Trabalho 2012 teo2010 regenerative therapy

13
Biochem. J. (2010) 428, 11–23 (Printed in Great Britain) doi:10.1042/BJ20100102 11 REVIEW ARTICLE Emerging use of stem cells in regenerative medicine Adrian K. K. TEO*and Ludovic VALLIER* 1 *Laboratory for Regenerative Medicine, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, U.K., and Institute of Medical Biology, A*STAR (Agency for Science, Technology and Research), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648 Stem cells represent a unique opportunity for regenerative medicine to cure a broad number of diseases for which current treatment only alleviates symptoms or retards further disease progression. However, the number of stem cells available has speedily increased these past 10 years and their diversity presents new challenges to clinicians and basic scientists who intend to use them in clinics or to study their unique properties. In addition, the recent possibility to derive pluripotent stem cells from somatic cells using epigenetic reprogramming has further increased the clinical interest of stem cells since induced pluripotent stem cells could render personalized cell-based therapy possible. The present review will attempt to summarize the advantages and challenges of each type of stem cell for current and future clinical applications using specific examples. Key words: cell replacement therapy, human, regenerative medicine, stem cell. INTRODUCTION During the past decade, stem cells have become a major focus for translational medicine. These cell types combine two properties that make them uniquely attractive for regenerative medicine. A stem cell is defined as a cell type which has the property to self- renew while maintaining the capacity to differentiate into diverse cell types. Stem cells are therefore different from progenitor cells which can differentiate into mature cell types but are incapable of self-renewing, or somatic cells which are capable of proliferating but unable to differentiate. By combining these properties of self- renewal and differentiation, stem cells could allow the production of an infinite quantity of cell types with a clinical interest. Stem cells are also becoming increasingly attractive for basic research since they can be expanded in vitro while maintaining their native properties, allowing basic studies which are impossible with primary tissue cultures or biopsy material. Consequently, specific stem cells such as ESCs (embryonic stem cells) have become a reference model for understanding key molecular mechanisms which control cell fate choice and organ differentiation. Importantly, stem cells are already present in clinics especially HSCs (haematopoietic stem cells), which have been successfully used for more than 40 years in bone marrow transplantations to treat diverse blood disorders such as leukaemia [1]. Future clinical applications of stem cells concern a broad number of degenerative diseases (i.e. disease in which one cell type or part of an organ fails) which could potentially be treated using stem-cell-based therapy. This includes major metabolic diseases such as T1D (Type 1 diabetes), caused by the destruction of insulin-secreting β -cells [2], diverse brain and myelin disorders in which specific neural cells are targeted such as MS (multiple sclerosis), PD (Parkinson’s disease) or HD (Huntington’s disease), heart disease, where some cardiac cells need to be replaced upon myocardial infarction, and genetic diseases like myopathy, where a specific subtype of cells are not functional. The number of diseases that could be cured using stem cells is extremely broad. Thus the present review will only focus on specific examples of current applications which illustrate the recent progress towards the use of stem cells in clinics. Human stem cells can be classified into three broad categories in accordance with their origin and their capacity of differentiation. Somatic stem cells are multipotent stem cells which originate from differentiated organs (adult or foetal). They can reside specifically in in vivo niches (endogenous somatic stem cells) and sometimes can be isolated and expanded in vitro (exogenous somatic stem cells). The third category (pluripotent stem cells) includes ESCs which are derived from embryos at the blastocyst stage and hiPSCs [human iPSCs (induced pluripotent stem cells)] which are generated from reprogrammed somatic cells. In the present review, we will examine the properties of each stem cell category and discuss their advantages for clinical application by describing relevant examples. In addition, we will define their drawbacks and the necessary progress for their future clinical applications. TOTIPOTENCE, PLURIPOTENCE AND MULTIPOTENCE The clinical use of a specific type of stem cell is ultimately defined by its capacity to generate a broad number of cell types. This capacity of differentiation can be divided into three categories. Totipotent stem cells are capable of differentiating into all adult and embryonic tissues, including extra-embryonic tissues such as trophectoderm. Embryos at the one and two cell stages and GCs (germ cells) are likely to be the only cells which display this capacity naturally in an embryonic environment [3]. So far, there is no known stem cell in vitro which has no restriction in its capacity of differentiation. The major issue here is to define the assays and Abbreviations used: AFP, α foetal protein; ALB, albumin; AT, adipose tissue; BM, bone marrow; CK, cytokeratin; DE, definitive endoderm; EGF, epidermal growth factor; ESC, embryonic stem cell; FD, familial dysautonomia; FGF, fibroblast growth factor; GC, germ cell; GvHD, graft versus host disease; hESC, human ESC; HNF, hepatocyte nuclear factor; HSC, haematopoietic stem cell; IGF1, insulin-like growth factor 1; iPSC, induced pluripotent stem cell; hiPSC, human iPSC; MAPC, multipotent adult progenitor cell; mESC, mouse ESC; MSC, mesenchymal stromal cell; NSC, neural stem cell; OPC, oligodendrocyte progenitor cell; PD, Parkinson’s disease; SCF, stem cell factor; SCNT, somatic cell nuclear transfer; SGL, subgranular layer; SMA, spinal muscular atrophy; SVZ, subventricular zone; T1D, Type 1 diabetes; TGF, transforming growth factor; UCB, umbilical cord blood. 1 To whom correspondence should be addressed (email [email protected]). c The Authors Journal compilation c 2010 Biochemical Society www.biochemj.org Biochemical Journal

Transcript of Trabalho 2012 teo2010 regenerative therapy

Biochem. J. (2010) 428, 11–23 (Printed in Great Britain) doi:10.1042/BJ20100102 11

REVIEW ARTICLEEmerging use of stem cells in regenerative medicineAdrian K. K. TEO*† and Ludovic VALLIER*1

*Laboratory for Regenerative Medicine, University of Cambridge, West Forvie Building, Forvie Site, Robinson Way, Cambridge CB2 0SZ, U.K., and †Institute of Medical Biology,A*STAR (Agency for Science, Technology and Research), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648

Stem cells represent a unique opportunity for regenerativemedicine to cure a broad number of diseases for which currenttreatment only alleviates symptoms or retards further diseaseprogression. However, the number of stem cells available hasspeedily increased these past 10 years and their diversity presentsnew challenges to clinicians and basic scientists who intend to usethem in clinics or to study their unique properties. In addition, therecent possibility to derive pluripotent stem cells from somaticcells using epigenetic reprogramming has further increased the

clinical interest of stem cells since induced pluripotent stem cellscould render personalized cell-based therapy possible. The presentreview will attempt to summarize the advantages and challengesof each type of stem cell for current and future clinical applicationsusing specific examples.

Key words: cell replacement therapy, human, regenerativemedicine, stem cell.

INTRODUCTION

During the past decade, stem cells have become a major focus fortranslational medicine. These cell types combine two propertiesthat make them uniquely attractive for regenerative medicine. Astem cell is defined as a cell type which has the property to self-renew while maintaining the capacity to differentiate into diversecell types. Stem cells are therefore different from progenitor cellswhich can differentiate into mature cell types but are incapable ofself-renewing, or somatic cells which are capable of proliferatingbut unable to differentiate. By combining these properties of self-renewal and differentiation, stem cells could allow the productionof an infinite quantity of cell types with a clinical interest. Stemcells are also becoming increasingly attractive for basic researchsince they can be expanded in vitro while maintaining their nativeproperties, allowing basic studies which are impossible withprimary tissue cultures or biopsy material. Consequently, specificstem cells such as ESCs (embryonic stem cells) have becomea reference model for understanding key molecular mechanismswhich control cell fate choice and organ differentiation.

Importantly, stem cells are already present in clinics especiallyHSCs (haematopoietic stem cells), which have been successfullyused for more than 40 years in bone marrow transplantations totreat diverse blood disorders such as leukaemia [1]. Future clinicalapplications of stem cells concern a broad number of degenerativediseases (i.e. disease in which one cell type or part of an organfails) which could potentially be treated using stem-cell-basedtherapy. This includes major metabolic diseases such as T1D(Type 1 diabetes), caused by the destruction of insulin-secretingβ-cells [2], diverse brain and myelin disorders in which specificneural cells are targeted such as MS (multiple sclerosis), PD(Parkinson’s disease) or HD (Huntington’s disease), heart disease,where some cardiac cells need to be replaced upon myocardial

infarction, and genetic diseases like myopathy, where a specificsubtype of cells are not functional. The number of diseases thatcould be cured using stem cells is extremely broad. Thus thepresent review will only focus on specific examples of currentapplications which illustrate the recent progress towards the useof stem cells in clinics.

Human stem cells can be classified into three broad categories inaccordance with their origin and their capacity of differentiation.Somatic stem cells are multipotent stem cells which originate fromdifferentiated organs (adult or foetal). They can reside specificallyin in vivo niches (endogenous somatic stem cells) and sometimescan be isolated and expanded in vitro (exogenous somatic stemcells). The third category (pluripotent stem cells) includes ESCswhich are derived from embryos at the blastocyst stage andhiPSCs [human iPSCs (induced pluripotent stem cells)] whichare generated from reprogrammed somatic cells. In the presentreview, we will examine the properties of each stem cell categoryand discuss their advantages for clinical application by describingrelevant examples. In addition, we will define their drawbacks andthe necessary progress for their future clinical applications.

TOTIPOTENCE, PLURIPOTENCE AND MULTIPOTENCE

The clinical use of a specific type of stem cell is ultimately definedby its capacity to generate a broad number of cell types. Thiscapacity of differentiation can be divided into three categories.Totipotent stem cells are capable of differentiating into all adultand embryonic tissues, including extra-embryonic tissues suchas trophectoderm. Embryos at the one and two cell stages andGCs (germ cells) are likely to be the only cells which display thiscapacity naturally in an embryonic environment [3]. So far, there isno known stem cell in vitro which has no restriction in its capacityof differentiation. The major issue here is to define the assays and

Abbreviations used: AFP, α foetal protein; ALB, albumin; AT, adipose tissue; BM, bone marrow; CK, cytokeratin; DE, definitive endoderm; EGF, epidermalgrowth factor; ESC, embryonic stem cell; FD, familial dysautonomia; FGF, fibroblast growth factor; GC, germ cell; GvHD, graft versus host disease; hESC,human ESC; HNF, hepatocyte nuclear factor; HSC, haematopoietic stem cell; IGF1, insulin-like growth factor 1; iPSC, induced pluripotent stem cell; hiPSC,human iPSC; MAPC, multipotent adult progenitor cell; mESC, mouse ESC; MSC, mesenchymal stromal cell; NSC, neural stem cell; OPC, oligodendrocyteprogenitor cell; PD, Parkinson’s disease; SCF, stem cell factor; SCNT, somatic cell nuclear transfer; SGL, subgranular layer; SMA, spinal muscular atrophy;SVZ, subventricular zone; T1D, Type 1 diabetes; TGF, transforming growth factor; UCB, umbilical cord blood.

1 To whom correspondence should be addressed (email [email protected]).

c© The Authors Journal compilation c© 2010 Biochemical Society

www.biochemj.org

Bio

chem

ical

Jo

urn

al

12 A. K. K. Teo and L. Vallier

Table 1 Types of pluripotent stem cells derived from embryos and thegermline

mEpiSC, mouse epiblast stem cell; m(h)EGC, mouse (human) embryonic GC; mGSC, multipotentgermline stem cell; PGC, primordial GC.

Type of pluripotent stem cell Source of derivation Reference

mESC Pre-implantation embryo [5,6]Primate ESC Pre-implantation embryo [176,177]hESC Pre-implantation embryo [111,178]

Morula and late blastocyst stage embryo [179,180]Single blastomere [181]Parthenogenetic embryo [182–184]

mEpiSC Post-implantation embryo [185,186]mEGC PGC [187,188]mGSC Neonatal and adult mouse testis [189,190]hEGC Cultured human PGCs [191]

standards that would allow the definition of totipotency of humanstem cells in vitro. Obvious ethical considerations preclude theuse of chimaera formation and germline transmission in humans(see below) and the development of in vitro assays to demonstratethat one cell type can differentiate into all the cell types existingin the human body will be extremely challenging.

Pluripotent stem cells are capable of differentiating into thederivatives of the three germ layers (ectoderm, mesoderm andendoderm) and GCs. However, they are not totipotent as theycannot differentiate into certain cell types especially extra-embryonic tissues such as trophectoderm. Pluripotent stem cellshave been derived from embryos at different developmentalstages and from the adult germline [4] (Table 1). Pluripotencyis currently defined by specific assays. The most commonly usedassay in non-human species is germline transmission. In this case,pluripotent stem cells are injected into embryos at the blastocyststage and the resulting chimaeric embryo is re-transplanted intoa pseudo-pregnant female to continue its development. Theoffspring is chimaeric in all the tissues, including the germline,which is validated by the transmission of the genetic backgroundoriginating from the injected stem cells to the next generation.The best example of pluripotent stem cells fulfilling this criteriaremains to be mESCs (mouse ESCs) [5,6], which can differentiateinto various tissues in vitro and can contribute to the germlinein vivo after blastocyst injection. However, mESCs are unableto differentiate into trophectoderm in the absence of geneticmodifications and thus are defined as pluripotent stem cells.Concerning hESCs (human ESCs), their pluripotent state hasbeen demonstrated by a broad number of studies showing thatthey can differentiate into a large number of cell types includingthe trophectoderm and germ cells [7,8]. However, germlinetransmission assays are unfeasible for obvious ethical reasonsand pluripotency in human cells is usually demonstrated by theircapacity to form teratomas. Indeed, pluripotent stem cells canform teratomas containing derivatives of the three germ layersonce injected into an immunodeficient host. However, this assayis not quantitative and relies on histological analyses. Thereforethe lack of proper standards to define pluripotency in human stemcells remains an issue which could explain the current confusionregarding the pluripotent status of somatic stem cells such asMAPCs (multipotent adult progenitor cells) [9,10].

Multipotent stem cells are able to differentiate into all the celltypes constituting an organ. The most prominent example remainsHSCs, which are capable of differentiating into all the cell types ofthe haematopoietic system. Two types of assays are currentlyused to demonstrate the multipotent state of stem cells. The firstis to demonstrate that one stem cell can reconstitute an

entire organ. For instance, HSCs can reconstitute an entirehaematopoietic system after transplantation into sub-lethallyirradiated animals. However, this type of assay is difficult toperform in humans since they require the destruction of thetargeted organ and also require clonal analysis which can betechnically challenging. Indeed, adult stem cells often requirea complex environment to survive and to self-renew in, and thisniche can be difficult to mimic in vitro. The second type of assayis based on lineage tracing experiments in which the stem cellof interest is genetically tagged and hence marks its progeny.Thus, using this approach, one can follow the origin of a celltype in a specific organ. The existence of gut stem cells [11] andskin stem cells [12] has been characterized using this approach.Multipotent stem cells have been identified in a broad numberof organs in rodents. However, the existence and functionalityof their human counterpart remains in some cases controversialsince the lack of appropriate technology renders their isolationand thus characterization extremely challenging.

SOMATIC STEM CELLS

Endogenous somatic stem cells

Somatic stem cells are multipotent and can usually differentiateinto all the cell types constituting their organ of origin. Somesomatic stem cells such as hepatic progenitor cells proliferatevery slowly and can be activated by specific conditions suchas stress response or injury [13]. Other somatic stem cellssuch as vascular progenitor cells are proliferating constantlyand are in high numbers [14]. They are often localized in aspecific niche that is essential to maintain their property ofself-renewal and differentiation [15]. Ultimately, their normalfunction could be to maintain a constant number of cells inan organ. Somatic stem cells have been reported to be foundin the brain [16,17], haematopoietic system, skin [18,19], heart[20,21], muscle [22,23], lung [24,25], intestine [26] and liver[27,28] (Figure 1 and Table 2). They represent the ideal target forregenerative medicine since their mobilization would not requireinvasive methods or immunosuppressive treatments.

For instance, a broad number of studies have focused onactivating endogenous NSCs (neural stem cells) to replace missingneural cells [29,30]. The use of FGF2 (fibroblast growth factor 2),EGF (epidermal growth factor) [31], TGFα (transforming growthfactor α) [32], VEGF (vascular endothelial growth factor) [33,34],SCF (stem cell factor) [35], glucocorticoids and IGF1 (insulin-likegrowth factor 1) [36] could possibly activate neuronal replacementin neurodegenerative diseases and also in stroke injury. Anotherexample would be the activation of in vivo muscle repair usinggrowth factors [37]. IGF1 has been strongly linked to promotecellular recruitment and muscular repair [38]. Together with theinhibition of the NF-κB (nuclear factor κB) pathway [39] andmodulation of TNFα (tumour necrosis factor α) function [40],muscular atrophy could be prevented [41].

However, the clinical use of endogenous somatic stem cellsis currently limited for diverse reasons. Their existence remainscontroversial in several organs especially in humans where lineagetracing or reconstitution experiments are impossible. Thereforetheir identification and characterization is difficult and pastexperience has shown that results obtained in animal models mightnot be translated directly to humans. This limits the study of themechanisms controlling the self-renewal and differentiation ofendogenous stem cells, which is necessary for the developmentof future clinical treatments. Two controversial examples concernthe pancreas and liver. Using lineage tracing techniques inthe mouse, it has been demonstrated that pancreatic β-cells

c© The Authors Journal compilation c© 2010 Biochemical Society

Emerging use of stem cells in regenerative medicine 13

Table 2 Types of adult stem cells reportedly found

Type of adult stem cell Location found Expandable in vitro? Used in clinical trials?

HSC BM, placenta, UCB and peripheral blood Yes YesMSC BM, amniotic fluid, placenta, AT and UCB Yes YesMuscle satellite stem cell Between basement membrane and cell membrane (sarcolemma) of muscle fibre No YesNSC SVZ of lateral ventricles and SGL of dentate gyrus of the hippocampus in the brain Yes Going to startLimbal stem cell Corneoscleral limbus Yes Going to startSkin stem cell Basal layer of epidermis and base of hair follicles Yes NoGut stem cell Crypts Yes NoLung stem cell Between airways (bronchioles) and alveoli No NoCardiac stem cell Heart No NoLiver stem cell Canals of Hering at the terminal bile ductules Yes NoPancreatic stem cell Pancreatic ducts No No

Figure 1 Endogenous and exogenous somatic stem cells for cell-based therapy

Endogenous adult stem cells (SC) can be activated with the use of drugs or growth factors to prompt regeneration in tissues or organs in vivo. Exogenous adult stem cells can be expanded in exvivo cultures and then transplanted into the human body for cell replacement or to stimulate regeneration via paracrine actions. Stem cells which remain to be fully characterized are indicated by (?).

‘regenerate’ via self-replication of existing β-cells and not fromstem cells [42,43]. However, recent reports have contradictedthese observations by showing that β-cells can be made denovo from islet progenitor cells expressing Ngn3 and residingin the pancreatic duct [44]. Nevertheless, most of these resultswere obtained in animal models of injury in which the pancreasis induced to regenerate after duct ligation, and recent studieshave not been able to repeat these experiments [45]. Thereforeit remains to be demonstrated that this model is physiologicallyrelevant and that a pool of pancreatic stem cells can be identifiedin normal conditions, especially in humans.

Hepatic oval cells/hepatic progenitors represent anothercontroversial adult stem cell [46,47]. These liver-specific stemcells originally identified in rodents could be used to makehepatocytes for cell-based therapy of liver failure and livermetabolic diseases [48–50]. They are bipotent (i.e. they arecapable to differentiate into duct cells or hepatocytes) as shownby the expression of bile duct markers [CK (cytokeratin)-7,

CK-19 and OV-6], and hepatocyte markers [AFP (α foetal protein)and ALB (albumin)]. They also express HNFs (hepatocyte nuclearfactors) which mark hepatic progenitors during embryonic liverdevelopment [51]. In humans, they are localized in the canals ofHering at the terminal bile ductules [52] and are reportedly activ-ated in disease conditions [53,54]. However, they are extremelyrare (if in existence) and possibly inaccessible [52]. There havebeen reports of the isolation of hepatic progenitor cells from theadult human liver [55–57]. However, the lack of definitive markersand the dissimilarity between human and rodents renders theidentification of a common hepatic progenitor cell difficult [58].

These two examples illustrate the difficulty of translatingobservations obtained in animal models directly to humans [59–61]. Indeed, stem cells in distant species might have differentfunctions and activity [62,63]. Another major drawback isthat endogenous somatic stem cells could be targeted by thediseases, explaining why endogenous somatic stem cells appearto be incapable or insufficient in stopping the progression of

c© The Authors Journal compilation c© 2010 Biochemical Society

14 A. K. K. Teo and L. Vallier

degenerative diseases. Finally, the use of endogenous stemcells could be precluded in genetic diseases whereby stem cellscontinue to produce progeny carrying the same phenotype.Consequently, genetic correction will be necessary, implicatingthe use of gene therapy approaches or complex manipulationsin vitro. In conclusion, mobilization of endogenous somatic stemcells represents the ultimate objective of regenerative medicine.This approach is the most ideal, ‘simple’ and natural as weare simply prompting and activating our innate body system torepair diseased tissues. There would not be complicated problemssuch as immunological response to exogenous foreign cells orethical issues related to ESCs. However, their clinical use willrequire better identification and characterization to develop newapproaches allowing their mobilization in vivo, especially in adiseased environment.

Exogenous somatic stem cells

Exogenous somatic stem cells are derived in vitro from diverseorgans. They have a high capacity of proliferation and can bemaintained in vitro for a prolonged period of time. Exogenoussomatic stem cells are commonly recognized to exhibit lineage-restricted multipotency. However, several reports have suggestedthat somatic stem cells could be pluripotent since they candifferentiate into a broad number of cell types unrelated totheir organ of origin [64]. In addition, controversial reportshave suggested that these cells could participate in germ layerformation [64]. However, these results have been difficult toreplicate and the current consensus is that adult somatic stemcells have a restricted capacity of differentiation compared withthose of ESCs. Furthermore, exogenous somatic stem cells oftenrepresent an in vitro state which has no equivalence in vivo.However, their use in regenerative medicine is promising sincethey are easy to isolate and can be expanded in vitro. In addition,immunosuppressive treatments are not required due to autologoustransplantations, making them ideal candidates for clinical trials.The two best examples are NSCs [65,66] and multipotent MSCs(mesenchymal stromal cells) [67,68] which are currently beingused in several clinical trials.

NSCs are progenitor cells located in the SVZ (subventricularzone) of the lateral ventricles or SGL (subgranular layer) of thedentate gyrus of the hippocampus in the brain. NSCs have beencultured in vitro as cell clumps known as ‘neurospheres’ [69,70].The neurospheres can be passaged several times, demonstratingtheir capacity of self-renewal. They are also multipotent sincethey can differentiate into neurons and glia [71,72]. ThereforeNSCs represent a unique source of cells for clinical application,and several clinical trials are currently testing their efficiency[73]. One of them is conducted by StemCells Inc. (San Francisco,CA, U.S.A.) and its objective is to use NSCs for the treatmentof Batten’s disease, a lysosomal storage disorder. In addition,several trials have used neuronal cells for stroke patients [74,75].ReNeuron (Guildford, U.K.) is currently proposing to transplantmyc-immortalized hNSCs (human NSCs) [76] derived fromhuman foetal tissue into stroke patients. Therefore the first clinicalapplications of NSCs have started. However, several challengeshave to be solved before larger trials could start, including betterisolation and expansion methods for NSCs, as well as preventionof tumour formation after transplantation [77].

MSCs are fibroblast-like cells which can be isolated fromdiverse sources including BM (bone marrow) [78], amnioticfluid [79], placenta [80,81], AT (adipose tissue) [82,83] andUCB (umbilical cord blood) [84]. MSCs are mainly identifiedby their adherent property, differentiation potential, and cell-surface markers such as CD73, CD90 and CD105 [67,85,86].

However, their capacity of differentiation in vivo and in vitrois limited as they are not pluripotent. Besides MSCs, severalrelated types of cells including MAPCs were isolated from theBM [9,10]. MSCs are multipotent [87] and have been reportedto be able to differentiate into osteoblasts, adipocytes [88],chondrocytes [89,90], and epithelial [91], endothelial [92] andneural cells [93]. It should be noted that MSCs derived fromdifferent tissues may exhibit slightly different propensities andcapacities to differentiate into specific cell types. For example,MSCs from synovial tissue may be more effective in repairingcartilage defects than MSCs from other tissues [94]. MSCs self-renew in vitro but not indefinitely since they do senesce after20 population doublings and ultimately lose their potential todifferentiate [95]. More importantly, they do tend to generateabnormal karyotypes in vitro as their genomes are highly unstable[96]. This could represent a major limitation for their use in cell-based therapy.

Nonetheless, the interest in MSCs for clinical applicationsresides less in their capacity of differentiation than in theirparacrine actions on tissue repair [97]. Indeed, MSCs expresslow levels of MHC class II molecules and thus should be weaklyimmunogenic, suggesting that immunosuppressive treatmentmight not be required. In addition, they have been shown to beable to modulate the immune response of cells in vitro and tolimit inflammation [98]. Hence they could be used for preventinggraft rejections and GvHD (graft versus host disease). MSCswere first used in clinical trials as a complementary treatment forbreast cancer patients [99]. Thereafter they were demonstratedfurther to be beneficial against GvHD [100]. On the basis ofthese successes, a broad number of studies are currently beingperformed to define their efficacy to treat Crohn’s disease [101],ALS (amyotrophic lateral sclerosis) [102] and heart diseases[103]. In particular for heart therapy, autologous BM-MSCs havebeen used in controlled clinical trials [104]. Importantly, MSCs donot repopulate damaged tissues and cells quickly disappear afterengraftment in vivo. Despite this, small but positive effects oncardiac performance have been observed, and this clinical benefitcould be achieved through immune modulation and increasedangiogenesis.

MSCs could also potentially be used to treat liver diseases.BM-MSCs apparently are able to differentiate into hepatocyte-like cells without cell fusion [105,106]. UCB-MSCs [107] andAT-MSCs [108] can also be differentiated into hepatocyte-likecells. hMSC (human MSC)-derived hepatocytes have recentlybeen reported to be able to restore liver function [109]. MSCs arenow being used in clinical trials for end-stage liver disease [110]and initial results appear promising. However, long-term recoveryof liver function remains to be demonstrated and further studieswill be required to understand the mechanisms by which MSCscan restore liver function. In summary, the mechanisms by whichMSCs can achieve their immunomodulatory and detoxificationfunctions, as well as paracrine secretions, remain to be defined.Further basic studies will be extremely helpful in validating theirreal function in vivo and the mechanisms by which they canalleviate so many symptoms. However, their interest for clinicalapplication cannot be denied and the broad number of clinicaltrials which are currently performed with MSCs should rapidlyprovide some information regarding their real efficacy and long-term effect.

The examples described above illustrate the advantagesof somatic stem cells for clinical applications. These cellsare relatively easy to isolate and they do not requireimmunosuppressive treatment. In addition, they can be used totreat a broad number of diseases without major risks. All ofthese aspects greatly simplify their use in a clinical context

c© The Authors Journal compilation c© 2010 Biochemical Society

Emerging use of stem cells in regenerative medicine 15

and thus make them attractive for clinical trials. However, anincreasing amount of data are suggesting that even a shortperiod of time in culture in vitro can modify the genetic andepigenetic characteristics of somatic stem cells. The consequencesof these modifications on cell proliferation and differentiationafter transplantation and after a prolonged period of timein vivo will have to be examined carefully. Finally, the mechanismsinvolving somatic stem cells in tissue repair and/or in modulatingimmune response are not always fully understood and additionalbasic studies are required to fully define the clinical potential ofthese different cell types.

EMBRYONIC STEM CELLS

hESCs are pluripotent stem cells derived from the innercell mass of embryos at the blastocyst stage [111].Their embryonic origin confers upon them the capacity toproliferate indefinitely in vitro while maintaining the capacityto differentiate into derivatives of the three germ layers from whichall adult organs are derived. Therefore hESCs represent a uniqueopportunity for regenerative medicine since they could be usedto generate an infinite quantity of cells with a clinical interest.However, the clinical application of these unique cell types iscurrently limited by two challenges: the difficulty to generatefully functional cell types and also the safety issue associatedwith teratoma formation.

Difficulty to generate fully functional cell types from hESCs

A major focus of the past 10 years has been the developmentof robust protocols to differentiate hESCs into homogenouspopulations of cell types with a clinical interest. Consequently,a broad number of protocols have been developed to generatediverse cell types, including retina cells [112,113], dopaminergicneurons [114], motor neurons [115,116], OPCs (oligodendrocyteprogenitor cells) [117], cardiomyocytes [118,119], pancreatic β-cells [120] and hepatocytes [121–124] (Figure 2 and Figure 3).These methods mark a clear progress towards the generation offully functional cell types. They have benefited from the lessonslearnt from mESCs as well as from the knowledge developedby developmental biology studies. Indeed, past experiences haveshown that following a natural path of development in vitroremains the best way to generate functional cell types frompluripotent stem cells of embryonic origin. Therefore most of theprotocols developed to drive differentiation of hESCs respect keystages of embryonic development. However, this methodologyalso implies that generation of fully mature and functionalcells from hESCs will require an extended period of time ofin vitro culture, mimicking a complex environment constantlychanging over time. Therefore such an approach is presentedwith unprecendented challenges which would require a strongcollaboration between stem cell biologists and developmentalbiologists. Generation of pancreatic β-cells is a good exampleto illustrate the progress and challenges regarding the generationof clinically relevant cells from hESCs.

The Edmonton Protocol has proven that islet transplantation viathe hepatic portal vein can correct insulin deficiency in diabeticpatients [125]. However, the lack of cadaveric donors severelylimits the number of patients that could benefit from this therapy.Generating pancreatic cells from hESCs represent a uniqueopportunity to bypass this limitation. However, the resultingβ-cells would have to synthesize and store insulin, and alsomaintain blood glucose in a physiological range when challengedwith sporadic food intake. Past experiences [126,127] have

demonstrated that strict adherence to the pancreas developmentalprocess is essential for the production of pancreatic cells fromESCs [2,128,129]. Accordingly, a robust method of differentiationshould respect four major stages: (i) formation of DE (definitiveendoderm) [130,131], (ii) formation of PE (pancreatic endoderm),(iii) formation of endocrine progenitors, and (iv) maturation ofendocrine progenitors into fully functional hormone-secretingcells. Several methods are now available to efficiently generateDE cells from hESCs and to further differentiate the resultingcells into pancreatic progenitors. However, the specification ofthese progenitors toward the endocrine pathway as well as theproduction of fully mature β-cells remains a challenge.

Indeed, the most advanced protocol for the differentiationof hESCs into pancreatic β-cells allows the production ofmulti-hormonal immature pancreatic endocrine cells which lackNkx6.1 and MafA co-expression. Despite this major limitation,the pancreatic progenitors generated using this approach candifferentiate into fully functional pancreatic cells in vivo aftertransplantation into immuno-deficient mice and the resultingβ-cells can re-establish euglycaemia in a mouse model ofdiabetes [120]. These results are extremely encouraging sincethey provide the first demonstration that stem-cell-based therapycould be used to treat diabetes. However, partially differentiatedpancreatic progenitors can easily be contaminated by pluripotentcells which further increase the risk of teratoma formationassociated with hESCs (see below). In addition, non-chemicallydefined culture conditions containing FBS (foetal bovine serum)are still used, increasing the difficulty for such approaches tobe approved for clinical trials. Finally, any cell-based therapyapproach will have to address specific problems associated withthe auto-immune disease involved in T1D. Therefore the resultsobtained with hESCs are very promising and they demonstratethe potential of pluripotent stem cells to develop new treatmentfor diabetes. However, more efforts are still required to generatefully functional β-cells from human pluripotent stem cells and todemonstrate that pancreatic progenitors can be used safely in aclinical context.

Importantly, the challenges described above for pancreaticcells are shared with many other cell types. For example, HSCsgenerated from hESCs resemble those of the yolk sac [132,133]and thus have a limited capacity of repopulation in immuno-deficient animals [134]. In addition, hESC-derived hepatocytesappear to have an embryonic identity since they have limitedcapacity to colonize the adult liver and they usually display limitedfunctionality when compared with their in vivo counterparts [121–124]. Therefore developing robust protocols of differentiationremains a major challenge to deliver the clinical promises ofhESCs. However, this issue can be solved by directly using theprogenitors already available as shown with pancreatic cells (seeabove) and/or by increasing our understanding of the mechanismscontrolling organ maturation. This last approach is currently usedsuccessfully for a large number of cell types and especially withneural cells.

Indeed, protocols currently available to generate functionalneurons, retina cells or OPCs have strongly benefited fromknowledge generated by developmental biologists concerningneural tube patterning and neuronal specification [135,136].Therefore these protocols generally respect key stages ofdevelopment and require extended periods of time of in vitroculture (between 45 days and up to 3 months) to generatefunctional cell types. In addition, the availability of animalmodels for specific neurodegenerative diseases and the existenceof robust in vitro functional tests allowed validation of thecells generated. For example, transplantation of ESC-deriveddopaminergic neurons appeared to restore motor function in

c© The Authors Journal compilation c© 2010 Biochemical Society

16 A. K. K. Teo and L. Vallier

Figure 2 Human pluripotent stem cells for cell-based therapy

hESCs can be derived from the inner cell mass of the human blastocyst, whereas hiPSCs can be derived via reprogramming of human somatic cells. These human pluripotent stem cells can bedifferentiated into clinically useful cell types, such as neural cells, retinal cells, cardiomyocytes, hepatocytes and pancreatic β-cells, and be used for cell replacement therapy.

animal models of PD [114], and studies have shown thathESCs can be differentiated into retinal pigment cells whichcan restore retinal structure and function in animal models ofretinal degeneration [137]. Finally, OPCs generated from hESCshave been transplanted into animal models of spinal cord injuryand the transplanted cells appear not only to recolonize thescarred area, but also to promote the recruitment of endogenousstem cells, which ultimately results in restoration of mobility[117,138]. These exciting preliminary findings led to the firsthESC-based clinical trial for treatment of spinal cord injuries byGeron Corporation (Menlo Park, CA, U.S.A.). This clinical trial,despite being put on hold at the moment, represents an importantstep towards clinical applications of human pluripotent stem cellsand the outcome will bring essential information concerningthe feasibility of stem-cell-based therapy for a large number ofdiseases.

Safety issue with hESCs

Safety issues continue to be the main limitation for the useof hESCs in clinics, and teratoma formation represents oneof the most difficult challenges (Figure 4). Indeed, hESCs arenaturally tumourous by definition. hESC lines are consideredto be pluripotent based on the capacity to form teratomasonce injected into an immunodeficient host. Therefore thereis a risk of tumour formation if the transplanted cells arecontaminated by residual pluripotent stem cells. Importantly,several reports have shown that this risk is increased whenearly progenitors are used [120,139] and that methods producingheterogeneous populations of cells favour contamination byundifferentiated cells. Hence teratoma formation can easily beavoided using robust methods of differentiation allowing thegeneration of a homogenous population of fully differentiatedcells. A second cause of teratoma formation could be more

problematic. Differentiated cells generated from ESCs could havethe capacity to dedifferentiate into pluripotent stem cells whengrown in a specific environment in vivo. However, there is noevidence that fully differentiated cells generated from hESCs canbe reprogrammed in vivo or in vitro without overexpression ofpluripotency factors. Systematic studies regarding the occurrenceof teratoma formation after transplantation of hESC-derivedsomatic cells could be necessary to completely rule out such ahypothesis.

Graft overgrowth represents another issue associated withprogenitors generated from hESCs. Indeed, the injection of earlyprogenitors in the adult environment can cause uncontrolledproliferation of these progenitors, resulting in a tumour-likestructure incompatible with the function of the transplantedorgan. For example, a recent report has described neuronalprogenitors generated from hESCs to continue proliferating aftertransplantation in the brain, leading to abnormal growth [140].This issue illustrates once again the importance of generatingfully differentiated cell types from hESCs. The issue of tumourformation associated with hESCs is also heightened by theirrelative karyotypic instability [141]. Indeed, hESCs are proneto acquire chromosomal translocations when grown over aprolonged period of time in culture. This is a common featurewith any primary cell culture [142], which can be solved bykaryotyping hESCs regularly and by maintaining robust cultureconditions [143]. However, the most robust method currentlyavailable to expand hESCs involves co-culture with feeder cellsand serum, which are not always fully compatible with clinicalapplications.

To conclude, the risk of tumour formation by hESCs remainsan issue, but it can be easily controlled by using methods ofdifferentiation allowing the generation of homogenous populationof fully differentiated cells. Importantly, technical solutionscan be used to guarantee the absence of pluripotent stem

c© The Authors Journal compilation c© 2010 Biochemical Society

Emerging use of stem cells in regenerative medicine 17

Figure 3 An example of an hESC differentiation protocol

Directed differentiation of hESCs to hepatic progenitors using fully defined culture conditions [175]. hESCs were differentiated into endoderm using activin, BMP4 (bone morphogenetic protein 4),FGF2 and the PI3K (phosphoinositide 3-kinase) inhibitor LY294002. Hepatic specification was then initiated with FGF10, RA (retinoic acid) and an inhibitor of the activin/nodal receptor SB431542.Addition of FGF4, HGF and EGF resulted in maturation of the hepatic progenitors expressing some markers of mature hepatocytes, such as ALB, AFP, AAT (α1-antitrypsin) and CK18.

Figure 4 Roadmap from human stem cells to cell replacement therapy

Some selection criteria for the type of human stem cells (SCs) and the methodology of differentiation prior to approval for clinical use. Thereafter the end-products can be used for cell replacementtherapy or for drug testing. FDA, Federal Drug Administration.

c© The Authors Journal compilation c© 2010 Biochemical Society

18 A. K. K. Teo and L. Vallier

cells in transplanted cells. Methods of purification allowingtransplantation of homogenous populations of differentiatedcells are currently available and are developed by severallaboratories [144–147]. This can be achieved with FACS usingspecific cell-surface markers for pluripotent stem cells and/or fordifferentiated cells. hESCs can also be engineered to contain aselection gene which can be used to avoid the presence of anyresidual undifferentiated cells [148]. Finally, encapsulation ofdifferentiated cells when compatible with their functionality canalso be an advantageous solution. Therefore there are multiplesolutions to increase the safety of cells generated from hESCs,which should accelerate their use in clinic.

HUMAN-INDUCED PLURIPOTENT STEM CELLS

Pluripotent stem cells can also be generated by reprogrammingsomatic cells using either SCNT (somatic cell nuclear transfer)or overexpression of pluripotency factors. However, SCNThas never been performed successfully in humans and thedifficulty in obtaining human oocytes remains a major limitation.The recent possibility to generate hiPSCs from somatic cellsby overexpressing pluripotency factors such as Oct4, Sox2,Klf4 and c-Myc represents a more immediate opportunity forregenerative medicine. Indeed, this approach can be used togenerate hiPSCs from a large number of patients of differentage and gender [149,150]. In addition, a broad number ofsomatic cells can be reprogrammed by the four factors approach,including skin fibroblasts [151], keratinocytes [152], NSCs[153], B-lymphocytes [154], pancreatic β-cells [155], hepatocytesand intestinal cells [156]. Therefore hiPSCs could enablethe production of patient-specific cell types which are fullyimmune-compatible with the original donor, thereby avoiding theuse of immunosuppressive treatment during cell-based therapy(Figure 2). In addition, hiPSCs can be generated from somaticcells isolated from patients suffering from diverse diseases. Then,the resulting ‘diseased’ hiPSCs can be differentiated into the celltype targeted by the disease and thus provide in vitro models toperform large-scale studies impossible with primary cell culture orwith biopsy material. Two recent studies have shown that hiPSCscan be used to study SMA (spinal muscular atrophy) and FD(familial dysautonomia) [156a,156b]. In both studies, ‘diseased’hiPSCs were differentiated into neuronal cells which then displayphenotype characteristic of cells from patients with SMA or FD.Therefore these culture systems provide, for the first time, anin vitro model to study these complex diseases and also to screenpharmaceutical compounds in vitro using relevant tissues. HencehiPSCs represent a unique opportunity to develop new treatmentsfor a large number of diseases which are lacking relevant in vitromodels (Figure 4).

Limitations to in vivo use

The use of hiPSCs in vivo is currently limited by several issues.hiPSCs are commonly generated using viruses overexpressing thefactors necessary to achieve epigenetic reprogramming. Thereforethe resulting hiPSCs contain multiple copies of each transgenein their genome and these insertions can provoke mutationsin key genes, such as oncogenes or cell-cycle regulators. Thisphenomenon has been well described in gene therapy clinicaltrials for SCID (severe combined immunodeficiency) [157].Indeed, several patients developed leukaemia after insertion ofthe lentiviruses in proto-oncogene LMO2 (LIM domain only2) [158,159]. Therefore the use of viruses to generate hiPSCsrepresents a major limitation for their in vivo use. A solutioncould be to sequence the entire genome of each hiPSC to select

lines that only integrate viruses in regions of the genome devoid ofgenes. Alternatively, a large number of groups are now developingnon-integrative methods to generate hiPSCs, including the useof transitory transfection of episomal viruses [160–162]. Thedevelopment of small molecules represents the ultimate solutionto the problems associated with genetic modifications and severalreports have shown that c-Myc and Sox2 could be replacedrespectively, by molecules such as the histone modifier inhibitorBIX-01294 [163] and the TGFβ receptor inhibitor SB431542[164]. However, all of these methods have been used on a limitednumber of somatic cells of embryonic origin and more work isrequired to demonstrate that they can be used with a broad numberof adult patients.

Generation of hiPSCs also require the use of c-Myc and/or theinhibition of the p53 pathway, both of which present safety issues.Indeed, studies in the mouse have shown that reactivation of theexogenous c-Myc transgene can induce tumour formation [165].Recent reports have also shown that inhibition of the p53 pathwaystrongly increases the efficiency of epigenetic reprogramming andthus suggests that the main pathway protecting cells against DNAdamage needs to be turned off to allow the generation of hiPSCs[166–171]. The consequences on the genomic integrity of hiPSCsremain to be demonstrated, but inhibition of p53 could increasethe karyotypic instability already observed with hESCs. Finally,the pluripotent status of hiPSCs confers upon them the capacityto form teratomas in vivo and this safety issue, which is sharedwith hESCs, could be aggravated in hiPSCs. Indeed, a recentreport has shown that the capacity of iPSCs to form teratomascan vary in function of their origin [172]. This phenomenon couldbe explained by the persistence of undifferentiated cells afterdifferentiation, suggesting that some iPSC lines are more difficultto differentiate than others. Alternatively, differentiated cellsgenerated from iPSCs could re-acquire a pluripotent status aftertransplantation in vivo. Therefore further studies are necessary tofully demonstrate that differentiated cells generated from iPSCsare epigenetically stable over a prolonged period of time in vitroand in vivo.

Generation of fully differentiated cells

hiPSCs and hESCs share many characteristics, including theircapacity of differentiation, the expression of pluripotency markersand their dependence on activin/nodal signalling to maintaintheir pluripotent status [173]. These functional similarities alsoimply that hiPSCs and hESCs suffer from the same limitations,which include the risk of teratoma formation (see above),variability in their capacity to differentiate and difficulty ingenerating fully functional cells. Moreover, incomplete epigeneticreprogramming, epigenetic memory, transgene insertion andgenetic instability could worsen the variability between hiPSCsand thus render its use almost impossible for personalized therapy.Indeed, each hiPSC line could require the development of aspecific method of differentiation. Consequently, further studiesare needed to definitively demonstrate that hiPSCs generated froma broad number of patients can all be efficiently differentiatedinto one particular cell type using a unique protocol. In addition,the absence of robust protocols to generate fully differentiatedcells from hiPSCs is currently limiting the development ofin vitro models of disease. Indeed, the cells generated fromdiseased hiPSCs display features of embryonic progenitors whichcannot be used to mimic disease only occurring in the adult[174]. Therefore much more effort will be required to deliverthe clinical promises of hiPSCs. However, this major objectivecould be achieved very rapidly since hiPSCs have become themain focus of many laboratories and they will plainly benefit

c© The Authors Journal compilation c© 2010 Biochemical Society

Emerging use of stem cells in regenerative medicine 19

from the experience accumulated over the past 10 years withhESCs.

CONCLUSIONS AND PERSPECTIVES

The increasing types of stem cells available represent a uniqueopportunity for regenerative medicine as they will offer newtreatment for a broad number of diseases which cannot be fullycured by drug-based therapy. Importantly, stem cells are alreadyreaching the clinic as shown by the large number of clinical trialsusing diverse types of stem cells. However, stem cells presentnew challenges that will need to be solved to fully realise theirclinical promises. The broad diversity of stem cells available isnow offering a large choice for clinicians and a universal stemcell for ‘all the diseases’ is unlikely to exist. Therefore, the‘right’ stem cell will have to be used for the ‘right’ disease. Inaddition, safety could block or delay a large number of clinicalapplications. Indeed, the necessity to demonstrate that a stemcell is not harmful represents a necessary hurdle, especially withpluripotent stem cells capable of forming teratomas. Thereforeregulatory agencies will have to define new criteria to evaluatethe risk associated with specific stem cells and their differentiatedprogeny. This process is often delayed by the difficulty to definewhat is an acceptable risk/benefit with stem-cell-based therapy. Isteratoma an acceptable risk compared with a terminal disease? Inaddition, novel animal models are urgently needed to reinforcethe work currently performed in rodents. Indeed, the mousepresents clear advantages but also important limitations. Theirsize is not relevant for human treatment and mouse modelsrarely reproduce, in full, the human disease. Finally, monitoringfunctionality of human cells in a fully immunosuppressed xeno-environment present obvious issues. Therefore the use of largeanimals or non-human primates will be necessary to validate thefunctionality and safety of the cells generated from hESCs/hiPSCsand also other stem cells. To conclude, clinical applications ofstem cells are becoming a reality and future applications willcontinue to require a strong collaboration between basic scientists,clinicians, regulatory agencies and, more importantly, patients,whose support is essential.

ACKNOWLEDGEMENTS

We thank Dr Tamir Rashid for helpful discussions, Ms Stephanie Brown for proof-readingprior to submission, and Dr Nicholas Hannan for providing the immunostaining figures.

FUNDING

A. K. K. T. is supported by A*STAR Graduate Academy of A*STAR (Agency for Science,Technology and Research), Singapore, and L.V. is supported by the Medical ResearchCouncil (U.K.).

REFERENCES

1 Burt, R. K., Loh, Y., Pearce, W., Beohar, N., Barr, W. G., Craig, R., Wen, Y., Rapp, J. A.and Kessler, J. (2008) Clinical applications of blood-derived and marrow-derived stemcells for nonmalignant diseases. JAMA, J. Am. Med. Assoc. 299, 925–936

2 Baetge, E. E. (2008) Production of β-cells from human embryonic stem cells. DiabetesObes. Metab. 10 (Suppl. 4) 186–194

3 Seydoux, G. and Braun, R. E. (2006) Pathway to totipotency: lessons from germ cells.Cell 127, 891–904

4 Yu, J. and Thomson, J. A. (2008) Pluripotent stem cell lines. Genes Dev. 22,1987–1997

5 Martin, G. R. (1981) Isolation of a pluripotent cell line from early mouse embryoscultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl. Acad. Sci.U.S.A. 78, 7634–7638

6 Evans, M. J. and Kaufman, M. H. (1981) Establishment in culture of pluripotential cellsfrom mouse embryos. Nature 292, 154–156

7 Clark, A. T., Bodnar, M. S., Fox, M., Rodriquez, R. T., Abeyta, M. J., Firpo, M. T. andPera, R. A. (2004) Spontaneous differentiation of germ cells from human embryonicstem cells in vitro. Hum. Mol. Genet. 13, 727–739

8 Xu, R. H., Chen, X., Li, D. S., Li, R., Addicks, G. C., Glennon, C., Zwaka, T. P. andThomson, J. A. (2002) BMP4 initiates human embryonic stem cell differentiation totrophoblast. Nat. Biotechnol. 20, 1261–1264

9 Aranguren, X. L., McCue, J. D., Hendrickx, B., Zhu, X. H., Du, F., Chen, E., Pelacho, B.,Penuelas, I., Abizanda, G., Uriz, M. et al. (2008) Multipotent adult progenitor cellssustain function of ischemic limbs in mice. J. Clin. Invest. 118, 505–514

10 Jiang, Y., Jahagirdar, B. N., Reinhardt, R. L., Schwartz, R. E., Keene, C. D.,Ortiz-Gonzalez, X. R., Reyes, M., Lenvik, T., Lund, T., Blackstad, M. et al. (2002)Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418, 41–49

11 van der Flier, L. G. and Clevers, H. (2009) Stem cells, self-renewal, and differentiationin the intestinal epithelium. Annu. Rev. Physiol. 71, 241–260

12 Blanpain, C. and Fuchs, E. (2009) Epidermal homeostasis: a balancing act of stem cellsin the skin. Nat. Rev. Mol. Cell. Biol. 10, 207–217

13 Gaudio, E., Carpino, G., Cardinale, V., Franchitto, A., Onori, P. and Alvaro, D. (2009)New insights into liver stem cells. Digest. Liver Dis. 41, 455–462

14 Pacilli, A. and Pasquinelli, G. (2009) Vascular wall resident progenitor cells: a review.Exp. Cell Res. 315, 901–914

15 Yin, T. and Li, L. (2006) The stem cell niches in bone. J. Clin. Invest. 116, 1195–120116 Doetsch, F. (2003) A niche for adult neural stem cells. Curr. Opin. Genet. Dev. 13,

543–55017 Lederer, C. W. and Santama, N. (2008) Neural stem cells: mechanisms of fate

specification and nuclear reprogramming in regenerative medicine. Biotechnol. J. 3,1521–1538

18 Bickenbach, J. R. and Grinnell, K. L. (2004) Epidermal stem cells: interactions indevelopmental environments. Differentiation 72, 371–380

19 Bickenbach, J. R., Stern, M. M., Grinnell, K. L., Manuel, A. and Chinnathambi, S.(2006) Epidermal stem cells have the potential to assist in healing damaged tissues. J.Investig. Dermatol. Symp. Proc. 11, 118–123

20 Beltrami, A. P., Barlucchi, L., Torella, D., Baker, M., Limana, F., Chimenti, S., Kasahara,H., Rota, M., Musso, E., Urbanek, K. et al. (2003) Adult cardiac stem cells aremultipotent and support myocardial regeneration. Cell 114, 763–776

21 Messina, E., De Angelis, L., Frati, G., Morrone, S., Chimenti, S., Fiordaliso, F., Salio,M., Battaglia, M., Latronico, M. V., Coletta, M. et al. (2004) Isolation and expansion ofadult cardiac stem cells from human and murine heart. Circ. Res. 95, 911–921

22 Martin, C. M., Russell, J. L., Ferdous, A. and Garry, D. J. (2006) Molecular signaturesdefine myogenic stem cell populations. Stem Cell Rev. 2, 37–42

23 Shi, X. and Garry, D. J. (2006) Muscle stem cells in development, regeneration, anddisease. Genes Dev. 20, 1692–1708

24 Neuringer, I. P. and Randell, S. H. (2004) Stem cells and repair of lung injuries. Respir.Res. 5, 6

25 Neuringer, I. P. and Randell, S. H. (2006) Lung stem cell update: promise andcontroversy. Monaldi Arch. Chest Dis. 65, 47–51

26 Casali, A. and Batlle, E. (2009) Intestinal stem cells in mammals and Drosophila. CellStem Cell 4, 124–127

27 Kung, J. W. and Forbes, S. J. (2009) Stem cells and liver repair. Curr. Opin. Biotechnol.20, 568–574

28 Ochiya, T., Yamamoto, Y. and Banas, A. (2009) Commitment of stem cells intofunctional hepatocytes. Differentiation 79, 65–73

29 Cameron, H. A., Hazel, T. G. and McKay, R. D. (1998) Regulation of neurogenesis bygrowth factors and neurotransmitters. J. Neurobiol. 36, 287–306

30 Androutsellis-Theotokis, A., Rueger, M. A., Mkhikian, H., Korb, E. and McKay, R. D.(2008) Signaling pathways controlling neural stem cells slow progressive braindisease. Cold Spring Harb. Symp. Quant. Biol. 73, 403–410

31 Kuhn, H. G., Winkler, J., Kempermann, G., Thal, L. J. and Gage, F. H. (1997) Epidermalgrowth factor and fibroblast growth factor-2 have different effects on neural progenitorsin the adult rat brain. J. Neurosci. 17, 5820–5829

32 Fallon, J., Reid, S., Kinyamu, R., Opole, I., Opole, R., Baratta, J., Korc, M., Endo, T. L.,Duong, A., Nguyen, G. et al. (2000) In vivo induction of massive proliferation, directedmigration, and differentiation of neural cells in the adult mammalian brain. Proc. Natl.Acad. Sci. U.S.A. 97, 14686–14691

33 Jin, K., Zhu, Y., Sun, Y., Mao, X. O., Xie, L. and Greenberg, D. A. (2002) Vascularendothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proc.Natl. Acad. Sci. U.S.A. 99, 11946–11950

34 Cao, L., Jiao, X., Zuzga, D. S., Liu, Y., Fong, D. M., Young, D. and During, M. J. (2004)VEGF links hippocampal activity with neurogenesis, learning and memory. Nat. Genet.36, 827–835

c© The Authors Journal compilation c© 2010 Biochemical Society

20 A. K. K. Teo and L. Vallier

35 Jin, K., Mao, X. O., Sun, Y., Xie, L. and Greenberg, D. A. (2002) Stem cell factorstimulates neurogenesis in vitro and in vivo. J. Clin. Invest. 110, 311–319

36 Aberg, M. A., Aberg, N. D., Hedbacker, H., Oscarsson, J. and Eriksson, P. S. (2000)Peripheral infusion of IGF-I selectively induces neurogenesis in the adult rathippocampus. J. Neurosci. 20, 2896–2903

37 Rosenthal, N., Santini, M. P. and Musaro, A. (2006) Growth factor enhancement ofcardiac regeneration. Cell Transplant. 15 (Suppl. 1), S41–S45

38 Mourkioti, F. and Rosenthal, N. (2005) IGF-1, inflammation and stem cells: interactionsduring muscle regeneration. Trends Immunol. 26, 535–542

39 Mourkioti, F. and Rosenthal, N. (2008) NF-κB signaling in skeletal muscle: prospectsfor intervention in muscle diseases. J. Mol. Med. 86, 747–759

40 Chen, S. E., Gerken, E., Zhang, Y., Zhan, M., Mohan, R. K., Li, A. S., Reid, M. B. and Li,Y. P. (2005) Role of TNF-α signaling in regeneration of cardiotoxin-injured muscle.Am. J. Physiol. Cell Physiol. 289, C1179–C1187

41 Acharyya, S., Villalta, S. A., Bakkar, N., Bupha-Intr, T., Janssen, P. M., Carathers, M., Li,Z. W., Beg, A. A., Ghosh, S., Sahenk, Z. et al. (2007) Interplay of IKK/NF-κB signalingin macrophages and myofibers promotes muscle degeneration in Duchenne musculardystrophy. J. Clin. Invest. 117, 889–901

42 Dor, Y., Brown, J., Martinez, O. I. and Melton, D. A. (2004) Adult pancreatic β-cells areformed by self-duplication rather than stem-cell differentiation. Nature 429,41–46

43 Teta, M., Rankin, M. M., Long, S. Y., Stein, G. M. and Kushner, J. A. (2007) Growth andregeneration of adult β cells does not involve specialized progenitors. Dev. Cell 12,817–826

44 Xia, B., Zhan, X. R., Yi, R. and Yang, B. (2009) Can pancreatic duct-derived progenitorsbe a source of islet regeneration? Biochem. Biophys. Res. Commun. 383, 383–385

45 Solar, M., Cardalda, C., Houbracken, I., Martin, M., Maestro, M. A., De Medts, N.,Xu, X., Grau, V., Heimberg, H., Bouwens, L. and Ferrer, J. (2009) Pancreatic exocrineduct cells give rise to insulin-producing β cells during embryogenesis but not afterbirth. Dev. Cell 17, 849–860

46 Fausto, N. (2004) Liver regeneration and repair: hepatocytes, progenitor cells, andstem cells. Hepatology 39, 1477–1487

47 Shafritz, D. A., Oertel, M., Menthena, A., Nierhoff, D. and Dabeva, M. D. (2006) Liverstem cells and prospects for liver reconstitution by transplanted cells. Hepatology 43,S89–S98

48 Malhi, H., Irani, A. N., Gagandeep, S. and Gupta, S. (2002) Isolation of humanprogenitor liver epithelial cells with extensive replication capacity and differentiationinto mature hepatocytes. J. Cell Sci. 115, 2679–2688

49 Wang, X., Foster, M., Al-Dhalimy, M., Lagasse, E., Finegold, M. and Grompe, M.(2003) The origin and liver repopulating capacity of murine oval cells. Proc. Natl. Acad.Sci. U.S.A. 100 (Suppl. 1), 11881–11888

50 Dan, Y. Y., Riehle, K. J., Lazaro, C., Teoh, N., Haque, J., Campbell, J. S. and Fausto, N.(2006) Isolation of multipotent progenitor cells from human fetal liver capable ofdifferentiating into liver and mesenchymal lineages. Proc. Natl. Acad. Sci. U.S.A. 103,9912–9917

51 Nagy, P., Bisgaard, H. C. and Thorgeirsson, S. S. (1994) Expression of hepatictranscription factors during liver development and oval cell differentiation. J. Cell Biol.126, 223–233

52 Dan, Y. Y. and Yeoh, G. C. (2008) Liver stem cells: a scientific and clinical perspective.J. Gastroenterol. Hepatol. 23, 687–698

53 Lowes, K. N., Brennan, B. A., Yeoh, G. C. and Olynyk, J. K. (1999) Oval cell numbers inhuman chronic liver diseases are directly related to disease severity. Am. J. Pathol.154, 537–541

54 Roskams, T., Yang, S. Q., Koteish, A., Durnez, A., DeVos, R., Huang, X., Achten, R.,Verslype, C. and Diehl, A. M. (2003) Oxidative stress and oval cell accumulation inmice and humans with alcoholic and nonalcoholic fatty liver disease. Am. J. Pathol.163, 1301–1311

55 Herrera, M. B., Bruno, S., Buttiglieri, S., Tetta, C., Gatti, S., Deregibus, M. C., Bussolati,B. and Camussi, G. (2006) Isolation and characterization of a stem cell population fromadult human liver. Stem Cells 24, 2840–2850

56 Schmelzer, E., Wauthier, E. and Reid, L. M. (2006) The phenotypes of pluripotenthuman hepatic progenitors. Stem Cells 24, 1852–1858

57 Schmelzer, E., Zhang, L., Bruce, A., Wauthier, E., Ludlow, J., Yao, H. L., Moss, N.,Melhem, A., McClelland, R., Turner, W. et al. (2007) Human hepatic stem cells fromfetal and postnatal donors. J. Exp. Med. 204, 1973–1987

58 Theise, N. D., Saxena, R., Portmann, B. C., Thung, S. N., Yee, H., Chiriboga, L., Kumar,A. and Crawford, J. M. (1999) The canals of Hering and hepatic stem cells in humans.Hepatology 30, 1425–1433

59 Bhindi, R., Witting, P. K., McMahon, A. C., Khachigian, L. M. and Lowe, H. C. (2006)Rat models of myocardial infarction. Pathogenetic insights and clinical relevance.Thromb. Haemostasis 96, 602–610

60 DiBernardo, A. B. and Cudkowicz, M. E. (2006) Translating preclinical insights intoeffective human trials in ALS. Biochim. Biophys. Acta 1762, 1139–1149

61 Rivera, J. and Tessarollo, L. (2008) Genetic background and the dilemma of translatingmouse studies to humans. Immunity 28, 1–4

62 Hanley, N. A., Hanley, K. P., Miettinen, P. J. and Otonkoski, T. (2008) Weighing upβ-cell mass in mice and humans: self-renewal, progenitors or stem cells? Mol. Cell.Endocrinol. 288, 79–85

63 Ausoni, S. and Sartore, S. (2009) From fish to amphibians to mammals: in search ofnovel strategies to optimize cardiac regeneration. J. Cell Biol. 184, 357–364

64 Kuci, S., Kuci, Z., Latifi-Pupovci, H., Niethammer, D., Handgretinger, R., Schumm, M.,Bruchelt, G., Bader, P. and Klingebiel, T. (2009) Adult stem cells as an alternativesource of multipotential (pluripotential) cells in regenerative medicine. Curr. Stem CellRes. Ther. 4, 107–117

65 Gage, F. H. (2000) Mammalian neural stem cells. Science 287, 1433–143866 Goldman, S. A. and Windrem, M. S. (2006) Cell replacement therapy in neurological

disease. Philos. Trans. R. Soc. London Ser. B 361, 1463–147567 Bernardo, M. E., Locatelli, F. and Fibbe, W. E. (2009) Mesenchymal stromal cells. Ann.

N.Y. Acad. Sci. 1176, 101–11768 Prockop, D. J. (2009) Repair of tissues by adult stem/progenitor cells (MSCs):

controversies, myths, and changing paradigms. Mol. Ther. 17, 939–94669 Morshead, C. M., Reynolds, B. A., Craig, C. G., McBurney, M. W., Staines, W. A.,

Morassutti, D., Weiss, S. and van der Kooy, D. (1994) Neural stem cells in the adultmammalian forebrain: a relatively quiescent subpopulation of subependymal cells.Neuron 13, 1071–1082

70 Reynolds, B. A. and Weiss, S. (1992) Generation of neurons and astrocytes fromisolated cells of the adult mammalian central nervous system. Science 255,1707–1710

71 Song, H., Stevens, C. F. and Gage, F. H. (2002) Astroglia induce neurogenesis fromadult neural stem cells. Nature 417, 39–44

72 Song, H. J., Stevens, C. F. and Gage, F. H. (2002) Neural stem cells from adulthippocampus develop essential properties of functional CNS neurons. Nat. Neurosci.5, 438–445

73 Taupin, P. (2006) Therapeutic potential of adult neural stem cells. Recent Pat. CNSDrug Discov. 1, 299–303

74 Kondziolka, D., Wechsler, L., Goldstein, S., Meltzer, C., Thulborn, K. R., Gebel, J.,Jannetta, P., DeCesare, S., Elder, E. M., McGrogan, M. et al. (2000) Transplantation ofcultured human neuronal cells for patients with stroke. Neurology 55, 565–569

75 Meltzer, C. C., Kondziolka, D., Villemagne, V. L., Wechsler, L., Goldstein, S., Thulborn,K. R., Gebel, J., Elder, E. M., DeCesare, S. and Jacobs, A. (2001) Serial [18F]fluorodeoxyglucose positron emission tomography after human neuronal implantationfor stroke. Neurosurgery 49, 586–591

76 Choi, C. Q. (2007) A stroke for stem cells. Sci. Am. 296, 8–977 Amariglio, N., Hirshberg, A., Scheithauer, B. W., Cohen, Y., Loewenthal, R.,

Trakhtenbrot, L., Paz, N., Koren-Michowitz, M., Waldman, D., Leider-Trejo, L. et al.(2009) Donor-derived brain tumor following neural stem cell transplantation in anataxia telangiectasia patient. PLoS Med. 6, e1000029

78 Friedenstein, A. J., Petrakova, K. V., Kurolesova, A. I. and Frolova, G. P. (1968)Heterotopic of bone marrow. Analysis of precursor cells for osteogenic andhematopoietic tissues. Transplantation 6, 230–247

79 De Coppi, P., Bartsch, Jr, G., Siddiqui, M. M., Xu, T., Santos, C. C., Perin, L.,Mostoslavsky, G., Serre, A. C., Snyder, E. Y., Yoo, J. J. et al. (2007) Isolation ofamniotic stem cell lines with potential for therapy. Nat. Biotechnol. 25, 100–106

80 In ‘t Anker, P. S., Scherjon, S. A., Kleijburg-van der Keur, C., de Groot-Swings, G. M.,Claas, F. H., Fibbe, W. E. and Kanhai, H. H. (2004) Isolation of mesenchymal stem cellsof fetal or maternal origin from human placenta. Stem Cells 22, 1338–1345

81 Chien, C. C., Yen, B. L., Lee, F. K., Lai, T. H., Chen, Y. C., Chan, S. H. and Huang, H. I.(2006) In vitro differentiation of human placenta-derived multipotent cells intohepatocyte-like cells. Stem Cells 24, 1759–1768

82 Zuk, P. A., Zhu, M., Ashjian, P., De Ugarte, D. A., Huang, J. I., Mizuno, H., Alfonso,Z. C., Fraser, J. K., Benhaim, P. and Hedrick, M. H. (2002) Human adipose tissue is asource of multipotent stem cells. Mol. Biol. Cell 13, 4279–4295

83 Zuk, P. A., Zhu, M., Mizuno, H., Huang, J., Futrell, J. W., Katz, A. J., Benhaim, P.,Lorenz, H. P. and Hedrick, M. H. (2001) Multilineage cells from human adipose tissue:implications for cell-based therapies. Tissue Eng. 7, 211–228

84 Bieback, K., Kern, S., Kluter, H. and Eichler, H. (2004) Critical parameters for theisolation of mesenchymal stem cells from umbilical cord blood. Stem Cells 22,625–634

85 Tyndall, A. and Uccelli, A. (2009) Multipotent mesenchymal stromal cells forautoimmune diseases: teaching new dogs old tricks. Bone Marrow Transplant. 43,821–828

86 Karp, J. M. and Leng Teo, G. S. (2009) Mesenchymal stem cell homing: the devil is inthe details. Cell Stem Cell 4, 206–216

c© The Authors Journal compilation c© 2010 Biochemical Society

Emerging use of stem cells in regenerative medicine 21

87 Pittenger, M. F., Mackay, A. M., Beck, S. C., Jaiswal, R. K., Douglas, R., Mosca, J. D.,Moorman, M. A., Simonetti, D. W., Craig, S. and Marshak, D. R. (1999) Multilineagepotential of adult human mesenchymal stem cells. Science 284, 143–147

88 in ‘t Anker, P. S., Noort, W. A., Scherjon, S. A., Kleijburg-van der Keur, C.,Kruisselbrink, A. B., van Bezooijen, R. L., Beekhuizen, W., Willemze, R., Kanhai, H. H.and Fibbe, W. E. (2003) Mesenchymal stem cells in human second-trimester bonemarrow, liver, lung, and spleen exhibit a similar immunophenotype but aheterogeneous multilineage differentiation potential. Haematologica 88, 845–852

89 Sekiya, I., Colter, D. C. and Prockop, D. J. (2001) BMP-6 enhances chondrogenesis ina subpopulation of human marrow stromal cells. Biochem. Biophys. Res. Commun.284, 411–418

90 Indrawattana, N., Chen, G., Tadokoro, M., Shann, L. H., Ohgushi, H., Tateishi, T.,Tanaka, J. and Bunyaratvej, A. (2004) Growth factor combination for chondrogenicinduction from human mesenchymal stem cell. Biochem. Biophys. Res. Commun.320, 914–919

91 Sueblinvong, V., Loi, R., Eisenhauer, P. L., Bernstein, I. M., Suratt, B. T., Spees, J. L.and Weiss, D. J. (2008) Derivation of lung epithelium from human cord blood-derivedmesenchymal stem cells. Am. J. Respir. Crit. Care Med. 177, 701–711

92 Yue, W. M., Liu, W., Bi, Y. W., He, X. P., Sun, W. Y., Pang, X. Y., Gu, X. H. and Wang,X. P. (2008) Mesenchymal stem cells differentiate into an endothelial phenotype,reduce neointimal formation, and enhance endothelial function in a rat vein graftingmodel. Stem Cells Dev. 17, 785–793

93 Greco, S. J. and Rameshwar, P. (2007) Enhancing effect of IL-1α on neurogenesis fromadult human mesenchymal stem cells: implication for inflammatory mediators inregenerative medicine. J. Immunol. 179, 3342–3350

94 Koga, H., Muneta, T., Nagase, T., Nimura, A., Ju, Y. J., Mochizuki, T. and Sekiya, I.(2008) Comparison of mesenchymal tissues-derived stem cells for in vivochondrogenesis: suitable conditions for cell therapy of cartilage defects in rabbit. CellTissue Res. 333, 207–215

95 Digirolamo, C. M., Stokes, D., Colter, D., Phinney, D. G., Class, R. and Prockop, D. J.(1999) Propagation and senescence of human marrow stromal cells in culture: asimple colony-forming assay identifies samples with the greatest potential to propagateand differentiate. Br. J. Haematol. 107, 275–281

96 Li, H., Fan, X., Kovi, R. C., Jo, Y., Moquin, B., Konz, R., Stoicov, C., Kurt-Jones, E.,Grossman, S. R., Lyle, S. et al. (2007) Spontaneous expression of embryonic factorsand point mutations in aged mesenchymal stem cells: a model of age-relatedtumorigenesis in mice. Cancer Res. 67, 10889–10898

97 Togel, F., Weiss, K., Yang, Y., Hu, Z., Zhang, P. and Westenfelder, C. (2007)Vasculotropic, paracrine actions of infused mesenchymal stem cells are important to therecovery from acute kidney injury. Am. J. Physiol. Renal Physiol. 292, F1626–F1635

98 Spitkovsky, D. and Hescheler, J. (2008) Adult mesenchymal stromal stem cells fortherapeutic applications. Minim. Invasive Ther. Allied Technol. 17, 79–90

99 Koc, O. N., Gerson, S. L., Cooper, B. W., Dyhouse, S. M., Haynesworth, S. E., Caplan,A. I. and Lazarus, H. M. (2000) Rapid hematopoietic recovery after coinfusion ofautologous-blood stem cells and culture-expanded marrow mesenchymal stem cells inadvanced breast cancer patients receiving high-dose chemotherapy. J. Clin. Oncol. 18,307–316

100 Le Blanc, K., Rasmusson, I., Sundberg, B., Gotherstrom, C., Hassan, M., Uzunel, M.and Ringden, O. (2004) Treatment of severe acute graft-versus-host disease with thirdparty haploidentical mesenchymal stem cells. Lancet 363, 1439–1441

101 Bocelli-Tyndall, C., Bracci, L., Spagnoli, G., Braccini, A., Bouchenaki, M., Ceredig, R.,Pistoia, V., Martin, I. and Tyndall, A. (2007) Bone marrow mesenchymal stromal cells(BM-MSCs) from healthy donors and auto-immune disease patients reduce theproliferation of autologous- and allogeneic-stimulated lymphocytes in vitro.Rheumatology 46, 403–408

102 Mazzini, L., Ferrero, I., Luparello, V., Rustichelli, D., Gunetti, M., Mareschi, K., Testa,L., Stecco, A., Tarletti, R., Miglioretti, M. et al. (2009) Mesenchymal stem celltransplantation in amyotrophic lateral sclerosis: a Phase I clinical trial. Exp, Neurol.,doi:10.1016/j.expneurol.2009.08.007

103 Psaltis, P. J., Zannettino, A. C., Worthley, S. G. and Gronthos, S. (2008) Concisereview: mesenchymal stromal cells: potential for cardiovascular repair. Stem Cells 26,2201–2210

104 Yi, B. A., Wernet, O. and Chien, K. R. (2010) Pregenerative medicine: developmentalparadigms in the biology of cardiovascular regeneration. J. Clin. Invest. 120, 20–28

105 Schwartz, R. E., Reyes, M., Koodie, L., Jiang, Y., Blackstad, M., Lund, T., Lenvik, T.,Johnson, S., Hu, W. S. and Verfaillie, C. M. (2002) Multipotent adult progenitor cellsfrom bone marrow differentiate into functional hepatocyte-like cells. J. Clin. Invest.109, 1291–1302

106 Sato, Y., Araki, H., Kato, J., Nakamura, K., Kawano, Y., Kobune, M., Sato, T., Miyanishi,K., Takayama, T., Takahashi, M. et al. (2005) Human mesenchymal stem cellsxenografted directly to rat liver are differentiated into human hepatocytes withoutfusion. Blood 106, 756–763

107 Hong, S. H., Gang, E. J., Jeong, J. A., Ahn, C., Hwang, S. H., Yang, I. H., Park, H. K.,Han, H. and Kim, H. (2005) In vitro differentiation of human umbilical cordblood-derived mesenchymal stem cells into hepatocyte-like cells. Biochem. Biophys.Res. Commun. 330, 1153–1161

108 Banas, A., Teratani, T., Yamamoto, Y., Tokuhara, M., Takeshita, F., Quinn, G., Okochi, H.and Ochiya, T. (2007) Adipose tissue-derived mesenchymal stem cells as a source ofhuman hepatocytes. Hepatology 46, 219–228

109 Banas, A., Teratani, T., Yamamoto, Y., Tokuhara, M., Takeshita, F., Osaki, M., Kato, T.,Okochi, H. and Ochiya, T. (2009) Rapid hepatic fate specification of adipose-derivedstem cells and their therapeutic potential for liver failure. J. Gastroenterol. Hepatol. 24,70–77

110 Kharaziha, P., Hellstrom, P. M., Noorinayer, B., Farzaneh, F., Aghajani, K., Jafari, F.,Telkabadi, M., Atashi, A., Honardoost, M., Zali, M. R. and Soleimani, M. (2009)Improvement of liver function in liver cirrhosis patients after autologous mesenchymalstem cell injection: a phase I-II clinical trial. Eur. J. Gastroenterol. Hepatol. 21,1199–1205

111 Thomson, J. A., Itskovitz-Eldor, J., Shapiro, S. S., Waknitz, M. A., Swiergiel, J. J.,Marshall, V. S. and Jones, J. M. (1998) Embryonic stem cell lines derived from humanblastocysts. Science 282, 1145–1147

112 Klimanskaya, I., Hipp, J., Rezai, K. A., West, M., Atala, A. and Lanza, R. (2004)Derivation and comparative assessment of retinal pigment epithelium from humanembryonic stem cells using transcriptomics. Cloning Stem Cells 6, 217–245

113 Lund, R. D., Wang, S., Klimanskaya, I., Holmes, T., Ramos-Kelsey, R., Lu, B., Girman,S., Bischoff, N., Sauve, Y. and Lanza, R. (2006) Human embryonic stem cell-derivedcells rescue visual function in dystrophic RCS rats. Cloning Stem Cells 8, 189–199

114 Takagi, Y., Takahashi, J., Saiki, H., Morizane, A., Hayashi, T., Kishi, Y., Fukuda, H.,Okamoto, Y., Koyanagi, M., Ideguchi, M. et al. (2005) Dopaminergic neurons generatedfrom monkey embryonic stem cells function in a Parkinson primate model. J. Clin.Invest. 115, 102–109

115 Wichterle, H., Lieberam, I., Porter, J. A. and Jessell, T. M. (2002) Directeddifferentiation of embryonic stem cells into motor neurons. Cell 110, 385–397

116 Li, X. J., Du, Z. W., Zarnowska, E. D., Pankratz, M., Hansen, L. O., Pearce, R. A. andZhang, S. C. (2005) Specification of motoneurons from human embryonic stem cells.Nat. Biotechnol. 23, 215–221

117 Nistor, G. I., Totoiu, M. O., Haque, N., Carpenter, M. K. and Keirstead, H. S. (2005)Human embryonic stem cells differentiate into oligodendrocytes in high purity andmyelinate after spinal cord transplantation. Glia 49, 385–396

118 Dai, W., Field, L. J., Rubart, M., Reuter, S., Hale, S. L., Zweigerdt, R., Graichen, R. E.,Kay, G. L., Jyrala, A. J., Colman, A. et al. (2007) Survival and maturation of humanembryonic stem cell-derived cardiomyocytes in rat hearts. J. Mol. Cell. Cardiol. 43,504–516

119 Caspi, O., Huber, I., Kehat, I., Habib, M., Arbel, G., Gepstein, A., Yankelson, L., Aronson,D., Beyar, R. and Gepstein, L. (2007) Transplantation of human embryonic stemcell-derived cardiomyocytes improves myocardial performance in infarcted rat hearts.J. Am. Coll. Cardiol. 50, 1884–1893

120 Kroon, E., Martinson, L. A., Kadoya, K., Bang, A. G., Kelly, O. G., Eliazer, S., Young, H.,Richardson, M., Smart, N. G., Cunningham, J. et al. (2008) Pancreatic endodermderived from human embryonic stem cells generates glucose-responsiveinsulin-secreting cells in vivo. Nat. Biotechnol. 26, 443–452

121 Chen, Y., Soto-Gutierrez, A., Navarro-Alvarez, N., Rivas-Carrillo, J. D., Yamatsuji, T.,Shirakawa, Y., Tanaka, N., Basma, H., Fox, I. J. and Kobayashi, N. (2006) Instanthepatic differentiation of human embryonic stem cells using activin A and a deletedvariant of HGF. Cell Transplant. 15, 865–871

122 Cai, J., Zhao, Y., Liu, Y., Ye, F., Song, Z., Qin, H., Meng, S., Chen, Y., Zhou, R., Song, X.et al. (2007) Directed differentiation of human embryonic stem cells into functionalhepatic cells. Hepatology 45, 1229–1239

123 Duan, Y., Catana, A., Meng, Y., Yamamoto, N., He, S., Gupta, S., Gambhir, S. S. andZern, M. A. (2007) Differentiation and enrichment of hepatocyte-like cells from humanembryonic stem cells in vitro and in vivo. Stem Cells 25, 3058–3068

124 Oertel, M. and Shafritz, D. A. (2008) Stem cells, cell transplantation and liverrepopulation. Biochim. Biophys. Acta 1782, 61–74

125 Shapiro, A. M., Lakey, J. R., Ryan, E. A., Korbutt, G. S., Toth, E., Warnock, G. L.,Kneteman, N. M. and Rajotte, R. V. (2000) Islet transplantation in seven patients withtype 1 diabetes mellitus using a glucocorticoid-free immunosuppressive regimen. N.Engl. J. Med. 343, 230–238

126 Rajagopal, J., Anderson, W. J., Kume, S., Martinez, O. I. and Melton, D. A. (2003)Insulin staining of ES cell progeny from insulin uptake. Science 299, 363

127 Lumelsky, N., Blondel, O., Laeng, P., Velasco, I., Ravin, R. and McKay, R. (2001)Differentiation of embryonic stem cells to insulin-secreting structures similar topancreatic islets. Science 292, 1389–1394

c© The Authors Journal compilation c© 2010 Biochemical Society

22 A. K. K. Teo and L. Vallier

128 Madsen, O. D. (2005) Stem cells and diabetes treatment. APMIS 113, 858–875129 Docherty, K., Bernardo, A. S. and Vallier, L. (2007) Embryonic stem cell therapy for

diabetes mellitus. Semin. Cell Dev. Biol. 18, 827–838130 D’Amour, K. A., Agulnick, A. D., Eliazer, S., Kelly, O. G., Kroon, E. and Baetge, E. E.

(2005) Efficient differentiation of human embryonic stem cells to definitive endoderm.Nat. Biotechnol. 23, 1534–1541

131 Kubo, A., Shinozaki, K., Shannon, J. M., Kouskoff, V., Kennedy, M., Woo, S., Fehling,H. J. and Keller, G. (2004) Development of definitive endoderm from embryonic stemcells in culture. Development 131, 1651–1662

132 Wang, L. (2006) Endothelial and hematopoietic cell fate of human embryonic stemcells. Trends Cardiovasc. Med. 16, 89–94

133 Bhatia, M. (2007) Hematopoiesis from human embryonic stem cells. Ann. N.Y. Acad.Sci. 1106, 219–222

134 Bhatia, M. (2007) Hematopoietic development from human embryonic stem cells.Hematology Am. Soc. Hematol. Educ. Program, 11–16

135 Caviness, Jr, V. S., Nowakowski, R. S. and Bhide, P. G. (2009) Neocorticalneurogenesis: morphogenetic gradients and beyond. Trends Neurosci. 32, 443–450

136 Tanaka, E. M. and Ferretti, P. (2009) Considering the evolution of regeneration in thecentral nervous system. Nat. Rev. Neurosci. 10, 713–723

137 Vugler, A., Lawrence, J., Walsh, J., Carr, A., Gias, C., Semo, M., Ahmado, A., da Cruz,L., Andrews, P. and Coffey, P. (2007) Embryonic stem cells and retinal repair. Mech.Dev. 124, 807–829

138 Sharp, J., Frame, J., Siegenthaler, M., Nistor, G. and Keirstead, H. S. (2010) Humanembryonic stem cell-derived oligodendrocyte progenitor cell transplants improverecovery after cervical spinal cord injury. Stem Cells 28, 152–163

139 Basma, H., Soto-Gutierrez, A., Yannam, G. R., Liu, L., Ito, R., Yamamoto, T., Ellis, E.,Carson, S. D., Sato, S., Chen, Y. et al. (2009) Differentiation and transplantation ofhuman embryonic stem cell-derived hepatocytes. Gastroenterology. 136, 990–999

140 Aubry, L., Bugi, A., Lefort, N., Rousseau, F., Peschanski, M. and Perrier, A. L. (2008)Striatal progenitors derived from human ES cells mature into DARPP32 neurons in vitroand in quinolinic acid-lesioned rats. Proc. Natl. Acad. Sci. U.S.A. 105, 16707–16712

141 Hanson, C. and Caisander, G. (2005) Human embryonic stem cells and chromosomestability. APMIS 113, 751–755

142 Cervantes, R. B., Stringer, J. R., Shao, C., Tischfield, J. A. and Stambrook, P. J. (2002)Embryonic stem cells and somatic cells differ in mutation frequency and type. Proc.Natl. Acad. Sci. U.S.A. 99, 3586–3590

143 Mitalipova, M. M., Rao, R. R., Hoyer, D. M., Johnson, J. A., Meisner, L. F., Jones, K. L.,Dalton, S. and Stice, S. L. (2005) Preserving the genetic integrity of human embryonicstem cells. Nat. Biotechnol. 23, 19–20

144 Choi, K. D., Vodyanik, M. A. and Slukvin, II. (2009) Generation of mature humanmyelomonocytic cells through expansion and differentiation of pluripotent stemcell-derived lin-CD34+CD43+CD45+ progenitors. J. Clin. Invest. 119, 2818–2829

145 Fong, C. Y., Peh, G. S., Gauthaman, K. and Bongso, A. (2009) Separation of SSEA-4and TRA-1-labelled undifferentiated human embryonic stem cells from aheterogeneous cell population using magnetic-activated cell sorting (MACS) andfluorescence-activated cell sorting (FACS). Stem Cell Rev. Rep. 5, 72–80

146 Choo, A. B., Tan, H. L., Ang, S. N., Fong, W. J., Chin, A., Lo, J., Zheng, L., Hentze, H.,Philp, R. J., Oh, S. K. and Yap, M. (2008) Selection against undifferentiated humanembryonic stem cells by a cytotoxic antibody recognizing podocalyxin-like protein-1.Stem Cells 26, 1454–1463

147 Tan, H. L., Fong, W. J., Lee, E. H., Yap, M. and Choo, A. (2009) mAb 84, a cytotoxicantibody that kills undifferentiated human embryonic stem cells via oncosis. StemCells 27, 1792–1801

148 Schuldiner, M., Itskovitz-Eldor, J. and Benvenisty, N. (2003) Selective ablation ofhuman embryonic stem cells expressing a “suicide” gene. Stem Cells 21, 257–265

149 Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K. andYamanaka, S. (2007) Induction of pluripotent stem cells from adult human fibroblastsby defined factors. Cell 131, 861–872

150 Yu, J., Vodyanik, M. A., Smuga-Otto, K., Antosiewicz-Bourget, J., Frane, J. L., Tian, S.,Nie, J., Jonsdottir, G. A., Ruotti, V., Stewart, R. et al. (2007) Induced pluripotent stemcell lines derived from human somatic cells. Science 318, 1917–1920

151 Lowry, W. E., Richter, L., Yachechko, R., Pyle, A. D., Tchieu, J., Sridharan, R., Clark,A. T. and Plath, K. (2008) Generation of human induced pluripotent stem cells fromdermal fibroblasts. Proc. Natl. Acad. Sci. U.S.A. 105, 2883–2888

152 Aasen, T., Raya, A., Barrero, M. J., Garreta, E., Consiglio, A., Gonzalez, F., Vassena, R.,Bilic, J., Pekarik, V., Tiscornia, G. et al. (2008) Efficient and rapid generation of inducedpluripotent stem cells from human keratinocytes. Nat. Biotechnol. 26, 1276–1284

153 Kim, J. B., Zaehres, H., Wu, G., Gentile, L., Ko, K., Sebastiano, V., Arauzo-Bravo, M. J.,Ruau, D., Han, D. W., Zenke, M. and Scholer, H. R. (2008) Pluripotent stem cellsinduced from adult neural stem cells by reprogramming with two factors. Nature 454,646–650

154 Hanna, J., Markoulaki, S., Schorderet, P., Carey, B. W., Beard, C., Wernig, M.,Creyghton, M. P., Steine, E. J., Cassady, J. P., Foreman, R. et al. (2008) Directreprogramming of terminally differentiated mature B lymphocytes to pluripotency. Cell133, 250–264

155 Stadtfeld, M., Brennand, K. and Hochedlinger, K. (2008) Reprogramming of pancreaticβ cells into induced pluripotent stem cells. Curr. Biol. 18, 890–894

156 Aoi, T., Yae, K., Nakagawa, M., Ichisaka, T., Okita, K., Takahashi, K., Chiba, T. andYamanaka, S. (2008) Generation of pluripotent stem cells from adult mouse liver andstomach cells. Science 321, 699–702

156a Lee, G., Papapetrou, E. P., Kim, H., Chambers, S. M., Tomishima, M. J., Fasano, C. A.,Ganat, Y. M., Menon, J., Shimizu, F., Viale, A. et al. (2009) Modelling pathogenesis andtreatment of familial dysautonomia using patient-specific iPSCs. Nature 461, 402–406

156b Ebert, A. D., Yu, J., Rose, Jr, F. F., Mattis, V. B., Lorson, C. L., Thomson, J. A. andSvendsen, C. N. (2009) Induced pluripotent stem cells from a spinal muscular atrophypatient. Nature 457, 277–280

157 Cassani, B., Montini, E., Maruggi, G., Ambrosi, A., Mirolo, M., Selleri, S., Biral, E.,Frugnoli, I., Hernandez-Trujillo, V., Di Serio, C. et al. (2009) Integration of retroviralvectors induces minor changes in the transcriptional activity of T cells from ADA-SCIDpatients treated with gene therapy. Blood 114, 3546–3556

158 Hacein-Bey-Abina, S., Garrigue, A., Wang, G. P., Soulier, J., Lim, A., Morillon, E.,Clappier, E., Caccavelli, L., Delabesse, E., Beldjord, K. et al. (2008) Insertionaloncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J. Clin.Invest. 118, 3132–3142

159 Howe, S. J., Mansour, M. R., Schwarzwaelder, K., Bartholomae, C., Hubank, M.,Kempski, H., Brugman, M. H., Pike-Overzet, K., Chatters, S. J., de Ridder, D. et al.(2008) Insertional mutagenesis combined with acquired somatic mutations causesleukemogenesis following gene therapy of SCID-X1 patients. J. Clin. Invest. 118,3143–3150

160 Okita, K., Nakagawa, M., Hyenjong, H., Ichisaka, T. and Yamanaka, S. (2008) Generationof mouse induced pluripotent stem cells without viral vectors. Science 322, 949–953

161 Yu, J., Hu, K., Smuga-Otto, K., Tian, S., Stewart, R., Slukvin, I. I. and Thomson, J. A.(2009) Human induced pluripotent stem cells free of vector and transgene sequences.Science 324, 797–801

162 Stadtfeld, M., Nagaya, M., Utikal, J., Weir, G. and Hochedlinger, K. (2008) Inducedpluripotent stem cells generated without viral integration. Science 322, 945–949

163 Shi, Y., Desponts, C., Do, J. T., Hahm, H. S., Scholer, H. R. and Ding, S. (2008)Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4with small-molecule compounds. Cell Stem Cell 3, 568–574

164 Ichida, J. K., Blanchard, J., Lam, K., Son, E. Y., Chung, J. E., Egli, D., Loh, K. M.,Carter, A. C., Di Giorgio, F. P., Koszka, K. et al. (2009) A small-molecule inhibitor oftgf-β signaling replaces sox2 in reprogramming by inducing nanog. Cell Stem Cell 5,491–503

165 Okita, K., Ichisaka, T. and Yamanaka, S. (2007) Generation of germline-competentinduced pluripotent stem cells. Nature 448, 313–317

166 Kawamura, T., Suzuki, J., Wang, Y. V., Menendez, S., Morera, L. B., Raya, A., Wahl,G. M. and Belmonte, J. C. (2009) Linking the p53 tumour suppressor pathway tosomatic cell reprogramming. Nature 460, 1140–1144

167 Hong, H., Takahashi, K., Ichisaka, T., Aoi, T., Kanagawa, O., Nakagawa, M., Okita, K.and Yamanaka, S. (2009) Suppression of induced pluripotent stem cell generation bythe p53-p21 pathway. Nature 460, 1132–1135

168 Li, H., Collado, M., Villasante, A., Strati, K., Ortega, S., Canamero, M., Blasco, M. A.and Serrano, M. (2009) The Ink4/Arf locus is a barrier for iPS cell reprogramming.Nature 460, 1136–1139

169 Marion, R. M., Strati, K., Li, H., Murga, M., Blanco, R., Ortega, S., Fernandez-Capetillo,O., Serrano, M. and Blasco, M. A. (2009) A p53-mediated DNA damage response limitsreprogramming to ensure iPS cell genomic integrity. Nature 460, 1149–1153

170 Utikal, J., Polo, J. M., Stadtfeld, M., Maherali, N., Kulalert, W., Walsh, R. M., Khalil, A.,Rheinwald, J. G. and Hochedlinger, K. (2009) Immortalization eliminates a roadblockduring cellular reprogramming into iPS cells. Nature 460, 1145–1148

171 Banito, A., Rashid, S. T., Acosta, J. C., Li, S., Pereira, C. F., Geti, I., Pinho, S., Silva,J. C., Azuara, V., Walsh, M. et al. (2009) Senescence impairs successfulreprogramming to pluripotent stem cells. Genes Dev. 23, 2134–2139

172 Miura, K., Okada, Y., Aoi, T., Okada, A., Takahashi, K., Okita, K., Nakagawa, M.,Koyanagi, M., Tanabe, K., Ohnuki, M. et al. (2009) Variation in the safety of inducedpluripotent stem cell lines. Nat. Biotechnol. 27, 743–745

173 Vallier, L., Touboul, T., Brown, S., Cho, C., Bilican, B., Alexander, M., Cedervall, J.,Chandran, S., Ahrlund-Richter, L., Weber, A. and Pedersen, R. A. (2009) Signalingpathways controlling pluripotency and early cell fate decisions of human inducedpluripotent stem cells. Stem Cells 27, 2655–2666

174 Saha, K. and Jaenisch, R. (2009) Technical challenges in using human inducedpluripotent stem cells to model disease. Cell Stem Cell 5, 584–595

c© The Authors Journal compilation c© 2010 Biochemical Society

Emerging use of stem cells in regenerative medicine 23

175 Touboul, T., Hannan, N., Corbineau, S., Martinez, A., Martinet, C., Branchereau, S.,Mainot, S., Strick-Marchand, H., Pedersen, R., Santo, J. D. et al. (2009) Generation offunctional hepatocytes from human embryonic stem cells under chemically definedconditions which recapitulate liver development. Hepatology, doi: 10.1002/hep.23506

176 Thomson, J. A., Kalishman, J., Golos, T. G., Durning, M., Harris, C. P., Becker, R. A.and Hearn, J. P. (1995) Isolation of a primate embryonic stem cell line. Proc. Natl.Acad. Sci. U.S.A. 92, 7844–7848

177 Thomson, J. A., Kalishman, J., Golos, T. G., Durning, M., Harris, C. P. and Hearn, J. P.(1996) Pluripotent cell lines derived from common marmoset (Callithrix jacchus)blastocysts. Biol. Reprod. 55, 254–259

178 Bongso, A., Fong, C. Y., Ng, S. C. and Ratnam, S. (1994) Isolation and culture of innercell mass cells from human blastocysts. Hum. Reprod. 9, 2110–2117

179 Stojkovic, M., Lako, M., Stojkovic, P., Stewart, R., Przyborski, S., Armstrong, L., Evans,J., Herbert, M., Hyslop, L., Ahmad, S. et al. (2004) Derivation of human embryonicstem cells from day-8 blastocysts recovered after three-step in vitro culture. Stem Cells22, 790–797

180 Strelchenko, N., Verlinsky, O., Kukharenko, V. and Verlinsky, Y. (2004) Morula-derivedhuman embryonic stem cells. Reprod. Biomed. Online 9, 623–629

181 Klimanskaya, I., Chung, Y., Becker, S., Lu, S. J. and Lanza, R. (2006) Human embryonicstem cell lines derived from single blastomeres. Nature 444, 481–485

182 Lin, G., OuYang, Q., Zhou, X., Gu, Y., Yuan, D., Li, W., Liu, G., Liu, T. and Lu, G. (2007)A highly homozygous and parthenogenetic human embryonic stem cell line derivedfrom a one-pronuclear oocyte following in vitro fertilization procedure. Cell Res. 17,999–1007

183 Mai, Q., Yu, Y., Li, T., Wang, L., Chen, M. J., Huang, S. Z., Zhou, C. and Zhou, Q.(2007) Derivation of human embryonic stem cell lines from parthenogeneticblastocysts. Cell Res. 17, 1008–1019

184 Revazova, E. S., Turovets, N. A., Kochetkova, O. D., Kindarova, L. B., Kuzmichev, L. N.,Janus, J. D. and Pryzhkova, M. V. (2007) Patient-specific stem cell lines derived fromhuman parthenogenetic blastocysts. Cloning Stem Cells 9, 432–449

185 Brons, I. G., Smithers, L. E., Trotter, M. W., Rugg-Gunn, P., Sun, B., Chuva de SousaLopes, S. M., Howlett, S. K., Clarkson, A., Ahrlund-Richter, L., Pedersen, R. A. andVallier, L. (2007) Derivation of pluripotent epiblast stem cells from mammalianembryos. Nature 448, 191–195

186 Tesar, P. J., Chenoweth, J. G., Brook, F. A., Davies, T. J., Evans, E. P., Mack, D. L.,Gardner, R. L. and McKay, R. D. (2007) New cell lines from mouse epiblastshare defining features with human embryonic stem cells. Nature 448,196–199

187 Matsui, Y., Zsebo, K. and Hogan, B. L. (1992) Derivation of pluripotentialembryonic stem cells from murine primordial germ cells in culture. Cell 70,841–847

188 Resnick, J. L., Bixler, L. S., Cheng, L. and Donovan, P. J. (1992) Long-termproliferation of mouse primordial germ cells in culture. Nature 359,550–551

189 Kanatsu-Shinohara, M., Inoue, K., Lee, J., Yoshimoto, M., Ogonuki, N., Miki, H., Baba,S., Kato, T., Kazuki, Y., Toyokuni, S. et al. (2004) Generation of pluripotent stem cellsfrom neonatal mouse testis. Cell 119, 1001–1012

190 Guan, K., Nayernia, K., Maier, L. S., Wagner, S., Dressel, R., Lee, J. H., Nolte, J., Wolf,F., Li, M., Engel, W. and Hasenfuss, G. (2006) Pluripotency of spermatogonial stemcells from adult mouse testis. Nature 440, 1199–1203

191 Shamblott, M. J., Axelman, J., Wang, S., Bugg, E. M., Littlefield, J. W., Donovan, P. J.,Blumenthal, P. D., Huggins, G. R. and Gearhart, J. D. (1998) Derivation of pluripotentstem cells from cultured human primordial germ cells. Proc. Natl. Acad. Sci. U.S.A.95, 13726–13731

Received 15 January 2010/4 March 2010; accepted 9 March 2010Published on the Internet 28 April 2010, doi:10.1042/BJ20100102

c© The Authors Journal compilation c© 2010 Biochemical Society