microRNA-based cancer cell reprogramming technology (Review)

  • Authors:
    • Shimpei Nishikawa
    • Hideshi Ishii
    • Naotsugu Haraguchi
    • Yoshihiro Kano
    • Takahito Fukusumi
    • Katsuya Ohta
    • Miyuki Ozaki
    • Dyah Laksmi Dewi
    • Daisuke Sakai
    • Taroh Satoh
    • Hiroaki Nagano
    • Yuichiro Doki
    • Masaki Mori
  • View Affiliations

  • Published online on: April 23, 2012     https://doi.org/10.3892/etm.2012.558
  • Pages: 8-14
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epigenetic modifications play crucial roles in cancer initiation and development. Complete reprogramming can be achieved through the introduction of defined biological factors such as Oct4, Sox2, Klf4, and cMyc into mouse and human fibroblasts. Introduction of these transcription factors resulted in the modification of malignant phenotype behavior. Recent studies have shown that human and mouse somatic cells can be reprogrammed to become induced pluripotent stem cells using forced expression of microRNAs, which completely eliminates the need for ectopic protein expression. Considering the usefulness of RNA molecules, microRNA-based reprogramming technology may have future applications in regenerative and cancer medicine.

Contents

Introduction

Complete reprogramming

Reprogramming-like phenomenon in cancer cells

Investigation of other factors

Perspectives

Introduction

MicroRNAs (miRNAs) modulate mRNA expression through base pairing between seed sequences in miRNA and complementary sequences within the open reading frame or an untranslated region of the target mRNA, thereby destabilizing mRNA and/or inhibiting protein synthesis (1,2). miRNAs play crucial roles in developmental biology (3,4), stress response (5,6), stem cell physiology (3,4,7), and diseases such as cancer (812).

Complete reprogramming

Introduction of the transcription factor MyoD into a muscle lineage caused differentiated cells to become myoblasts (13), indicating that single defined biological factors can successfully execute reprogramming of differentiated cells. Three cDNAs, including MyoD, were identified by screening a myocyte cDNA library with proliferating myoblast-specific subtracted cDNA probes. The MyoD cDNA contained a short protein segment similar to an amino acid sequence present in the myc protein family. MyoD cDNA was then transfected into fibroblasts, where it is not normally expressed (mouse C3H10T1/2, NIH3T3, Swiss 3T3, and L cells), and this was sufficient to convert these cells into stable myoblasts. Furthermore, myogenesis occurs to a lesser extent when MyoD is expressed in other lineage-committed adipoblasts.

The hypothesis that one or few transcription factors are sufficient for lineage conversion of terminally differentiated cells is supported by a study showing that C/EBP overexpression can lead to a stepwise reprogramming of B cells, thereby causing them to become macrophages (14). Initial C/EBP expression in B cells inhibits the B-cell commitment transcription factor Pax5, leading to downregulation of its target CD19. This increases the activity of PU.1, an ETS family transcription factor, which results in the upregulation of its target gene Mac-1 and other myeloid markers. This ultimately leads to the conversion of B cells to macrophages (15). Furthermore, three transcription factors, Ngn3 (also known as Neurog3), Pdx1, and Mafa, are capable of reprogramming differentiated pancreatic exocrine cells in adult mice into cells that closely resemble β-cells. These and several other studies (16,17) clearly indicate that defined biological factors are sufficient for complete lineage conversion of differentiated cells. Lineage conversion, i.e., direct cell reprogramming without reversion to a pluripotent stem cell (PSC) state, is indeed a major step, but the main aim of reprogramming studies is the successful generation of PSCs from differentiated cells. Understanding the mechanism responsible for the successful conversion of adult cells into other cell types would be beneficial for tissue repair, regeneration, and cancer therapy.

Takahashi and Yamanaka reported that complete reprogramming can be achieved by the introduction of defined biological factors such as Oct4 (also known as Pou5f1), Sox2, Klf4, and cMyc into mouse (18) and human fibroblasts (19,20). This is a major breakthrough considering the ethical issues related to the use of fertilized oocytes for the establishment and production of embryonic stem (ES) cells and the immunological incompatibility that occurs when unrelated individuals act as donor sources.

Reprogramming-like phenomenon in cancer cells

Genetic and epigenetic modifications are the hallmarks of cancer (21), but their roles in the determination of biological behavior have not been completely elucidated (22,23). Considering that cancer initiation and development is based on genetic information, the uncontrolled behavior of cancer cells is probably due to the combined deregulation of genetic and epigenetic programs (23). In tumor cells, genes can be controlled by epigenetic modifications (24) and underlying genomic mutations that lead to reversible and irreversible changes, thereby resulting in activation of oncogenes and inactivation of tumor-suppressor genes (21,23,2527). In other words, modifications in genetic information, such as double-strand breaks and fusions, can cause irreversible and stable changes in a cell (28), whereas epigenetic controls exert reversible and transient effects on cell behavior (23,29). Tumor development is a complex process driven by active (driver) and passive (passenger) mutations (30). The resultant tumors generally comprise heterogeneous tissues with tumor cell characteristics. It is generally considered that molecular mutations can lead to growth advantages during heterogeneous tumor formations, which ultimately increases tumor size. Furthermore, anti-apoptotic survival signals also support tumor development (31). The understanding of the involvement of changes in differentiation during tumor development remains somewhat elusive (23).

Recent studies indicate that aggressive tumors are associated with the preferential expression of ES-expressing genes, which suggests that common mechanisms are involved in the regulation of cancer stem cells (CSCs) and ES cells. Epigenetic regulating agents or events (including cell fusion and several epigenetic agents for DNA demethylation and histone acetylation) may reverse epigenetic information to normal biological characteristics in malignant tumors. A recent study that investigated the overexpression of the ES-expressing transcription factor indicated a modification in the epigenetic status and cancer cell behavior, after which the cells mimicked induced PSC (iPSC) behavior (32). Taken together, it is suggested that the viral-mediated transfection of transcription factors resulted in marked modification of epigenetic alterations in cancer cells due to its high magnitude of exogenous gene expression, whereas somewhat distinct alterations may develop in de novo cancer in terms of epigenetic net works, i.e., the activation of growth-promoting oncogenes and inactivation of tumor-suppressor genes. This suggests a possible experimental approach for using a reprogramming-like event in cancer research.

Cell reprogramming events observed in normal cells should be distinguished from reprogramming-like events observed in cancer cells. i) Numerous studies have suggested that the genetic and epigenetic pathways in cancer cells differ from those in normal cells. Although some pathways may appear to be common, recent studies have indicated that only aggressive tumors are associated with the preferential expression of ES-expressing genes. ii) Cancer cells involve irreversible genetic changes that can be controlled by a one-way irreversible program; however, normal cells largely exert their differentiation program via reversible epigenetic modifications. For example, pancreatic PDX-1 was expressed transiently in pancreatic somatic stem or progenitor cells, whereas it was not expressed in differentiated endocrine or pancreatic cancer cells (33). iii) The introduction of four defined transcription factors, Oct4, Sox2, Klf4, and cMyc, resulted in complete reprogramming of mouse (18) and human fibroblasts (19). The introduction of the same four transcription factors into cancer cells induced the expression of genes related to an undifferentiated status and showed multipotentiality by differentiating into three germ layers, although they did not form a teratoma, a hallmark of pluripotency. This suggests that the induced pluripotency program may be partially altered by genetic mutations in cancer cells or that the cancer program initiated in normal multipotent cells may be different from the pluripotency program in normal cells.

Investigation of other factors

Reprogramming factors such as Oct3, Sox2, Klf4, and c-Myc were identified on the basis of their differential expression in fibroblasts and ES cells (18). However, like previous studies (34,35), future studies may elucidate the role of other new important factors in cellular reprogramming that may contribute considerably to the development of safe and effective clinical therapeutic options. Recent studies using an expression library of transcription factors expressed by unfertilized oocytes and fertilized one-cell zygotes have identified a novel maternal transcription factor, Gli-like transcription factor (Glis1; Glis family zinc finger 1). Glis1 considerably enhances the generation of iPSCs from mouse and human fibroblasts, when it is expressed together with Oct4/Sox2/Klf4 (36). Glis1 effectively promotes multiple reprogramming pathways, including Myc, Nanog, Lin28, Wnt, Essrb, and the EMT related-pathways, thereby indicating the usefulness of Glis1 in effectively promoting the direct reprogramming of somatic cells during the generation of iPSC (36).

Small non-coding RNAs such as miRNAs, endogenous small interfering RNAs (endo-siRNAs), and Piwi-interacting RNAs (piRNAs) (3739), have different but essential roles in mammalian development (40). Differences among the small non-coding RNA types can be determined using numerous properties, such as the size of endo-siRNAs (21 nucleotides, nt), which is distinct from that of piRNAs (31 nt). The sequence of an endosiRNA is completely complementary to its target transcripts, whereas that of an miRNA is usually only partially complementary to its target (3739). Mature miRNAs are derived from precursor miRNAs that possess short stem-loop structures, whereas endo-siRNAs are processed from long dsRNAs without short stem-loop structures (3739). It is generally accepted that piRNAs are expressed during male gametogenesis where they play a crucial role, whereas endosiRNAs are essential during oocyte meiosis (3740). In contrast, miRNAs are ubiquitously expressed in somatic tissues and function throughout postimplantation development (3740).

Recent studies indicate that small non-coding RNAs undergo a complex, but finely tuned regulation in the early stages of cell reprogramming, i.e., during transition from a differentiated oocyte to a pluripotent blastomere. piRNAs and endo-siRNAs correspond to mRNAs or retrotransposons in growing oocytes (41). In oocytes, piRNAs are bound to Mili and regulate retrotransposons, while siRNAs are exclusively mapped to retrotransposons or other genomic regions that form dsRNA structures (41). The components of the piRNA pathway are also required for de novo methylation of the differentially methylated region (DMR) of the imprinted mouse Rasgrf1 locus, but not other paternally imprinted loci. A retrotransposon sequence within a non-coding RNA region that spans DMR was targeted by piRNAs generated from a different locus (42). Male germ cells express abundant endogenous siRNAs that can potentially target hundreds of transcripts and/or thousands of DNA regions in the genome, including retrotransposon sequences. This suggests the possibility of complex uncharacterized regulation imposed by small non-coding RNAs during male germ cell development (43). Interestingly, miRNAs were shown to be non-essential during preimplantation embryonic development and their function was suppressed during oocyte meiosis. Furthermore, endo-siRNAs, rather than miRNAs, play a role during oocyte maturation and preimplantation development (40,44,45). The introduction of miRNAs is sufficient to induce a pluripotent phenotype in differentiated somatic cells (46,47) without the addition of transcription factors. miRNA-induced reprogramming is probably related to the alteration of aggressive cancer phenotypes (32,48,49). Thus, these studies suggest that a fraction of endogenous siRNAs are involved in oocyte reprogramming. Future studies are required to search for endogenous siRNAs in meiotic oocytes, mitotic zygotes, and early embryos. A detailed understanding of these processes may lead to small non-coding RNAs proving beneficial in altering cancerous phenotypes.

Retrotransposon-type repetitive sequences are involved in cancer chromosome translocation. These repetitive sequences are characteristic genomic sequences in fragile sites commonly found in various chromosomes, including FRA3B and FRA16D (5054). Rare fragile sites are often associated with particular sequences (55). Fragile sites often coincide with genes that are frequently rearranged or deleted in human cancers. More than half of cancer-specific translocations contain breakpoints within fragile sites (56,57). Replication stress is usually caused by carcinogen exposure or other cancer-induction events, and the ataxia-telangiectasia-related (Atr) DNA damage checkpoint pathway plays a crucial role in maintaining genomic stability at fragile chromosome sites (58). Recent findings have confirmed that Atr protein binds to three regions of FRA3B during mild replication stress (57). During dysplasia, loss of Fhit expression leads to a direct and significant influence on changes in checkpoint proteins, suggesting a connection between Fhit loss and modulation of checkpoint activity (57,59). In cancer, changes in miRNA expression are frequently associated with changes at fragile chromosome sites (60). Interestingly, the DNA damage-susceptible FRA3B/FHIT chromosome fragile site encodes a protein required for protecting cells from accumulated DNA damage through its ability to modulate the checkpoint proteins Atr and Chk1, whereas inactivation of Fhit contributes to the accumulation of abnormal checkpoint phenotypes during cancer development (6163). The absence of Fhit protein in stem cells, a reduction in oxidative stress, and efficient but not error-free DNA damage repair, facilitate the unscheduled long-term survival of genotoxin-exposed Fhit-deficient hematopoietic stem cells that carry deleterious mutations (64). Thus, the small noncoding RNAs are nucleotides that can affect cancer risk. The catalog of known miRNAs and a large fraction of genomic ultraconserved regions (UCRs) that encode a particular set of non-coding RNAs located at fragile sites and/or cancer susceptibility loci, the expression of which is altered in human cancers, was recently updated (65). These types of studies are the first step toward discovering novel approaches for cancer therapies (65). The next logical research step would be to study the relationship between UCRs, the stemness-regulating region of CSCs, and the therapeutic control of CSCs and EMT (66,67). The regulation of cancer phenotypes by small noncoding RNAs has emerged as an important component of pharmacogenomics, and one day it may be used as an efficient therapeutic approach for cancer (6870).

Perspectives

The regulatory mechanism of cell differentiation in cancer is unclear, and it is debatable whether cancer cells can actually be reprogrammed. The cell of origin for cancer remains unknown, and hence, the underlying mechanism controlling initial programming during cancer remains elusive (32,48,71,72). Epigenetic modifications are involved in the aggressive behavior of cancer cells; therefore, utilizing these modifications for therapeutic purposes may be challenging.

References

1 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

2 

John B, Enright AJ, Aravin A, Tuschl T, Sander C and Marks DS: Human MicroRNA targets. PLoS Biol. 2:e3632004. View Article : Google Scholar

3 

Houbaviy HB, Murray MF and Sharp PA: Embryonic stem cell-specific microRNAs. Dev Cell. 5:351–358. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Lim LP, Glasner ME, Yekta S, Burge CB and Bartel DP: Vertebrate microRNA genes. Science. 299:15402003. View Article : Google Scholar : PubMed/NCBI

5 

Leung AK and Sharp PA: MicroRNA functions in stress responses. Mol Cell. 40:205–215. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Marsit CJ, Eddy K and Kelsey KT: MicroRNA responses to cellular stress. Cancer Res. 66:10843–10848. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Ng HH and Surani MA: The transcriptional and signalling networks of pluripotency. Nat Cell Biol. 13:490–496. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Calin GA, Dumitru CD, Shimizu M, et al: Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 99:15524–15529. 2002. View Article : Google Scholar : PubMed/NCBI

9 

Calin GA, Ferracin M, Cimmino A, et al: A MicroRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med. 353:1793–1801. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Cerchia L and De Franciscis V: Noncoding RNAs in cancer medicine. J Biomed Biotechnol. 2006:1–8. 2006. View Article : Google Scholar

11 

Cummins JM, He Y, Leary RJ, et al: The colorectal microRNAome. Proc Natl Acad Sci USA. 103:3687–3692. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Wijnhoven B, Michael MZ and Watson DI: MicroRNAs and cancer. Br J Surg. 94:23–30. 2007. View Article : Google Scholar

13 

Davis RL, Weintraub H and Lassar AB: Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 51:987–1000. 1987. View Article : Google Scholar : PubMed/NCBI

14 

Xie H, Ye M, Feng R and Graf T: Stepwise reprogramming of B cells into macrophages. Cell. 117:663–676. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Zhou Q, Brown J, Kanarek A, Rajagopal J and Melton DA: In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 455:627–632. 2008. View Article : Google Scholar : PubMed/NCBI

16 

French SW, Hoyer KK, Shen RR and Teitell MA: Transdifferentiation and nuclear reprogramming in hematopoietic development and neoplasia. Immunol Rev. 187:22–39. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Ungefroren H and Fändrich F: The programmable cell of monocytic origin (PCMO): a potential adult stem/progenitor cell source for the generation of islet cells. Adv Exp Med Biol. 654:667–682. 2010.PubMed/NCBI

18 

Takahashi K and Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 126:663–676. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K and Yamanaka S: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 131:861–872. 2007. View Article : Google Scholar

20 

Yamanaka S: Elite and stochastic models for induced pluripotent stem cell generation. Nature. 460:49–52. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar

22 

McKenna ES and Roberts CW: Epigenetics and cancer without genomic instability. Cell Cycle. 8:23–26. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Hanahan D and Weinberg RA: Hallmarks of cancer: the next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Rodríguez-Paredes M and Esteller M: Cancer epigenetics reaches mainstream oncology. Nat Med. 17:330–339. 2011.

25 

Cavenee W, Dryja TP, Phillips RA, et al: Expression of recessive alleles by chromosomal mechanisms in retinoblastoma. Nature. 305:779–784. 1983. View Article : Google Scholar : PubMed/NCBI

26 

Weinberg R: Tumor suppressor genes. Science. 254:1138–1146. 1991. View Article : Google Scholar : PubMed/NCBI

27 

Weinberg RA: The retinoblastoma protein and cell cycle control. Cell. 81:323–330. 1995. View Article : Google Scholar : PubMed/NCBI

28 

Wang JY: Cellular responses to DNA damage. Curr Opin Cell Biol. 10:240–247. 1998. View Article : Google Scholar : PubMed/NCBI

29 

Solinas G, Marchesi F, Garlanda C, Mantovani A and Allavena P: Inflammation-mediated promotion of invasion and metastasis. Cancer Metastasis Rev. 29:243–248. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Vogelstein B and Kinzler KW: The multistep nature of cancer. Trends Genet. 9:138–141. 1993. View Article : Google Scholar : PubMed/NCBI

31 

Bonavia R, Inda MD, Cavenee WK and Furnari FB: Heterogeneity maintenance in glioblastoma: a social network. Cancer Res. 71:4055–4060. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Miyoshi N, Ishii H, Nagai K, et al: Defined factors induce reprogramming of gastrointestinal cancer cells. Proc Natl Acad Sci USA. 107:40–45. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Friedlander SYG, Chu GC, Snyder EL, et al: Context-dependent transformation of adult pancreatic cells by oncogenic K-Ras. Cancer Cell. 16:379–389. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Orkin SH and Hochedlinger K: Chromatin connections to pluripotency and cellular reprogramming. Cell. 145:835–850. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Gurdon JB and Wilmut I: Nuclear transfer to eggs and oocytes. Cold Spring Harb Perspect Biol. 3:1–14. 2011. View Article : Google Scholar

36 

Maekawa M, Yamaguchi K, Nakamura T, et al: Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature. 474:225–229. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Nilsen TW: Endo-siRNAs: Yet another layer of complexity in RNA silencing. Nat Struct Mol Biol. 15:546–548. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Golden DE, Gerbasi VR and Sontheimer EJ: An inside job for siRNAs. Mol Cell. 31:309–312. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Okamura K and Lai EC: Endogenous small interfering RNAs in animals. Nat Rev Mol Cell Biol. 9:673–678. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Suh N and Blelloch R: Small RNAs in early mammalian development: from gametes to gastrulation. Development. 138:1653–1661. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Watanabe T, Totoki Y, Toyoda A, et al: Endogenous siRNAs from naturally formed dsRNAs regulate transcripts in mouse oocytes. Nature. 453:539–543. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Watanabe T, Tomizawa S, Mitsuya K, et al: Role for piRNAs and noncoding RNA in de novo DNA methylation of the imprinted mouse Rasgrf1 locus. Science. 332:848–852. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Song R, Hennig GW, Wu Q, Jose C, Zheng H and Yan W: Male germ cells express abundant endogenous siRNAs. Proc Natl Acad Sci USA. 108:13159–13164. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Suh N, Baehner L, Moltzahn F, Melton C, Shenoy A, Chen J and Blelloch R: MicroRNA function is globally suppressed in mouse oocytes and early embryos. Curr Biol. 20:271–277. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Ma J, Flemr M, Stein P, et al: MicroRNA activity is suppressed in mouse oocytes. Curr Biol. 20:265–270. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Anokye-Danso F, Trivedi CM, Juhr D, et al: Highly efficient miRNA-mediated reprogramming of mouse and human somatic cells to pluripotency. Cell Stem Cell. 8:376–388. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Miyoshi N, Ishii H, Nagano H, et al: Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 8:633–638. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Lin SL, Chang DC, Chang-Lin S, Lin CH, Wu DT, Chen DT and Ying SY: Mir-302 reprograms human skin cancer cells into a pluripotent ES-cell-like state. RNA. 14:2115–2124. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Lin SL, Chang DC, Ying SY, Leu D and Wu DT: MicroRNA miR-302 inhibits the tumorigenicity of human pluripotent stem cells by coordinate suppression of the CDK2 and CDK4/6 cell cycle pathways. Cancer Res. 70:9473–9482. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Sozzi G, Huebner K and Croce CM: FHIT in human cancer. Adv Cancer Res. 74:141–166. 1998. View Article : Google Scholar

51 

Huebner K and Croce CM: FRA3B and other common fragile sites: the weakest link. Nat Rev Cancer. 1:214–221. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Arlt MF, Casper AM and Glover TW: Common fragile sites. Cytogenet Genome Res. 100:92–100. 2003. View Article : Google Scholar

53 

Cimprich KA: Fragile sites: breaking up over a slowdown. Curr Biol. 13:R231–R233. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Durkin SG and Glover TW: Chromosome fragile sites. Annu Rev Genet. 41:169–192. 2007. View Article : Google Scholar

55 

Sutherland GR: Rare fragile sites. Cytogenet Genome Res. 100:77–84. 2003. View Article : Google Scholar

56 

Croce CM, Sozzi G and Huebner K: Role of FHIT in human cancer. J Clin Oncol. 17:1618–1624. 1999.PubMed/NCBI

57 

Dillon LW, Burrow AA and Wang YH: DNA instability at chromosomal fragile sites in cancer. Curr Genomics. 11:326–337. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Casper AM, Nghiem P, Arlt MF and Glover TW: ATR regulates fragile site stability. Cell. 111:779–789. 2002. View Article : Google Scholar : PubMed/NCBI

59 

Cirombella R, Montrone G, Stoppacciaro A, et al: Fhit loss in lung preneoplasia: relation to DNA damage response checkpoint activation. Cancer Lett. 291:230–236. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Calin GA, Sevignani C, Dumitru CD, et al: Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 101:2999–3004. 2004. View Article : Google Scholar : PubMed/NCBI

61 

Hu B, Wang H, Wang X, et al: Fhit and CHK1 have opposing effects on homologous recombination repair. Cancer Res. 65:8613–8616. 2005. View Article : Google Scholar : PubMed/NCBI

62 

Ishii H, Mimori K, Inoue H, et al: Fhit modulates the DNA damage checkpoint response. Cancer Res. 66:11287–11292. 2006. View Article : Google Scholar : PubMed/NCBI

63 

Okumura H, Ishii H, Pichiorri F, Croce CM, Mori M and Huebner K: Fragile gene product, Fhit, in oxidative and replicative stress responses. Cancer Sci. 100:1145–1150. 2009. View Article : Google Scholar : PubMed/NCBI

64 

Ishii H, Mimori K, Ishikawa K, et al: Fhit-deficient hematopoietic stem cells survive hydroquinone treatment long-term, carrying precancerous alterations. Cancer Res. 68:3662–3670. 2008. View Article : Google Scholar : PubMed/NCBI

65 

Rossi S, Sevignani C, Nnadi SC, Siracusa LD and Calin GA: Cancer-associated genomic regions (CAGRs) and noncoding RNAs: bioinformatics and therapeutic implications. Mamm Genome. 19:526–540. 2008. View Article : Google Scholar : PubMed/NCBI

66 

Wang Z, Li Y, Ahmad A, Azmi AS, Kong D, Banerjee S and Sarkar FH: Targeting miRNAs involved in cancer stem cell and EMT regulation: An emerging concept in overcoming drug resistance. Drug Resist Updat. 13:109–118. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Zimmerman AL and Wu S: MicroRNAs, cancer and cancer stem cells. Cancer Lett. 300:10–19. 2011. View Article : Google Scholar : PubMed/NCBI

68 

Cortez MA, Ivan C, Zhou P, Wu X, Ivan M and Calin GA: microRNAs in cancer: from bench to bedside. Adv Cancer Res. 108:113–157. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Garzon R, Marcucci G and Croce CM: Targeting microRNAs in cancer: Rationale, strategies and challenges. Nat Rev Drug Discov. 9:775–789. 2010. View Article : Google Scholar : PubMed/NCBI

70 

White NM, Fatoohi E, Metias M, Jung K, Stephan C and Yousef GM: Metastamirs: a stepping stone towards improved cancer management. Nat Rev Clin Oncol. 8:75–84. 2011. View Article : Google Scholar : PubMed/NCBI

71 

Kasemeier-Kulesa JC, Teddy JM, Postovit LM, Seftor EA, Seftor RE, Hendrix MJ and Kulesa PM: Reprogramming multipotent tumor cells with the embryonic neural crest micro-environment. Dev Dyn. 237:2657–2666. 2008. View Article : Google Scholar : PubMed/NCBI

72 

Wang J, Emadali A, Le Bescont A, Callanan M, Rousseaux S and Khochbin S: Induced malignant genome reprogramming in somatic cells by testis-specific factors. Biochim Biophys Acta. 1809:221–225. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Yu J, Vodyanik MA, Smuga-Otto K, et al: Induced pluripotent stem cell lines derived from human somatic cells. Science. 318:1917–1920. 2007. View Article : Google Scholar : PubMed/NCBI

74 

Okita K, Nakagawa M, Hyenjong H, Ichisaka T and Yamanaka S: Generation of mouse induced pluripotent stem cells without viral vectors. Science. 322:949–953. 2008. View Article : Google Scholar : PubMed/NCBI

75 

Stadtfeld M, Nagaya M, Utikal J, Weir G and Hochedlinger K: Induced pluripotent stem cells generated without viral integration. Science. 322:945–949. 2008. View Article : Google Scholar : PubMed/NCBI

76 

Aoi T, Yae K, Nakagawa M, et al: Generation of pluripotent stem cells from adult mouse liver and stomach cells. Science. 321:699–702. 2008. View Article : Google Scholar : PubMed/NCBI

77 

Kim JB, Zaehres H, Wu G, Gentile L, et al: Pluripotent stem cells induced from adult neural stem cells by reprogramming with two factors. Nature. 454:646–650. 2008. View Article : Google Scholar : PubMed/NCBI

78 

Huangfu D, Maehr R, Guo W, Eijkelenboom A, Snitow M, Chen AE and Melton DA: Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds. Nat Biotechnol. 26:795–797. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Wernig M, Lengner CJ, Hanna J, et al: A drug-inducible transgenic system for direct reprogramming of multiple somatic cell types. Nat Biotechnol. 26:916–924. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Sun N, Panetta NJ, Gupta DM, et al: Feeder-free derivation of induced pluripotent stem cells from adult human adipose stem cells. Proc Natl Acad Sci USA. 106:15720–15725. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Zhou H, Wu S, Joo JY, et al: Generation of induced pluripotent stem cells using recombinant proteins. Cell Stem Cell. 4:381–384. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Shi Y, Do JT, Desponts C, Hahm HS, Schöler HR and Ding S: A combined chemical and genetic approach for the generation of induced pluripotent stem cells. Cell Stem Cell. 2:525–528. 2008. View Article : Google Scholar : PubMed/NCBI

83 

Woltjen K, Michael IP, Mohseni P, et al: piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature. 458:766–770. 2009. View Article : Google Scholar : PubMed/NCBI

84 

Judson RL, Babiarz JE, Venere M and Blelloch R: Embryonic stem cell-specific microRNAs promote induced pluripotency. Nat Biotechnol. 27:459–461. 2009. View Article : Google Scholar : PubMed/NCBI

85 

Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P and Woltjen K: Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature. 458:771–775. 2009. View Article : Google Scholar : PubMed/NCBI

86 

Seki T, Yuasa S, Oda M, et al: Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells. Cell Stem Cell. 7:11–14. 2010. View Article : Google Scholar : PubMed/NCBI

87 

Warren L, Manos PD, Ahfeldt T, et al: Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell. 7:618–630. 2010. View Article : Google Scholar : PubMed/NCBI

88 

Zhu S, Li W, Zhou H, et al: Reprogramming of human primary somatic cells by OCT4 and chemical compounds. Cell Stem Cell. 7:651–655. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Li Z, Yang CS, Nakashima K and Rana TM: Small RNA-mediated regulation of iPS cell generation. EMBO J. 30:823–834. 2011. View Article : Google Scholar : PubMed/NCBI

90 

Subramanyam D, Lamouille S, Judson RL, Liu JY, Bucay N, Derynck R and Blelloch R: Multiple targets of miR-302 and miR-372 promote reprogramming of human fibroblasts to induced pluripotent stem cells. Nat Biotechnol. 29:443–448. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Li L, Connelly MC, Wetmore C, Curran T and Morgan JI: Mouse embryos cloned from brain tumors. Cancer Res. 63:2733–2736. 2003.PubMed/NCBI

92 

Hochedlinger K, Blelloch R, Brennan C, Yamada Y, Kim M, Chin L and Jaenisch R: Reprogramming of a melanoma genome by nuclear transplantation. Genes Dev. 18:1875–1885. 2004. View Article : Google Scholar : PubMed/NCBI

93 

Blelloch RH, Hochedlinger K, Yamada Y, et al: Nuclear cloning of embryonal carcinoma cells. Proc Natl Acad Sci USA. 101:13985–13990. 2004.PubMed/NCBI

94 

Postovit LM, Margaryan NV, Seftor EA, et al: Human embryonic stem cell microenvironment suppresses the tumorigenic phenotype of aggressive cancer cells. Proc Natl Acad Sci USA. 105:4329–4334. 2008. View Article : Google Scholar : PubMed/NCBI

95 

Utikal J, Maherali N, Kulalert W and Hochedlinger K: Sox2 is dispensable for the reprogramming of melanocytes and melanoma cells into induced pluripotent stem cells. J Cell Sci. 122:3502–3510. 2009. View Article : Google Scholar : PubMed/NCBI

96 

Carette JE, Pruszak J, Varadarajan M, Blomen VA and Gokhale S: Generation of iPSCs from cultured human malignant cells. Blood. 115:4039–4042. 2010. View Article : Google Scholar : PubMed/NCBI

97 

Mathieu J, Zhang Z, Zhou W, Wang AJ and Heddleston JM: HIF induces human embryonic stem cell markers in cancer cells. Cancer Res. 71:4640–4652. 2011. View Article : Google Scholar : PubMed/NCBI

98 

Allegrucci C, Rushton MD, Dixon JE, et al: Epigenetic reprogramming of breast cancer cells with oocyte extracts. Mol Cancer. 10:72011.PubMed/NCBI

Related Articles

Journal Cover

July 2012
Volume 4 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Nishikawa S, Ishii H, Haraguchi N, Kano Y, Fukusumi T, Ohta K, Ozaki M, Dewi D, Sakai D, Satoh T, Satoh T, et al: microRNA-based cancer cell reprogramming technology (Review). Exp Ther Med 4: 8-14, 2012
APA
Nishikawa, S., Ishii, H., Haraguchi, N., Kano, Y., Fukusumi, T., Ohta, K. ... Mori, M. (2012). microRNA-based cancer cell reprogramming technology (Review). Experimental and Therapeutic Medicine, 4, 8-14. https://doi.org/10.3892/etm.2012.558
MLA
Nishikawa, S., Ishii, H., Haraguchi, N., Kano, Y., Fukusumi, T., Ohta, K., Ozaki, M., Dewi, D., Sakai, D., Satoh, T., Nagano, H., Doki, Y., Mori, M."microRNA-based cancer cell reprogramming technology (Review)". Experimental and Therapeutic Medicine 4.1 (2012): 8-14.
Chicago
Nishikawa, S., Ishii, H., Haraguchi, N., Kano, Y., Fukusumi, T., Ohta, K., Ozaki, M., Dewi, D., Sakai, D., Satoh, T., Nagano, H., Doki, Y., Mori, M."microRNA-based cancer cell reprogramming technology (Review)". Experimental and Therapeutic Medicine 4, no. 1 (2012): 8-14. https://doi.org/10.3892/etm.2012.558