WNT signaling in stem cell differentiation and tumor formation

Hong , … , Yehong ,

J Clin Invest. 2013;123(4):1422-1424. https://doi.org/10.1172/JCI69324.

Commentary

Embryonic stem cells (ESCs) hold great therapeutic promise for the regeneration of functional cell types and clinical applications. However, tumorigenic potential of stem cells in a transplanted host remains a major obstacle. In this issue of the JCI, and colleagues identified TCF7-mediated canonical WNT signaling as a critical determinant of both the tumorigenicity and therapeutic function of ESC-derived retinal progenitor cells (ESC-RPCs). Their findings suggested that addressing key extracellular signaling and related intrinsic factors will be essential for the successful use of ESC-derived progenitor transplantation.

Find the latest version: https://jci.me/69324/pdf commentaries

6. Koch PJ, Franke WW. Desmosomal cadherins: 12. Brennan D, et al. Suprabasal Dsg2 expression in 17. Matsunaga-Udagawa R, et al. The scaffold protein another growing multigene family of adhesion transgenic mouse skin confers a hyperproliferative Shoc2/SUR-8 accelerates the interaction of Ras and molecules. Curr Opin Cell Biol. 1994;6(5):682–687. and apoptosis-resistant phenotype to keratino- Raf. J Biol Chem. 2010;285(10):7818–7826. 7. Harmon RM, et al. Desmoglein-1/Erbin inter- cytes. J Cell Sci. 2007;120(pt 5):758–771. 18. Siegel DH, Mann JA, Krol AL, Rauen KA. Derma- action suppresses ERK activation to support 13. Getsios S, et al. Desmoglein 1-dependent suppres- tological phenotype in Costello syndrome: conse- epidermal differentiation. J Clin Invest. 2013; sion of EGFR signaling promotes epidermal dif- quences of Ras dysregulation in development. Br J 123(4):1556–1570. ferentiation and morphogenesis. J Cell Biol. 2009; Dermatol. 2012;166(3):601–607. 8. Stanley JR, Amagai M. Pemphigus, bullous impe- 185(7):1243–1258. 19. Nava C, et al. Cardio-facio-cutaneous and Noonan tigo, and staphylococcal scalded skin syndrome. 14. Rickman L, et al. N-terminal deletion in a desmoso- syndromes due to mutations in the RAS/MAPK N Engl J Med. 2006;355(17):1800–1810. mal cadherin causes the autosomal dominant skin signalling pathway: genotype-phenotype relation- 9. Ishii K, C, Siegel DL, Stanley JR. Isolation disease striate palmoplantar keratoderma. Hum ships and overlap with Costello syndrome. J Med of pathogenic monoclonal anti-desmoglein 1 Mol Genet. 1999;8(6):971–976. Genet. 2007;44(12):763–771. human antibodies by phage display of pemphigus 15. Wan H, et al. Striate palmoplantar keratoderma 20. McGrath JA. Inherited disorders of desmosomes. foliaceus autoantibodies. J Invest Dermatol. 2008; arising from desmoplakin and desmoglein 1 muta- Australas J Dermatol. 2005;46(4):221–229. 128(4):939–948. tions is associated with contrasting perturbations 21. Dusek RL, Attardi LD. Desmosomes: new perpetra- 10. H, et al. Protection of neonates against pemphi- of desmosomes and the keratin filament network. tors in tumour suppression. Nat Rev Cancer. 2011; gus foliaceus by desmoglein 3. N Engl J Med. 2000; Br J Dermatol. 2004;150(5):878–891. 11(5):317–323. 343(1):31–35. 16. Lebeau S, et al. Comparative analysis of the expres- 22. Tada H, Hatoko M, Tanaka A, Kuwahara M, 11. Elias PM, et al. Desmoglein isoform distribution sion of ERBIN and Erb-B2 in normal human skin Muramatsu T. Expression of desmoglein I and pla- affects stratum corneum structure and function. and cutaneous carcinomas. Br J Dermatol. 2005; koglobin in skin carcinomas. J Cutan Pathol. 2000; J Cell Biol. 2001;153(2):243–249. 152(6):1248–1255. 27(1):24–29. WNT signaling in stem cell differentiation and tumor formation Hong Ouyang,1,2 Yehong Zhuo,3 and Kang Zhang1,2,3,4

1Molecular Medicine Research Center and Department of Ophthalmology, State Key Laboratory of Biotherapy, West China Hospital, University, Chengdu, People’s Republic of China. 2Department of Ophthalmology and Shiley Eye Center and Institute for Genomic Medicine, UCSD, San Diego, California, USA. 3State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Yat-sen University, Guangzhou, People’s Republic of China. 4Veterans Administration Healthcare System, San Diego, California, USA.

Embryonic stem cells (ESCs) hold great therapeutic promise for the regener- degeneration improved visual acuity, with ation of functional cell types and clinical applications. However, tumorigenic no signs of hyperproliferation or tumorige- potential of stem cells in a transplanted host remains a major obstacle. In nicity after 4 months (4). These examples this issue of the JCI, Cui and colleagues identified TCF7-mediated canonical illustrate purity, stability, and proper local- WNT signaling as a critical determinant of both the tumorigenicity and ther- ization of transplanted cells in vivo and apeutic function of ESC-derived retinal progenitor cells (ESC-RPCs). Their prompted the development of numerous findings suggested that addressing key extracellular signaling and related differentiation protocols. Still, the risk intrinsic factors will be essential for the successful use of ESC-derived pro- of tumor formation remains a barrier, genitor transplantation. because there are no parameters to quan- tify safety factors and because the appro- Photoreceptor degeneration underlies side effects. It has been previously demon- priate stage of differentiation at which major causes of blindness, including macu- strated that ESCs or ESC-derived progen- ESC-derived progenitors should be used lar degeneration and retinitis pigmentosa, itors spontaneously form tumors upon has not been well evaluated. In this issue, affecting tens of millions of people world- transplantation in vivo, even when the Cui and colleagues attempted to address wide. In theory, vision of affected patients cells are predifferentiated or presorted (2). the above concerns by identifying major may improve if the diseased cells (rods and Despite this issue, successful cell replace- extracellular signaling and intrinsic factors cones) are replaced with new, healthy cells ment of human ESC–derived (hESC-de- controlling tumorigenicity and therapeutic that form appropriate connections with the rived) retinal cells for vision restoration in potential of ESC-derived retinal progenitor host retina. Embryonic stem cells (ESCs) animal models of photoreceptor degener- cells (ESC-RPCs) (5). possess unlimited self-renewal capabilities ation has been reported recently. Photore- and the ability to differentiate into any ceptor precursors derived from hESCs have ESC tumorigenic potential adult cell type (1). These unique features been shown to migrate into Crx-deficient The best proof of pluripotency of ESCs is make ESC-based therapy appealing for the mouse retina following intraocular injec- their ability to form teratomas in which treatment of various degenerative disor- tion, express appropriate markers for both the stem cells differentiate to various tis- ders but may also cause unwanted serious rod and cone photoreceptors, and subse- sue types of the embryo in a disordered quently restore some light responses (3). In fashion following transplantation into another study, subretinal transplantation immunosuppressed mice. Teratomas Conflict of interest: The authors have declared that no conflict of interest exists. of hESC-derived retinal pigment epithe- usually contain all three germ layers and Citation for this article: J Clin Invest. 2013; lium in patients with Stargardt’s macular have typical tumor characteristics (6). 123(4):1422–1424. doi:10.1172/JCI69324. dystrophy and dry age-related macular In other words, ESCs are naturally tum-

1422 The Journal of Clinical Investigation http://www.jci.org Volume 123 Number 4 April 2013 commentaries

Figure 1 Canonical WNT signaling determines the pro- liferation and differentiation state of ESC-de- rived progenitors via full-length TCF7 (fTCF7). SOX2 and NESTIN act as direct targets of fTCF7 in vitro. Blocking the pathway by delet- ing TCF7 or treating cells with the WNT inhib- itor DKK1 could reduce tumorigenicity and improve therapeutic efficiency. ΔNTCF7, nat- urally truncated form of TCF7.

origenic: like cancer cells, they are capable of β-catenin, intracellular mediator of Differentiation of ESCs into retinal of unlimited proliferation potential and the pathway, is frequently associated with progenitor cells clonal propagation without anchorage the differentiation potential of ESCs in Despite the concern of tumorigenic poten- dependence. Indeed, tumor formation teratomas (7). To determine whether WNT tial, stem cell–based therapy still holds is commonly considered to be a conse- activation was related to the increased the most promise to restore lost vision in quence of hyperproliferation of residual tumorigenicity of ESC-RPCs, Cui and col- patients with retinal diseases, due to our ESCs or precursor cells. Accordingly, leagues treated the ESC-RPCs with DKK1, extensive knowledge of retinal development purification of tissue-committed progen- an extracellular inhibitor of WNT signal- (10). To date, a number of protocols have itor cells from undifferentiated ESCs or ing, prior to transplantation. Expression been developed for differentiation of mature removal of undifferentiated ESCs before profiling revealed that neural progeni- retinal cells, including photoreceptors, by a transplantation becomes a necessary tor markers and cell proliferation mark- stepwise treatment with defined factors (3, 8). prerequisite in order to reduce the risk ers were repressed, whereas large num- Notably, the rates of tumor formation and of tumor growth. Several strategies have ber of committed retinal markers were successful host integration vary among dif- been implemented to address this critical upregulated by WNT inhibition. The pre- ferent reports, even using a same selective issue, including the selection of speci- treatment with DKK1 dramatically sup- marker. Cui et al. selected SOX1.EGFP–pos- fied cells based on their surface mark- pressed tumor formation and improved itive neural progenitor cells first and then ers (e.g., by FACS or magnetic-activated integration of ESC-RPCs into the trans- obtained ESC-RPCs and subsequent pho- cell sorting), isolation of lineages using planted host retina (5). toreceptor precursors by adding small mol- genetic manipulation (e.g., lineage-pro- Cui et al. further demonstrated that ecule inhibitors. Unlike most other groups, moter-driven antibiotic resistance), or upregulation of the direct transcriptional they did not choose DKK1 as a common fac- the knockdown of or pharmacological targets of canonical WNT signaling, tor to direct ESCs to retinal progenitors and interference with intracellular signaling NESTIN and SOX2 was critical to the tum- found a high rate of neural tumor formation pathways that increase tumorigenicity. origenicity of ESC-RPCs following trans- following ocular injection as a result (5). plantation (Figure 1). The authors also Thus, a more in-depth comparison between Role of WNT signaling in confirmed a previous study demonstrating P-RPCs and ESC-RPCs, such as assessment tumorigenesis and differentiation the importance of the WNT pathway in eye of epigenetic signatures using genome-wide Cui et al. demonstrated that ESC-RPCs development, in which a high percentage tools (11), will be required to optimize differ- had a high propensity for neural tumor of embryonic eye field–specific cells were entiation conditions and to make progress formation following ocular transplanta- generated from hESCs by upregulation of toward translational applications. tion (rate 60.58%); however, transplanta- DKK1, and the addition of WNT3A and As an alternative to ESC-RPCs or P-RPCs tion of primary retinal progenitor cells BMP4 to cultures abolished the expression for cell therapy, pluripotent stem cells (P-RPCs) from neonatal mice resulted in of PAX6 and RAX (8). Notably, a report could be differentiated into more mature, efficient integration to host retina with that WNT/β-catenin signaling promotes lineage-restricted stem cells, such as neural no evidence of tumor development. Tran- dedifferentiation and proliferation of stem cells, to reduce tumorigenicity (12). scriptomic profiling revealed increased Muller glia-derived retinal progenitors and However, as Cui et al. discuss, donor cells activity of the WNT signaling pathway in neural regeneration after damage (9) also might lose desired functions and the abil- ESC-RPCs compared with that in P-RPCs suggested that sustained activation of the ity to integrate to host retina by prolonged (5). WNT signaling is one of the key sig- WNT pathway may maintain the prolifer- differentiation. Thus, it will be critical to naling pathways in regulating cell prolif- ation ability of ESC-RPCs and contribute identify the appropriate stage of ESC-de- eration, motility, and differentiation as to the potential of tumor formation upon rived cells and ensure the balance between well as tumorigenesis, and upregulation transplantation. safety and efficiency (13). It remains to be

The Journal of Clinical Investigation http://www.jci.org Volume 123 Number 4 April 2013 1423 commentaries seen how broadly the approach to retinal Sun Yat-sen University, 54 South Xianlie 5. Cui L, et al. WNT signaling determines tumorige- nicity and function of ESC-derived retinal progen- progenitor cell differentiation by the WNT Road, Guangzhou, 510060, China. Phone: itors. J Clin Invest. 2013;123(4):1647–1661. pathway modulation can be applied to dif- 86.20.87330371; Fax: 86.20.87333271; 6. Przyborski SA. Differentiation of human embry- ferentiation of other tissue or cell types. E-mail: [email protected]. Or to: onic stem cells after transplantation in immune-de- Nevertheless, predifferentiation of ESCs Kang Zhang, Institute for Genomic Med- ficient mice. Stem Cells. 2005;23(9):1242–1250. 7. Kielman MF, et al. Apc modulates embryonic in vitro to a desired cell population before icine, University of California, San Diego, stem-cell differentiation by controlling the dos- transplantation both to ensure efficiency Department of Ophthalmology and Shiley age of beta-catenin signaling. Nat Genet. 2002; of transplantation as well as to minimize Eye Center, 9415 Campus Point Drive, La 32(4):594–605. 8. Meyer JS, et al. Modeling early retinal development the risk of tumor formation may be a good Jolla, California 92093-0946, USA. Phone: with human embryonic and induced pluripo- general strategy in stem cell therapy. 858.246.0814; Fax: 858.246.0873; E-mail: tent stem cells. Proc Natl Acad Sci U S A. 2009; [email protected]. 106(39):16698–16703. 9. Osakada F, Ooto S, Akagi T, Mandai M, Akaike A, Acknowledgments 1. Evans MJ, Kaufman MH. Establishment in cul- Takahashi M. Wnt signaling promotes regeneration This work was supported by 973 Program ture of pluripotential cells from mouse embryos. in the retina of adult mammals. J Neurosci. 2007; grants (2013CB967504); NSFC grants (no. Nature. 1981;292(5819):154–156. 27(15):4210–4219. 2. Doi D, et al. Prolonged maturation culture favors a 10. Zhang K, S. Stem cells and eye development. 81270992 and no. 81130017); NEI/NIH reduction in the tumorigenicity and the dopamin- N Eng J Med. 2011;365(4):370–372. grants EY014428, EY018660, EY019270, ergic function of human ESC-derived neural cells 11. Hannum G, et al. Genome-wide methylation pro- EY021374; VA Merit Award; and the Bur- in a primate model of Parkinson’s disease. Stem files reveal quantitative views of human aging rates. roughs Wellcome Fund Clinical Scientist Cells. 2012;30(5):935–945. Mol Cell. 2013;49(2):359–367. 3. Lamba DA, Gust J, Reh TA. Transplantation of 12. W, et al. Rapid induction and -term self-re- Award in Translational Research. human embryonic stem cell-derived photorecep- newal of primitive neural precursors from human tors restores some visual function in Crx-deficient embryonic stem cells by small molecule inhibitors. Address correspondence to: Yehong mice. Cell Stem Cell. 2009;4(1):73–79. Proc Natl Acad Sci U S A. 2011;108(20):8299–8304. 4. Schwartz SD, et al. Embryonic stem cell trials for 13. West EL, et al. Defining the integration capacity of Zhuo, State Key Laboratory of Ophthal- macular degeneration: a preliminary report. Lancet. embryonic stem cell-derived photoreceptor precur- mology, Zhongshan Ophthalmic Center, 2012;379(9817):713–720. sors. Stem Cells. 2012;30(7):1424–1435. Striking the target in iron overload disorders Karin E. Finberg

Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA.

The liver, a major site of body iron stores, mediates key responses that pre- Hepcidin insufficiency in iron serve systemic iron homeostasis. In this issue of the JCI, et al. demon- overload disorders strate that administration of antisense oligonucleotides that reduce expres- Inherited forms of iron overload (hemo- sion of Tmprss6, a hepatic protein that plays an essential role in maintaining chromatosis) result from mutations in iron balance, can attenuate disease severity in mouse models of human iron gene products that are required locally overload disorders. These data reveal the potential of novel TMPRSS6-tar- in the liver for hepcidin production. In geted therapies for the treatment of clinical conditions such as hereditary these disorders, the resulting hepcidin hemochromatosis and β-thalassemia. insufficiency leads to gastrointestinal iron absorption that exceeds the body’s needs. Hepcidin and the regulation of Hepcidin mediates these effects by trigger- The accumulation of excess iron promotes systemic iron balance ing the internalization and degradation of oxidative damage to tissues, which can The majority of iron required daily by the ferroportin, a cellular iron exporter that is ultimately lead to failure of organs such adult human body is used to meet the highly expressed at the basolateral mem- as the heart, liver, and endocrine glands. demands of hemoglobin synthesis. Most of brane of enterocytes and the cell membrane Hepcidin levels are inappropriately low rel- this iron is obtained through the recycling of macrophages (Figure 1). In hepatocytes, ative to body iron stores in another class of of senescent erythrocytes by macrophages hepcidin transcription is modulated by an clinical disorders associated with systemic in the spleen, liver, and bone marrow, while intracellular signaling cascade that is acti- iron loading: congenital anemias that are a small amount is absorbed from the diet vated by binding of BMP ligands to a cell- characterized by ineffective erythropoie- in the duodenum. Hepcidin, a small cir- surface receptor complex (Figure 2). The sis (IE) (2). IE describes a defective form culating peptide released by the liver, reg- liver, a major site of iron storage, increases of erythroid maturation characterized by ulates iron balance by limiting both the production of the BMP family member an increased proportion of erythroid pre- absorption of iron from the diet and the BMP6 in response to rising local iron stores; cursors, which, due to excessive apoptosis, release of iron from macrophage stores (1). this leads to increased signaling for hep- fail to produce the normal complement of cidin production, which in turn limits fur- mature erythrocytes. In β-thalassemia, the ther dietary iron absorption. Appropriate most common inherited form of IE, the Conflict of interest: The author has declared that no conflict of interest exists. regulation of intestinal iron absorption is primary genetic defect leads to reduced Citation for this article: J Clin Invest. 2013; critical, as there is no regulated mechanism synthesis of the β-globin component of 123(4):1424–1427. doi:10.1172/JCI68889. for eliminating surplus iron from the body. adult hemoglobin. The result is an excess

1424 The Journal of Clinical Investigation http://www.jci.org Volume 123 Number 4 April 2013