gene therapy in patient‐specific stem cell lines and a preclinical model of retinitis pigmentosa with membrane frizzled‐related protein defects. Mol Ther. 2014; 22: 1688–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Howden SE, Gore A, Li Z, et al Genetic correction and analysis of induced pluripotent stem cells from a patient with gyrate atrophy. Proc Natl Acad Sci USA. 2011; 108: 6537–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Singh R, Shen W, Kuai D, et al iPS cell modeling of Best disease: insights into the pathophysiology of an inherited macular degeneration. Hum Mol Genet. 2013; 22: 593–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Qiu X, Yang J, Liu T, et al Efficient generation of lens progenitor cells from cataract patient‐specific induced pluripotent stem cells. PLoS ONE. 2012; 7: e32612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Shalom‐Feuerstein R, Serror L, Aberdam E, et al Impaired epithelial differentiation of induced pluripotent stem cells from ectodermal dysplasia‐related patients is rescued by the small compound APR‐246/PRIMA‐1MET. Proc Natl Acad Sci USA. 2013; 110: 2152–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Thatava T, Armstrong AS, De Lamo JG, et al Successful disease‐specific induced pluripotent stem cell generation from patients with kidney transplantation. Stem Cell Res Ther. 2011; 2: 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Li W, Wang X, Fan W, et al Modeling abnormal early development with induced pluripotent stem cells from aneuploid syndromes. Hum Mol Genet. 2012; 21: 32–45. [DOI] [PubMed] [Google Scholar]
  • 205. Ma Y, Li C, Gu J, et al Aberrant gene expression profiles in pluripotent stem cells induced from fibroblasts of a Klinefelter syndrome patient. J Biol Chem. 2012; 287: 38970–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206. Nakagawa M, Takizawa N, Narita M, et al Promotion of direct reprogramming by transformation‐deficient Myc. Proc Natl Acad Sci USA. 2010; 107: 14152–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207. Fang I‐M, Yang C‐M, Yang C‐H, et al Transplantation of induced pluripotent stem cells without C‐Myc attenuates retinal ischemia and reperfusion injury in rats. Exp Eye Res. 2013; 113: 49–59. [DOI] [PubMed] [Google Scholar]
  • 208. Sidhu KS. New approaches for the generation of induced pluripotent stem cells. Expert Opin. Biol. Ther. 2011; 11: 569–79. [DOI] [PubMed] [Google Scholar]
  • 209. Cho H‐J, Lee C‐S, Kwon Y‐W, et al Induction of pluripotent stem cells from adult somatic cells by protein‐based reprogramming without genetic manipulation. Blood. 2010; 116: 386–95. [DOI] [PubMed] [Google Scholar]
  • 210. Sun N, Longaker MT, Wu JC. Human iPS cell‐based therapy: considerations before clinical applications. Cell Cycle. 2010; 9: 880–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211. Okita K, Nagata N, Yamanaka S. Immunogenicity of induced pluripotent stem cells. Circ Res. 2011; 109: 720–1. [DOI] [PubMed] [Google Scholar]
  • 212. Yu J, Hu K, Smuga‐Otto K, et al Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009; 324: 797–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Condic ML, Rao M. Regulatory issues for personalized pluripotent cells. Stem Cells. 2008; 26: 2753–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Knoepfler PS. Key anticipated regulatory issues for clinical use of human induced pluripotent stem cells. Regen Med. 2012; 7: 713–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Harding J, Mirochnitchenko O. Preclinical studies for induced pluripotent stem cell‐based therapeutics. J Biol Chem. 2014; 289: 4585–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216. Takagi Y, Takahashi J, Saiki H, et al Dopaminergic neurons generated from monkey embryonic stem cells function in a Parkinson primate model. J. Clin. Invest. 2005; 115: 102–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. Ishii T, Yasuchika K, Machimoto T, et al Transplantation of embryonic stem cell‐derived endodermal cells into mice with induced lethal liver damage. Stem Cells. 2007; 25: 3252–60. [DOI] [PubMed] [Google Scholar]
  • 218. Leor J, Gerecht S, Cohen S, et al Human embryonic stem cell transplantation to repair the infarcted myocardium. Heart. 2007; 93: 1278–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219. Boyd AS, Wu DC, Higashi Y, et al A comparison of protocols used to generate insulin‐producing cell clusters from mouse embryonic stem cells. Stem Cells. 2008; 26: 1128–37. [DOI] [PubMed] [Google Scholar]
  • 220. 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] [PubMed] [Google Scholar]
  • 221. Hentze H, Graichen R, Colman A. Cell therapy and the safety of embryonic stem cell‐derived grafts. Trends Biotechnol. 2007; 25: 24–32. [DOI] [PubMed] [Google Scholar]
  • 222. Halme DG, Kessler DA. FDA regulation of stem‐cell‐based therapies. N Engl J Med. 2006; 355: 1730–5. [DOI] [PubMed] [Google Scholar]
  • 223. Rao M. Scalable human ES culture for therapeutic use: propagation, differentiation, genetic modification and regulatory issues. Gene Ther. 2008; 15: 82–8. [DOI] [PubMed] [Google Scholar]
  • 224. Liao M‐C, Diaconu M, Monecke S, et al Embryonic stem cell‐derived neural progenitors as non‐tumorigenic source for dopaminergic neurons. World J. Stem Cells. 2014; 6: 248–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Shibata H, Ageyama N, Tanaka Y, et al Improved safety of hematopoietic transplantation with monkey embryonic stem cells in the allogeneic setting. Stem Cells. 2006; 24: 1450–7. [DOI] [PubMed] [Google Scholar]
  • 226. Fujikawa T, Oh S‐H, Pi L, et al Teratoma formation leads to failure of treatment for type I diabetes using embryonic stem cell‐derived insulin‐producing cells. Am J Pathol. 2005; 166: 1781–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Fair JH, Cairns BA, Lapaglia MA, et al Correction of factor IX deficiency in mice by embryonic stem cells differentiated in vitro . Proc Natl Acad Sci USA. 2005; 102: 2958–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228. Arnhold S, Klein H, Semkova I, et al Neurally selected embryonic stem cells induce tumor formation after long‐term survival following engraftment into the subretinal space. Invest Ophthalmol Vis Sci. 2004; 45: 4251–5. [DOI] [PubMed] [Google Scholar]
  • 229. Park I‐H, Zhao R, West JA, et al Reprogramming of human somatic cells to pluripotency with defined factors. Nature. 2008; 451: 141–6. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Cellular and Molecular Medicine are provided here courtesy of Blackwell Publishing

RESOURCES