Skip to main content
Cell Transplantation logoLink to Cell Transplantation
. 2020 Apr 1;29:0963689720907565. doi: 10.1177/0963689720907565

SOX2 for Stem Cell Therapy and Medical Use: Pros or Cons?

Hong-Meng Chuang 1,2, Mao-Hsuan Huang 1,3, Yu-Shuan Chen 1,2, Horng-Jyh Harn 1,4,
PMCID: PMC7444200  PMID: 32233795

Abstract

Stem cell transplantation is a fast-developing technique, which includes stem cell isolation, purification, and storage, and it is in high demand in the industry. In addition, advanced applications of stem cell transplantation, including differentiation, gene delivery, and reprogramming, are presently being studied in clinical trials. In contrast to somatic cells, stem cells are self-renewing and have the ability to differentiate; however, the molecular mechanisms remain unclear. SOX2 (sex-determining region Y [SRY]-box 2) is one of the well-known reprogramming factors, and it has been recognized as an oncogene associated with cancer induction. The exclusion of SOX2 in reprogramming methodologies has been used as an alternative cancer treatment approach. However, the manner by which SOX2 induces oncogenic effects remains unclear, with most studies demonstrating its regulation of the cell cycle and no insight into the maintenance of cellular stemness. For controlling certain critical pathways, including Shh and Wnt pathways, SOX2 is considered irreplaceable and is required for the normal functioning of stem cells, particularly neural stem cells. In this report, we discussed the functions of SOX2 in both stem and cancer cells, as well as how this powerful regulator can be used to control cell fate.

Keywords: SOX2, cell transplantation, stem cells, cancer, stemness

Introduction

Stem cell transplantation is a well-established technique that has been part of the clinical treatment strategies for both malignant (e.g., acute myeloid leukemia and Hodgkin’s lymphoma) and nonmalignant (e.g., thalassemia and sickle cell anemia) diseases and disorders since 19591. In response to the increasing medical need, stem cells are now being isolated and transplanted from a variety of sources, including umbilical cord blood, placenta, amniotic fluid, dental pulp, and adipose tissue24. In addition, the discovery of induced pluripotent stem cells (iPSCs) has dramatically broadened the field5. These improvements are a reflection of the unmet medical need of regenerative medicine and the limitations of drugs and medical devices6. Despite organ transplantation having been successfully performed for the heart, kidney, liver, lung, pancreas, intestine, and thymus, organ sources are in very limited supply7,8. Even if a patient receives an organ and the transplantation is considered a success, the patient is required to take antirejection drugs, which have the risk of severe side effects. As such, the development of artificial organs may represent a promising solution; however, effective and efficient techniques for tissue engineering pose numerous challenges9.

Stem cells and/or progenitor cells may be a good choice for compensation of the functions of target tissues and for the secretion of appropriate cytokines and growth factors, including those that are considered immunomodulatory1012. However, the majority of studies have revealed that delivered cells rarely transdifferentiate into their target type and the survival time remains insufficient13,14. Nonetheless, while the mechanisms remain unclear, most experimental and clinical results have shown positive results1517. As such, the technologies underlying stem cell therapy continue to improve regarding cellular function and survival duration. The most important breakthrough has been the reprogramming of somatic cells into pluripotent stem cells with the exogenous expression of certain transcription factors18. The potential applications of stem cells, including cellular transplantation and organ development, have been tremendously enhanced after the discovery of iPSCs19,20.

As a member of the SOX gene family and SOXB group, which includes SOX1, SOX2, and SOX3, SOX2 (sex-determining region Y [SRY]-box 2) encodes a 34.3 kD protein21. As a key regulator of self-renewal, SOX2 protein binds to octamer-binding transcription factor 4 (Oct4) and enhances the expression of Nanog22,23. However, Tanaka et al. indicated that SOX2 is unnecessary as an enhancer, suggesting that it modulates the expression of Oct42426. The coupling of SOX2 to paired box protein 6 (PAX6) and BRN2 (encoded by POU3F2 in humans) has been shown to regulate eye and neural primordial cell functions27. Interestingly, SOX2 and/or the partner protein are not considered sufficient for transcriptional activation, but this complex is28. Once the complex is formed, downstream genes such as undifferentiated embryonic cell transcription factor 1 and fibroblast growth factor 4 activate and enhance embrionic stem cell development and survival29. Accordingly, the knockdown of Sox2 expression in mouse embryonic stem cells (ESCs) results in the failure of this self-renewal property and leads to differentiation22. In contrast to tumorigenesis, the expression level of SOX2 correlates with lower survival and treatment resistance30. Therefore, we evaluated the relationship between SOX2 and its functions in both stem and cancer cells and discovered a potential approach for improving stem cells and deteriorating cancer cells.

SOX2 Is Associated With an Enormous Expression Network

The characteristics of stemness are associated with the target genes of SOX2. In addition, stem cells possess regulatory mechanisms to maintain the appropriate expression of SOX2. For mouse ESCs, the exogenous elevated expression of Sox2 leads to differentiation of ESCs into a wide range of cell types, including neuroectoderm, mesoderm, and trophectoderm (TE)31. Moreover, feedback regulation involved in the Akt pathway reactivates endogenous Sox2 expression and serves to retain cellular stemness (Fig. 1)40. However, in comparison with iPSCs, the expression of SOX2 is artificial and lacks interactive control. Nevertheless, to reprogram cells into iPSCs, four genes, namely, Oct4, Klf-4, SOX2, and c-Myc (abbreviated to OKSM), are exogenously activated and these genes need a specific ratio to work adequately. Since the OKSM is necessary for pluripotency, other accessory factors such as Nanog and Sal-like protein 4 can only increase the efficiency of reprogramming and cannot replace SOX2 or OCT441,42. For example, a ratio increase of Klf4 is recommended in one of the commercial cellular reprogramming kits. Moreover, the expression of SOX2 is activated by the VP16 transactivator and further improves reprogramming efficiency43. These findings indicate that the OKSM acts as a driving force in the fertilization stage and should be tightly restricted or the cells may get out of control. Thus, the upstream and downstream regions of the SOX2 coding sequence contain large untranslated regions (UTRs) or the so-called gene deserts to prevent mutations and false binding44. Certain enhancers, including miRNA, long-noncoding RNA, and posttranslational modification have been shown to reversely regulate both transcriptional and posttranslational activity. At least 19 miRNAs, including miR-200b and miR-145, can influence the expression of SOX245. Indeed, miR-200 facilitates the reprogramming of fibroblasts into iPSCs in the presence of OKSM46. Moreover, miR-145 targets the 3′-UTR of SOX2 and inhibits self-renewal in human ESCs and iPSCs47. In other words, simply elevating the expression of SOX2 serves to attract negative regulation molecules such as the cyclin-dependent kinase inhibitor 1A (p21Cip1)48. Conversely, the zygote has a massive demethylation pattern compared with reprogrammed cells49. Although existing techniques, including single-cell RNA-Seq, methylation array, and bioinformatics, have confirmed that the methylation state changes in zygotes and somatic cells, the secrets of dedifferentiation remain a mystery50.

Fig. 1.

Fig. 1.

Molecular interactions illustrate various SOX2 signaling partners.3239

SOX2 Utilization in Stem Cell Therapy

Except for the stemness regulation of SOX2, the existence for cell differentiation and development is necessary. Notably, in the development of ESCs, when the blastocyst is formed and then divides into the TE and inner cell mass, the expression of SOX2 is decreased. However, the TE will not form if SOX2 is impaired or knocked down by siRNA51. This change is due to the complex formed with Oct4 and Nanog. For example, Oct4 and Nanog bind to SOX2 and regulate its functions of self-renewal and differentiation inhibition52. In adult humans, the olfactory nerve proliferates and is replaced every 3 to 4 weeks. The SOX2/PAX6-expressed epithelium plays an important role in maintaining the multipotency of the olfactory nerve53. These findings suggest further applications in the transplantation from iPSC-differentiated neural stem cells (NSCs). In particular, the in vitro-transcribed mRNA of SOX2 has been shown to induce NSC morphology in human dermal fibroblasts54. In addition, another study revealed that exogenous Sox2 expression in rat bone marrow–derived stem cells (BMSCs) benefits the cell transplantation treatment in a rat traumatic brain injury (TBI) model55. Especially, BMSCs retain their self-renewal property via the expression of Sirtuin1 (SIRT1)56. SIRT1 is a lysine deacetylase that contributes in maintaining SOX2 content by avoiding the acetylation and ubiquitination of SOX257. Moreover, proliferation and differentiation potential is conferred by the forced SOX2 expression of BMSC58. Using MRI tracking, Jiang et al. found that NSCs migrate into the injury site of rats with TBI59. Therefore, the existence of SOX2 is essential for the maintenance of self-renewal and multipotency. These studies suggested that Sox2-positive cells may play a role in neuron regeneration, enhancing neural functions after brain injury60.

Direct Evidence of SOX2 Initiating Tumorigenesis

SOX2 is generally considered an oncogene; however, its role in tumorigenesis remains controversial61,62. As part of the same lineage of breast cancer cells, the SOX2-positive population shows a greater colony-forming ability and would be abolished by SOX2 knockdown63,64. SOX2 is amplified in patients with cancer, and it contributes to the same stemness property observed in stem cells of patients with lung, brain, breast, and colon tumors65.

The clinical implications of SOX2 and cancers vary depending on the type of cancer, influencing patient survival and prognosis66. These molecules and pathways include VEGF, MAPK, Notch-Shh, BMP, Jak-STAT, and others, depending on the types of tumors6769. In brief, SOX2 regulates downstream genes and microRNAs by direct DNA binding, resulting in the alteration of thousands of genes and hundreds of microRNAs35. Moreover, a glioma cell subset with high levels of SOX2 has been shown to be resistant to platelet-derived growth factor (PDGF)- and insulin-like growth factor 1 (IGF-1)-receptor inhibitors70. Conversely, SOX2 may play a role in the maintenance of PDGF and IGF-1 pathways in cancer cells as well as in stem cells and may produce dysplasia or tumor cell initiation. However, in patients with gastric cancer, the overall survival rate is lower with SOX2 methylation than with unmethylated SOX2. Moreover, the exogenous expression of SOX2 results in cell cycle arrest through cyclin-dependent kinase inhibitor 1B (p27Kip1) and Rb phosphorylation71. Indeed, Sox2 expression in the mouse respiratory epithelium does not cause pulmonary tumors but induces the cellular proliferation of respiratory epithelial cells72. Direct induced-NSCs can also be obtained by SOX2 expression in human and mouse fibroblasts without tumorigenesis73.

Future Prospects of SOX2 Utilization

Due to the close relationship between SOX2 and cancer, studies that have investigated SOX2-dependent gene manipulation are limited. However, cell differentiation and proliferation has been achieved, including SOX2-expressing dental pulp stem cells, which lead to the differentiation of odontoblasts74. SOX2 also cooperates with various cofactors, including Oct4 for stemness, BRN2 for neural differentiation, and PAX3 for melanocyte maturation. These studies suggested that SOX2 (and possibly other members in the family) is one of the masters regulating cell fate and is associated with different kinds of cofactors75.

An urgent question we would ask is how to take advantage of SOX2 without eliciting detrimental effects? For the nerve system, SOX2 may prove to be a useful regulator for the maintenance of progenitor characteristics, allowing the cells to retain their ability to self-renew and differentiate into neurons, astrocytes, and oligodendrocytes76,77. Moreover, Hagey and Muhr found that decreased expression of Sox2 resulted in the differentiation of radial glia cells into their more developed progeny, intermediate progenitor cells78. However, mouse NSCs and progenitors have reduced differentiation ability and SOX2 expression with the loss of the E2f3a transcription factor, indicating that the expression of SOX2 is essential for neurogenesis79. These studies indicated that SOX2 is required for stemness but is unfavorable for differentiation. In other words, the expression of SOX2 should be controlled or there is a risk of the cells going corrupt.

Conversely, the SOX2-induced stemness ability is devastating and problematic when it appears in tumor cells. Since the existence of SOX2 is associated with the cell membrane, specific antibodies are not effective for disrupting its function. To control the stemness and metastatic properties, Tuhin et al. found that the downregulation effect of actinomycin D induced the cell death of breast cancer stem cells80. Moreover, SOX2 knockdown or small molecules reduced SOX2 expression and inhibited the stemness and metastatic properties61,81. A significant factor in patients with cancer is the selection of SOX2+ cells after anticancer therapies, including radiotherapy and chemotherapy, and SOX2-induced drug-resistant genes have been characterized in numerous studies82,83. These SOX2+ cells then form a new tumor bulk, with the most well-studied type being the quiescent Sonic Hedgehog subgroup medulloblastoma, which is activated into medulloblastoma-propagating cells after antimitotic drug treatment84. Moreover, the inhibition of the SOX2-driven transcriptional network arrests GBM growth by treatment with mithramycin, which is an antineoplastic antibiotic85. Indeed, SOX2 antigen and antibody were found in small-cell lung cancer (SCLC) cell lines and sera in SCLC patients. However, neither the antigen nor the antibody of SOX2 or other SOX group B genes exist in normal sera, suggesting that SOX2 might be a potential tumor target or marker86. However, these effects should be precisely targeted toward tumor cells to take advantage of the attributes of SOX2.

Summary

With the use of iPSC-derived retinal pigment epithelial cells transplanted in clinical trials, a large research and development effort has been undertaken to not only evaluate the effectiveness and safety but also improve the associated techniques87,88. In this report, we discussed the stemness-prone properties and some possible risks of SOX2 in Table 1. In addition, the safety issue in iPSC-derived cell therapy is the most important topic; therefore, Larsson et al. established a molecular beacon to compliment SOX2 mRNA, displaying the fluorescent signal when SOX2 is expressed89. Although the interaction networks remain incomplete, reactions with different expression levels of SOX2 are more apparent than ever. In summary, the potential applications of SOX2 are extremely promising but precise targeting and expression in the right place and at the right dosage are crucial.

Table 1.

A Summary Table of SOX2 Potential in Medical Use and Their Main Concerns for Cancer Therapy and Stem Cell Transplantation.

Potential in Medical Use Main Concerns Ways to Breakthrough
Manipulate cell fate The complicated upstream and downstream effectors of SOX2 Use the necessary domain of SOX266
Prolong stem cell lifespan during transplantation Risk of neoplastic or teratoma growth Use an additional molecular beacon that indicates abnormal SOX2 expression89
Retains stem cell potential May be involved in tumorigenesis The development of tumor-specific siRNA or antibodies90
Target as a tumor marker Constant expression in normal cells

Acknowledgments

We are grateful to Enago (www.enago.tw) for English proofreading.

Footnotes

Author Contributions: Writing—original draft preparation, HM Chuang; writing—review, MS Huang and YS Chen; conceptualization, HJ Harn.

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was funded by Buddhist Tzu Chi Bioinnovation Center, Tzu Chi Foundation, Hualien, Taiwan (project title: Development of a small molecule regulates transcription activity of SOX2 in type I collagen synthesis in fibroblasts for treating pulmonary fibrosis, MF00A130SS01) and Ministry of Science and Technology, Taiwan (MOST 106-2320-B-303-001-MY3 and MOST 106-2320-B-303-002-MY3).

References

  • 1. Copelan EA. Hematopoietic stem-cell transplantation. N Engl J Med. 2006;354(17):1813–1826. [DOI] [PubMed] [Google Scholar]
  • 2. Lynch W, Rezai S, Henderson CE. Human amniotic fluid: a source of stem cells for possible therapeutic use. Am J Obstet Gynecol. 2016;215(3):401. [DOI] [PubMed] [Google Scholar]
  • 3. Kfoury Y, Scadden DT. Mesenchymal cell contributions to the stem cell niche. Cell Stem Cell. 2015;16(3):239–253. [DOI] [PubMed] [Google Scholar]
  • 4. Mead B, Logan A, Berry M, Leadbeater W, Scheven BA. Concise review: dental pulp stem cells: A novel cell therapy for retinal and central nervous system repair. Stem Cells. 2017;35(1):61–67. [DOI] [PubMed] [Google Scholar]
  • 5. Hayashi R, Ishikawa Y, Sasamoto Y, Katori R, Nomura N, Ichikawa T, Araki S, Soma T, Kawasaki S, Sekiguchi K, Quantock AJ, et al. Co-ordinated ocular development from human iPS cells and recovery of corneal function. Nature. 2016;531(7594):376–380. [DOI] [PubMed] [Google Scholar]
  • 6. Chang CY, Ting HC, Liu CA, Su HL, Chiou TW, Harn HJ, Lin SZ. Induced pluripotent stem cells: a powerful neurodegenerative disease modeling tool for mechanism study and drug discovery [published online ahead of print January 1, 2018]. Cell Transplant. 2018;27(11):1588–1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Martin-Gandul C, Mueller NJ, Pascual M, Manuel O. The impact of infection on chronic allograft dysfunction and allograft survival after solid organ transplantation. Am J Transplant. 2015;15(12):3024–3040. [DOI] [PubMed] [Google Scholar]
  • 8. Manara AR, Murphy PG, O’Callaghan G. Donation after circulatory death. Br J Anaesth. 2012;108(Suppl 1):i108–i121. [DOI] [PubMed] [Google Scholar]
  • 9. Marx V. Tissue engineering: organs from the lab. Nature. 2015;522(7556):373–377. [DOI] [PubMed] [Google Scholar]
  • 10. Gao F, Chiu SM, Motan DA, Zhang Z, Chen L, Ji HL, Tse HF, Fu QL, Lian Q. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016;7:e2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Liao S, Zhang Y, Ting S, Zhen Z, Luo F, Zhu Z, Jiang Y, Sun S, Lai WH, Lian Q, Tse HF. Potent immunomodulation and angiogenic effects of mesenchymal stem cells versus cardiomyocytes derived from pluripotent stem cells for treatment of heart failure. Stem Cell Res Ther. 2019;10(1):78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Chuang HM, Shih TE, Lu KY, Tsai SF, Harn HJ, Ho LI. Mesenchymal stem cell therapy of pulmonary fibrosis: improvement with target combination [published online ahead of print January 1, 2018]. Cell Transplant. 2018;27(11):1581–1587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Hou D, Youssef EA, Brinton TJ, Zhang P, Rogers P, Price ET, Yeung AC, Johnstone BH, Yock PG, March KL. Radiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: implications for current clinical trials. Circulation. 2005;112(9 Suppl):1150–1156. [DOI] [PubMed] [Google Scholar]
  • 14. Kurtz A. Mesenchymal stem cell delivery routes and fate. Int J Stem Cells 2008;1(1):1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Squillaro T, Peluso G, Galderisi U. Clinical trials with mesenchymal stem cells: an update. Cell Transplant. 2016;25(5):829–848. [DOI] [PubMed] [Google Scholar]
  • 16. Trounson A, McDonald C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell. 2015;17(1):11–22. [DOI] [PubMed] [Google Scholar]
  • 17. Sykova E, Rychmach P, Drahoradova I, Konradova S, Ruzickova K, Vorisek I, Forostyak S, Homola A, Bojar M. Transplantation of mesenchymal stromal cells in patients with amyotrophic lateral sclerosis: results of phase I/IIa clinical Trial. Cell Transplant. 2017;26(4):647–658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Maza I, Caspi I, Zviran A, Chomsky E, Rais Y, Viukov S, Geula S, Buenrostro JD, Weinberger L, Krupalnik V, Hanna S, et al. Transient acquisition of pluripotency during somatic cell transdifferentiation with iPSC reprogramming factors. Nat Biotechnol. 2015;33(7):769–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Suchy F, Yamaguchi T, Nakauchi H. iPSC-Derived organs in vivo: challenges and promise. Cell Stem Cell. 2018;22(1):21–24. [DOI] [PubMed] [Google Scholar]
  • 20. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126(4):663–676. [DOI] [PubMed] [Google Scholar]
  • 21. Iwafuchi-Doi M, Yoshida Y, Onichtchouk D, Leichsenring M, Driever W, Takemoto T, Uchikawa M, Kamachi Y, Kondoh H. The Pou5f1/Pou3f-dependent but SoxB-independent regulation of conserved enhancer N2 initiates Sox2 expression during epiblast to neural plate stages in vertebrates. Dev Biol. 2011;352(2):354–366. [DOI] [PubMed] [Google Scholar]
  • 22. Chew JL, Loh YH, Zhang W, Chen X, Tam WL, Yeap LS, Li P, Ang YS, Lim B, Robson P, Ng HH. Reciprocal transcriptional regulation of Pou5f1 and Sox2 via the Oct4/Sox2 complex in embryonic stem cells. Mol Cell Biol. 2005;25(14):6031–6046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Mato Prado M, Frampton AE, Stebbing J, Krell J. Gene of the month: NANOG. J Clin Pathol. 2015;68(10):763–765. [DOI] [PubMed] [Google Scholar]
  • 24. Masui S, Nakatake Y, Toyooka Y, Shimosato D, Yagi R, Takahashi K, Okochi H, Okuda A, Matoba R, Sharov AA, Ko MS, et al. Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells. Nat Cell Biol. 2007;9(6):625–635. [DOI] [PubMed] [Google Scholar]
  • 25. Chen SM, Lee MS, Chang CY, Lin SZ, Cheng EH, Liu YH, Pan HC, Lee HC, Su HL. Prerequisite OCT4 maintenance potentiates the neural induction of differentiating human embryonic stem cells and induced pluripotent stem cells. Cell Transplant. 2015;24(5):829–844. [DOI] [PubMed] [Google Scholar]
  • 26. Tanaka S, Kamachi Y, Tanouchi A, Hamada H, Jing N, Kondoh H. Interplay of SOX and POU factors in regulation of the Nestin gene in neural primordial cells. Mol Cell Biol. 2004;24(20):8834–8846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Matsushima D, Heavner W, Pevny LH. Combinatorial regulation of optic cup progenitor cell fate by SOX2 and PAX6. Development. 2011;138(3):443–454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kamachi Y, Uchikawa M, Tanouchi A, Sekido R, Kondoh H. Pax6 and SOX2 form a co-DNA-binding partner complex that regulates initiation of lens development. Genes Dev. 2001;15(10):1272–1286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Yuan H, Corbi N, Basilico C, Dailey L. Developmental-specific activity of the FGF-4 enhancer requires the synergistic action of Sox2 and Oct-3. Genes Dev. 1995;9(21):2635–2645. [DOI] [PubMed] [Google Scholar]
  • 30. Chung JH, Jung HR, Jung AR, Lee YC, Kong M, Lee JS, Eun YG. SOX2 activation predicts prognosis in patients with head and neck squamous cell carcinoma. Sci Rep. 2018;8(1):1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Kopp JL, Ormsbee BD, Desler M, Rizzino A. Small increases in the level of Sox2 trigger the differentiation of mouse embryonic stem cells. Stem Cells. 2008;26(4):903–911. [DOI] [PubMed] [Google Scholar]
  • 32. Rimkus TK, Carpenter RL, Qasem S, Chan M, Lo HW. Targeting the sonic hedgehog signaling pathway: review of smoothened and GLI inhibitors. Cancers (Basel). 2016;8(2):E22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Kar S, Sengupta D, Deb M, Pradhan N, Patra SK. SOX2 function and Hedgehog signaling pathway are co-conspirators in promoting androgen independent prostate cancer. Biochim Biophys Acta Mol Basis Dis. 2017;1863(1):253–265. [DOI] [PubMed] [Google Scholar]
  • 34. Sarlak G, Vincent B. The roles of the stem cell-controlling sox2 transcription Factor: from neuroectoderm development to Alzheimer’s disease? Mol Neurobiol. 2016;53(3):1679–1698. [DOI] [PubMed] [Google Scholar]
  • 35. Schaefer T, Lengerke C. SOX2 protein biochemistry in stemness, reprogramming, and cancer: the PI3K/AKT/SOX2 axis and beyond. Oncogene. 2020;39(2):278–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Boyer LA, Plath K, Zeitlinger J, Brambrink T, Medeiros LA, Lee TI, Levine SS, Wernig M, Tajonar A, Ray MK, Bell GW, et al. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature. 2006;441(7091):349–353. [DOI] [PubMed] [Google Scholar]
  • 37. Basu-Roy U, Seo E, Ramanathapuram L, Rapp TB, Perry JA, Orkin SH, Mansukhani A, Basilico C. Sox2 maintains self renewal of tumor-initiating cells in osteosarcomas. Oncogene. 2012;31(18):2270–2282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Tang Y, Tian XC. JAK-STAT3 and somatic cell reprogramming. JAKSTAT 2013;2(4):e24935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Gordeeva O. TGFbeta Family Signaling pathways in pluripotent and teratocarcinoma stem cells’ fate decisions: balancing between self-renewal, differentiation, and cancer. Cells 2019;8(12):E1500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Ormsbee Golden BD, Wuebben EL, Rizzino A. Sox2 expression is regulated by a negative feedback loop in embryonic stem cells that involves AKT signaling and FoxO1. PLoS One. 2013;8(10):e76345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Gao Z, Cox JL, Gilmore JM, Ormsbee BD, Mallanna SK, Washburn MP, Rizzino A. Determination of protein interactome of transcription factor Sox2 in embryonic stem cells engineered for inducible expression of four reprogramming factors. J Biol Chem. 2012;287(14):11384–11397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Hanna J, Saha K, Pando B, van Zon J, Lengner CJ, Creyghton MP, van Oudenaarden A, Jaenisch R. Direct cell reprogramming is a stochastic process amenable to acceleration. Nature. 2009;462(7273):595–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Narayan S, Bryant G, Shah S, Berrozpe G, Ptashne M. OCT4 and SOX2 work as transcriptional activators in reprogramming human fibroblasts. Cell Rep. 2017;20(7):1585–1596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Zhou HY, Katsman Y, Dhaliwal NK, Davidson S, Macpherson NN, Sakthidevi M, Collura F, Mitchell JA. A Sox2 distal enhancer cluster regulates embryonic stem cell differentiation potential. Genes Dev. 2014;28(24):2699–2711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Vencken SF, Sethupathy P, Blackshields G, Spillane C, Elbaruni S, Sheils O, Gallagher MF, O’Leary JJ. An integrated analysis of the SOX2 microRNA response program in human pluripotent and nullipotent stem cell lines. BMC Genomics. 2014;15:711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Wang G, Guo X, Hong W, Liu Q, Wei T, Lu C, Gao L, Ye D, Zhou Y, Chen J, Wang J, et al. Critical regulation of miR-200/ZEB2 pathway in Oct4/Sox2-induced mesenchymal-to-epithelial transition and induced pluripotent stem cell generation. Proc Natl Acad Sci U S A. 2013;110(8):2858–2863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Xu N, Papagiannakopoulos T, Pan G, Thomson JA, Kosik KS. MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell. 2009;137(4):647–658. [DOI] [PubMed] [Google Scholar]
  • 48. Marques-Torrejon MA, Porlan E, Banito A, Gomez-Ibarlucea E, Lopez-Contreras AJ, Fernandez-Capetillo O, Vidal A, Gil J, Torres J, Farinas I. Cyclin-dependent kinase inhibitor p21 controls adult neural stem cell expansion by regulating Sox2 gene expression. Cell Stem Cell. 2013;12(1):88–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Seisenberger S, Peat JR, Hore TA, Santos F, Dean W, Reik W. Reprogramming DNA methylation in the mammalian life cycle: building and breaking epigenetic barriers. Philos Trans R Soc Lond B Biol Sci. 2013;368(1609):20110330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Huang Y, Liu H, Du H, Zhang W, Kang X, Luo Y, Zhou X, Li L. Developmental features of DNA methylation in CpG islands of human gametes and preimplantation embryos. Exp Ther Med. 2019;17(6):4447–4456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Keramari M, Razavi J, Ingman KA, Patsch C, Edenhofer F, Ward CM, Kimber SJ. Sox2 is essential for formation of trophectoderm in the preimplantation embryo. PLoS One. 2010;5(11):e13952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Boyer LA, Lee TI, Cole MF, Johnstone SE, Levine SS, Zucker JP, Guenther MG, Kumar RM, Murray HL, Jenner RG, Gifford DK, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell 2005;122(6):947–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Guo Z, Packard A, Krolewski RC, Harris MT, Manglapus GL, Schwob JE. Expression of pax6 and sox2 in adult olfactory epithelium. J Comp Neurol. 2010;518(21):4395–4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Kim BE, Choi SW, Shin JH, Kim JJ, Kang I, Lee BC, Lee JY, Kook MG, Kang KS. Single-factor SOX2 mediates direct neural reprogramming of human mesenchymal stem cells via transfection of in vitro transcribed mRNA. Cell Transplant. 2018;27(7):1154–1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Hao Q, Zheng J, Hu Y, Wang H. Bone marrow mesenchymal stem cells combined with Sox2 increase the functional recovery in rat with traumatic brain injury. Chinese Neurosurgical Journal. 2019;5(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Yuan HF, Zhai C, Yan XL, Zhao DD, Wang JX, Zeng Q, Chen L, Nan X, He LJ, Li ST. SIRT1 is required for long-term growth of human mesenchymal stem cells. J Mol Med (Berl) 2012;90(4):389–400. [DOI] [PubMed] [Google Scholar]
  • 57. Yoon DS, Choi Y, Jang Y, Lee M, Choi WJ, Kim SH, Lee JW. SIRT1 directly regulates SOX2 to maintain self-renewal and multipotency in bone marrow-derived mesenchymal stem cells. Stem Cells. 2014;32(12):3219–3231. [DOI] [PubMed] [Google Scholar]
  • 58. Go MJ, Takenaka C, Ohgushi H. Forced expression of Sox2 or Nanog in human bone marrow derived mesenchymal stem cells maintains their expansion and differentiation capabilities. Exp Cell Res. 2008;314(5):1147–1154. [DOI] [PubMed] [Google Scholar]
  • 59. Jiang L, Li R, Tang H, Zhong J, Sun H, Tang W, Wang H, Zhu J. MRI tracking of iPS cells-induced neural stem cells in traumatic brain injury rats. Cell Transplant 2019;28(6):747–755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Niu W, Zang T, Smith DK, Vue TY, Zou Y, Bachoo R, Johnson JE, Zhang CL. SOX2 reprograms resident astrocytes into neural progenitors in the adult brain. Stem Cell Reports. 2015;4(5):780–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Yen SY, Chuang HM, Huang MH, Lin SZ, Chiou TW, Harn HJ. n-Butylidenephthalide regulated tumor stem cell genes EZH2/AXL and reduced Its migration and invasion in glioblastoma. Int J Mol Sci. 2017;18(2):E372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Ferone G, Song JY, Sutherland KD, Bhaskaran R, Monkhorst K, Lambooij JP, Proost N, Gargiulo G, Berns A. SOX2 Is the determining oncogenic switch in promoting lung squamous cell carcinoma from different cells of origin. Cancer Cell. 2016;30(4):519–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Wu F, Zhang J, Wang P, Ye X, Jung K, Bone KM, Pearson JD, Ingham RJ, McMullen TP. Identification of two novel phenotypically distinct breast cancer cell subsets based on Sox2 transcription activity. Cell Signal. 2012;24(11):1989–1998. [DOI] [PubMed] [Google Scholar]
  • 64. Gangemi RM, Griffero F, Marubbi D, Perera M, Capra MC, Malatesta P, Ravetti GL, Zona GL, Daga A, Corte G. SOX2 silencing in glioblastoma tumor-initiating cells causes stop of proliferation and loss of tumorigenicity. Stem Cells. 2009;27(1):40–48. [DOI] [PubMed] [Google Scholar]
  • 65. Liu K, Lin B, Zhao M, Yang X, Chen M, Gao A, Liu F, Que J, Lan X. The multiple roles for Sox2 in stem cell maintenance and tumorigenesis. Cell Signal. 2013;25(5):1264–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Weina K, Utikal J. SOX2 and cancer: current research and its implications in the clinic. Clin Transl Med. 2014;3:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Chen S, Xu Y, Chen Y, Li X, Mou W, Wang L, Liu Y, Reisfeld RA, Xiang R, Lv D. SOX2 gene regulates the transcriptional network of oncogenes and affects tumorigenesis of human lung cancer cells. PLoS One. 2012;7(5):e36326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Fang X, Yu W, Li L, Shao J, Zhao N, Chen Q, Ye Z, Lin SC, Zheng S, Lin B. ChIP-seq and functional analysis of the SOX2 gene in colorectal cancers. OMICS. 2010;14(4):369–384. [DOI] [PubMed] [Google Scholar]
  • 69. Engelen E, Akinci U, Bryne JC, Hou J, Gontan C, Moen M, Szumska D, Kockx C, van Ijcken W, Dekkers DH. Sox2 cooperates with Chd7 to regulate genes that are mutated in human syndromes. Nat Genet. 2011;43(6):607–611. [DOI] [PubMed] [Google Scholar]
  • 70. Hagerstrand D, He X, Bradic Lindh M, Hoefs S, Hesselager G, Ostman A, Nister M. Identification of a SOX2-dependent subset of tumor- and sphere-forming glioblastoma cells with a distinct tyrosine kinase inhibitor sensitivity profile. Neuro Oncol. 2011;13(11):1178–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Otsubo T, Akiyama Y, Yanagihara K, Yuasa Y. SOX2 is frequently downregulated in gastric cancers and inhibits cell growth through cell-cycle arrest and apoptosis. Br J Cancer. 2008;98(4):824–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Tompkins DH, Besnard V, Lange AW, Keiser AR, Wert SE, Bruno MD, Whitsett JA. Sox2 activates cell proliferation and differentiation in the respiratory epithelium. Am J Respir Cell Mol Biol. 2011;45(1):101–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Ring KL, Tong LM, Balestra ME, Javier R, Andrews-Zwilling Y, Li G, Walker D, Zhang WR, Kreitzer AC, Huang Y. Direct reprogramming of mouse and human fibroblasts into multipotent neural stem cells with a single factor. Cell Stem Cell. 2012;11(1):100–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Yang Y, Zhao Y, Liu X, Chen Y, Liu P, Zhao L. Effect of SOX2 on odontoblast differentiation of dental pulp stem cells. Mol Med Rep. 2017;16(6):9659–9663. [DOI] [PubMed] [Google Scholar]
  • 75. Julian LM, McDonald AC, Stanford WL. Direct reprogramming with SOX factors: masters of cell fate. Curr Opin Genet Dev. 2017;46:24–36. [DOI] [PubMed] [Google Scholar]
  • 76. Graham V, Khudyakov J, Ellis P, Pevny L. SOX2 functions to maintain neural progenitor identity. Neuron. 2003;39(5):749–765. [DOI] [PubMed] [Google Scholar]
  • 77. Zappone MV, Galli R, Catena R, Meani N, De Biasi S, Mattei E, Tiveron C, Vescovi AL, Lovell-Badge R, Ottolenghi S, Nicolis SK. Sox2 regulatory sequences direct expression of a (beta)-geo transgene to telencephalic neural stem cells and precursors of the mouse embryo, revealing regionalization of gene expression in CNS stem cells. Development. 2000;127(11):2367–2382. [DOI] [PubMed] [Google Scholar]
  • 78. Hagey DW, Muhr J. Sox2 acts in a dose-dependent fashion to regulate proliferation of cortical progenitors. Cell Rep. 2014;9(5):1908–1920. [DOI] [PubMed] [Google Scholar]
  • 79. Julian LM, Vandenbosch R, Pakenham CA, Andrusiak MG, Nguyen AP, McClellan KA, Svoboda DS, Lagace DC, Park DS, Leone G. Opposing regulation of Sox2 by cell-cycle effectors E2f3a and E2f3b in neural stem cells. Cell Stem Cell 2013;12(4):440–452. [DOI] [PubMed] [Google Scholar]
  • 80. Das T, Nair RR, Green R, Padhee S, Howell M, Banerjee J, Mohapatra SS, Mohapatra S. Actinomycin D Down-regulates SOX2 Expression and Induces Death in Breast Cancer Stem Cells. Anticancer Res. 2017;37(4):1655–1663. [DOI] [PubMed] [Google Scholar]
  • 81. Li ZR, Jiang Y, Hu JZ, Chen Y, Liu QZ. SOX2 knockdown inhibits the migration and invasion of basal cell carcinoma cells by targeting the SRPK1-mediated PI3K/AKT signaling pathway. Oncol Lett. 2019;17(2):1617–1625. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 82. Mu P, Zhang Z, Benelli M, Karthaus WR, Hoover E, Chen CC, Wongvipat J, Ku SY, Gao D, Cao Z. SOX2 promotes lineage plasticity and antiandrogen resistance in TP53- and RB1-deficient prostate cancer. Science. 2017;355(6320):84–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Li D, Zhao LN, Zheng XL, Lin P, Lin F, Li Y, Zou HF, Cui RJ, Chen H, Yu XG. Sox2 is involved in paclitaxel resistance of the prostate cancer cell line PC-3 via the PI3K/Akt pathway. Mol Med Rep. 2014;10(6):3169–3176. [DOI] [PubMed] [Google Scholar]
  • 84. Vanner RJ, Remke M, Gallo M, Selvadurai HJ, Coutinho F, Lee L, Kushida M, Head R, Morrissy S, Zhu X. Quiescent sox2(+) cells drive hierarchical growth and relapse in sonic hedgehog subgroup medulloblastoma. Cancer Cell 2014;26(1):33–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Singh DK, Kollipara RK, Vemireddy V, Yang XL, Sun Y, Regmi N, Klingler S, Hatanpaa KJ, Raisanen J. Oncogenes activate an autonomous transcriptional regulatory circuit that drives glioblastoma. Cell Rep. 2017;18(4):961–976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Dhodapkar KM, Gettinger SN, Das R, Zebroski H, Dhodapkar MV. SOX2-specific adaptive immunity and response to immunotherapy in non-small cell lung cancer. Oncoimmunology. 2013;2(7) e25205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Mandai M, Kurimoto Y, Takahashi M. Autologous Induced Stem-Cell-Derived Retinal Cells for Macular Degeneration. N Engl J Med. 2017;377(8):792–793. [DOI] [PubMed] [Google Scholar]
  • 88. Sharma R, Khristov V, Rising A, Jha BS, Dejene R, Hotaling N, Li Y, Stoddard J, Stankewicz C, Wan Q. Clinical-grade stem cell-derived retinal pigment epithelium patch rescues retinal degeneration in rodents and pigs. Sci Transl Med. 2019;11(475). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Larsson HM, Lee ST, Roccio M, Velluto D, Lutolf MP, Frey P, Hubbell JA. Sorting live stem cells based on Sox2 mRNA expression. PLoS One. 2012;7(11): e49874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Malinee M, Kumar A, Hidaka T, Horie M, Hasegawa K, Pandian GN, Sugiyama H. Targeted suppression of metastasis regulatory transcription factor SOX2 in various cancer cell lines using a sequence-specific designer pyrrole-imidazole polyamide. Bioorg Med Chem. 2020;28(3):115248. [DOI] [PubMed] [Google Scholar]

Articles from Cell Transplantation are provided here courtesy of SAGE Publications

RESOURCES