MicroRNAs: key players in the immune system, differentiation ... - Nature

15 downloads 273 Views 300KB Size Report
Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen SM. (2003). bantam encodes ..... Hughes PD, Belz GT, Fortner KA, Budd RC, Strasser A, Bouillet P. (2008).
Oncogene (2008) 27, 5959–5974

& 2008 Macmillan Publishers Limited All rights reserved 0950-9232/08 $32.00 www.nature.com/onc

REVIEW

MicroRNAs: key players in the immune system, differentiation, tumorigenesis and cell death R Schickel, B Boyerinas, S-M Park and ME Peter The Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA

Micro (mi)RNAs are small, highly conserved noncoding RNAs that control gene expression post-transcriptionally either via the degradation of target mRNAs or the inhibition of protein translation. Each miRNA is believed to regulate the expression of multiple mRNA targets, and many miRNAs have been linked to the initiation and progression of human cancer. miRNAs control various activities of the immune system and different stages of hematopoietic development, and their misexpression is the cause of various blood malignancies. Certain miRNAs have oncogenic activities, whereas others have the potential to act as tumor suppressors. Because they control fundamental processes such as differentiation, cell growth and cell death, the study of the role of miRNAs in human neoplasms holds great promise for novel forms of therapy. Here, we summarize the role of miRNAs and their targets in contributing to human cancers and their function as regulators of apoptotic pathways and the immune system. Oncogene (2008) 27, 5959–5974; doi:10.1038/onc.2008.274 Keywords: apoptosis; progression

miRNA;

oncomiRs;

tumor

Introduction Micro RNAs (miRNAs) are small noncoding RNAs of about 19–22 nucleotides (nt) that regulate protein expression by posttranscriptional silencing. The relevance of this class of novel small RNA regulators has only become clear over the past few years. However, it was realized early on that miRNAs are fundamental regulators of cellular processes that have physiological significance, and hence deregulation of various miRNAs is emerging as an important contributor to many human diseases including cancer. It was long known in the Caenorhabditis elegans community that a decrease in the expression of the heterochronic gene lin-14 that regulates developmental timing was critical for early larval stage transition, and that the lin-4 gene was required to reduce lin-14 levels and allow the larval transition to Correspondence: Professor ME Peter, The Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA. E-mail: [email protected]

occur (Chalfie et al., 1981). The mechanism of repression was not known until groundbreaking work by two groups showed that repression occurred through complementary binding of the small RNA lin-4 to the 30 -UTR (untranslated region) of the lin-14 mRNA (Lee et al., 1993; Wightman et al., 1993). Subsequent discovery of the small non-coding RNA let-7 in C. elegans as a critical regulator in the determination of developmental cellular fate (Reinhart et al., 2000), and concurrent discovery of conserved sequence and temporal expression patterns of let-7-related genes across multiple species (Pasquinelli et al., 2000), suggested that these miRNAs may act as fundamental developmental regulators. In addition, these findings opened the floodgates of information to the understanding that miRNAs act as key participants in cellular differentiation. Later work revealed or suggested the role of miRNAs in neuronal patterning (Johnston and Hobert, 2003), lineage commitment in hematopoiesis (Chen et al., 2004), tissue homeostasis (Cui et al., 2006; Plaisance et al., 2006; Rodriguez et al., 2007) and apoptosis (Baehrecke, 2003; Cimmino et al., 2005). Currently, there are 678 mature human miRNA sequences listed in the miRNA registry (http://www. microrna.sanger.ac.uk/sequences) with approximately 1000 predicted miRNAs, each potentially targeting approximately 200 genes (Lewis et al., 2003). Lewis et al. (2005) further refined the identification of miRNA target sites and found that 5300 of 17 850 genes in their data set contained conserved miRNA target sites. Therefore, the possibility exists that >30% of the human genome may be under the translational regulation of miRNAs. The emerging role of miRNAs in such a diverse and fundamental set of cellular mechanisms clearly suggests that proper control of these regulatory elements is essential for the maintenance of a non-pathologic state. A basic understanding of cancer formation revolves around the oncogene/tumor suppressor model of carcinogenesis—with RAS as an example of a classic oncogene and p53 the most common example of a tumor suppressor that inhibits cellular proliferation and induces apoptosis. Despite the simplicity of this model, it provides a solid foundation for an understanding of how two main classes of proteins function in the development of cancer. We are now beginning to understand miRNAs as master regulators that, in some cases, can act as either an oncogene or a tumor

Regulation by miRNAs R Schickel et al

5960

suppressor, whereas in other cases, they can affect both phases of tumorigenesis. Loss of let-7, for example, contributes to multiple facets of tumor progression by wholesale alteration of differentiation status. As our understanding of miRNA biology continues to increase, we will likely need to comprehend miRNA function more on a systems level, where linear relationships between miRNAs and target genes are less important than changes in the overall gene expression pattern induced by altered miRNA levels.

miRNA biogenesis miRNAs negatively regulate protein expression at the point of protein translation. In the following, we will cover the basic pathway of miRNA biogenesis and maturation as well as the mechanism of translational repression. These topics have been reviewed in detail elsewhere (Bartel, 2004; Kim, 2005; Engels and Hutvagner, 2006; Zeng, 2006). miRNA biogenesis requires several posttranscriptional processing steps to yield the functional mature miRNA. Primary miRNAs (pri-miRNAs) are generally >1 kb transcriptional products of RNA polymerase-II and contain a 50 7-methylguanosine cap and 30 poly-A tail (Cullen, 2004). Key to miRNA production is a hairpin within an imperfectly base-paired double-stranded RNA. The hairpin of the pri-miRNA is recognized by the nuclear RNAse-III enzyme Drosha and its cofactor DGCR8. The Drosha/DGCR8 complex cleaves the pri-miRNA to an approximately 70 nt double-stranded RNA hairpin pre-miRNA with a 2 nt 30 overhang. The 2 nt 30 overhang is recognized by exportin-5 and the Ran-GTPase, mediating the pre-miRNA nuclear export (Zeng and Cullen, 2006). Once in the cytoplasm, the pre-miRNA is further digested by Dicer, the RNAse III enzyme of the RNA silencing pathway, yielding the B22 nt mature miRNA that enters the RNA-induced silencing complex (RISC) complex to mediate mRNA translational repression. Translational repression is a consequence of the miRNA/RISC complex binding to a target site in the 30 -UTR of the targeted mRNA that is complementary to the 50 -miRNA seed sequence, but usually not completely complementary along the entire miRNA/mRNA complex (Lai, 2002). Binding of the RISC/miRNA complex to the 30 -UTR of a target mRNA results in steric hindrance of the translational machinery and a consequent inability to translate the message. In the case of perfect complementarity between an miRNA and its target, the mRNA is degraded through the RNA silencing mechanism in a process distinct from miRNA-mediated translational repression (Engels and Hutvagner, 2006). The location, number and proximity of the miRNA binding sites along a target mRNA are all relevant in the translational repression, and this allows for prediction and ranking of putative miRNA targets based solely on sequence information (Grimson et al., 2007; Nielsen et al., 2007). Oncogene

miRNAs in hematopoiesis and immune function The study of miRNAs is now moving beyond descriptions of target analysis and toward a global understanding of how miRNAs impact entire developmental and differentiation processes. Several groups have targeted components within the miRNA-processing pathway and determined the effects of general loss of miRNA on development, differentiation, homeostasis and tumorigenesis. Particularly, it was found that Dicer-null mouse embryos fail to survive beyond embryonic day 7.5 and that Dicer is required for murine stem cell differentiation (Bernstein et al., 2003). Furthermore, knocking down Dicer implicated miRNAs in the maintenance of skeletal muscle development (O’Rourke et al., 2007), transformation and tumorigenesis (Kumar et al., 2007). Hematopoiesis represents an elegant developmental model to observe normal changes in miRNA expression and to test the effects of miRNA levels on differentiation and tumorigenesis. There are a number of miRNAs that are prominently expressed in hematopoietic cells (for example, miR-142, miR-146, miR-150, miR-155, miR-181 and miR-223) (Chen et al., 2004; Ramkissoon et al., 2006). Differential expression of miRNAs during hematopoiesis is most likely involved in the regulation of hematopoietic differentiation. One such observation implicates miR-221 and miR-222 in maintaining hematopoietic stem cells by preventing the formation of early erythroblasts by directly targeting the receptor c-kit (Felli et al., 2005). Thus, downmodulation of these miRNAs is believed to allow the formation of early erythroblasts. Furthermore, the introduction of miR-221 or miR-222 also leads to an increase in cell death at a time point consistent with that of the terminal maturation of erythrocytes. This mechanism may be an evolutionarily conserved response to protect cells from temporal misregulation and de-differentiation during erythropoiesis. Contrary to increasing cell death, miR-221 and miR-222 were shown to directly target p27Kip1 in a prostate tumor cell line model, resulting in proliferation and increased oncogenesis (Galardi et al., 2007; Gillies and Lorimer, 2007; le Sage et al., 2007). Therefore, it appears that these miRNAs, which help maintain a normal stem cell state during erythropoiesis, act as oncogenes when misexpressed in a prostate tumor model, supporting the notion that miRNAs most likely have unique temporal regulatory roles during both differentiation and normal physiologic responses. The descriptions of miR-181a and miR-150 clearly support the multifunctional physiologic roles of miRNAs. miR-181a directs hematopoietic stem cells toward differentiation into B cells (Chen et al., 2004). In addition, it was shown to modulate the intrinsic sensitivity of T cells to antigen by targeting a number of phosphatases that are known negative regulators of T-cell responses (Li et al., 2007). Furthermore, miR-150, found to exist at high levels in mature B and T cells but not their progenitors (Zhou et al., 2007), is required for mature B-cell development and the attenuation of the humoral immune response by translational repression of

Regulation by miRNAs R Schickel et al

5961

c-Myb (Xiao et al., 2007). The physiologic expression of mir-150 has a significant impact on limiting antibody production upon activation. Therefore, miR-181a and miR-150 have at least dual roles in normal physiologic processes. Elevated c-Myc expression is common in many cancers (Grandori et al., 2000). A hallmark of Burkitt’s lymphoma is a chromosomal translocation juxtaposing the c-myc and immunoglobulin genes and yielding high c-MYC expression (Janz, 2006). Furthermore, c-Myc is known to drive the expression of the pro-tumorigenic miR-17-92 cluster of miRNAs (O’Donnell et al., 2005; Dews et al., 2006), and a high level of c-Myc also leads to a decrease of multiple miRNAs known to have anti-tumorigenic properties (Chang et al., 2008). Chang et al. found that a majority of the c-Myc-repressed miRNAs provided a selective disadvantage when stably re-expressed in Myc3 cells, a B-cell lymphoma cell line, or in 38B9 cells, an Abl1-transformed pro-B cell line, and analysed 3 weeks after injection in an SCID mouse model. The miR-17–92 cluster is a group of miRNAs (consisting of miR-17-5p, miR-18, miR-19a, miR-20 and miR-92) located on chromosome 13q31 that is transcribed as a polycistronic unit. miR-17-92 members have been described to target p21, a critical regulator of G1–S phase checkpoint, as well as the pro-apoptotic factor BIM, allowing cells to bypass the DNA damage checkpoint and obtain a survival advantage. The 17–92 miRNA cluster also targets anti-angiogenic factors Tsp1 and connective tissue growth factor (CTGF), resulting in an increase in angiogenesis in solid tumors. Recently, it was shown that this cluster regulates monocyte differentiation by targeting the transcription factor AML1 (Fontana et al., 2007), which is required for the expression of the macrophage colony stimulating factor (M-CSF) receptor. Taken together, these studies suggest that elevated c-Myc levels can contribute to multiple levels of tumorigenesis by direct modulation of miR-17-92 levels. miR-223 has been shown to be involved in the differentiation of granulocytes (Fazi et al., 2005), and the transcription of miR-223 is competitively regulated by nuclear factor I/A (NFIA) and CCAAT/enhancer binding protein a (CEBPA); NFIA transcriptionally represses the expression of miR-223, whereas CEBPA causes the upregulation of miR-223 in response to differentiation induced by retinoic acid. Interestingly, miR-223 specifically targets NFIA favoring its own expression upon induced differentiation. Furthermore, CEBPA binding to the miR-223 promoter is required for the retinoic acid response, suggesting that miR-223 has a significant role in induced differentiation and that NFIA levels are critical in blocking this process. In most cases, the functions of miRNAs were identified by studying predicted targets, and only a few examples exist in which a functional role was established without knowing a putative target. An example of a miRNA that was studied both in terms of its regulation and its targets is miR-146a, which has an important function in the innate immune system. miR-146a was found to be upregulated in cells treated with lipopolysaccharide

(LPS) through the activation of toll-like receptor (TLR) 4 and concurrent activation of the nuclear factor-kB pathway (Taganov et al., 2006). Interestingly, two of its main predicted targets, interleukin-1 (IL-1) receptorassociated kinase 1 (IRAK1) and TRAF6, are both signaling components of the TLRs. Both were confirmed to be direct targets of miR-146a, placing this miRNA into a negative feedback loop that limits inflammatory signaling and could potentially cause chronic inflammation or become detrimental through the induction of septic shock if it became deregulated. Another TLR-responsive gene, miR-155, was identified to respond to the stimulation of TLR3 on macrophages (O’Connell et al., 2007) and immunoglobulin heavy chain enhancer (Em-miR-155 transgenic mice were hypersensitive to LPS/D-galactosamineinduced septic shock; Tili et al., 2007). Similar to miR-146a, evidence suggests that the putative targets of miR-155 are components of the TLR signaling machinery. Because miR-155 is best known through its involvement in the development of B-cell malignancies (see below), this discovery adds to the connection between inflammation and cancer that has recently attracted significant attention (Karin et al., 2006). In addition to its function in monocytes, miR-155 was shown to have a physiological function in B and T cells (Thai et al., 2007). miR-155-deficient mice have a defect in the germinal center (GC) reaction and a concurrent weak T-cell-dependent antibody response. It was suggested that this might be due to an impaired production of tumor necrosis factor a (TNFa) by the B cells that is required for a proper GC response. In the T-cell compartment, these mice were skewed toward TH2 differentiation, as their T cells produced more IL-4 and less interferon-g than T cells from control mice (Thai et al., 2007). These data illustrate that miRNAs play important roles in the development of various bloodderived cell types and often are part of complex feedback regulatory networks. Interestingly, in many reported cases, it is not the miRNA that drives differentiation, but rather miRNAs are expressed to prevent de-differentiation and/or maintain pools of cells at certain stages of differentiation. Hence, their downregulation seems to assist in driving cells down certain de-differentiation paths (Georgantas et al., 2007). OncomiRs as tumor suppressors and oncogenes We have just described the important role of various miRNAs in immune function and hematopoietic fate determination, and have suggested that the deregulation of this control system can lead to oncogenesis and other pathologies. Not surprisingly, multiple pieces of evidence point at miRNAs playing a profound role in cancer formation and progression (Table 1). Gene expression array analyses as well as real-time PCR analyses of cancers reveal cancer specific changes in the expression of miRNAs, when compared with noncancerous tissues (Liu et al., 2004; Lu et al., 2005; Volinia et al., 2006). In addition, an expression analysis of 217 Oncogene

5962

Oncogene

Table 1 miRNAs with relevance in cancer and cell death Normal function

Cancer relevance

Cell death relevance

Targets

References

Let-7/miR-98

Upregulated late during Caenorhabditis elegans development; increased expression in differentiated cells; may act to maintain differentiated states

Downregulated in lung colon and ovarian cancer; expression inversely correlates to patient survival in ovarian cancer, NSCLC and adenocarcinoma; possible growth suppressor in colon carcinoma (let-7a-1)

Contributes to IL-6-directed survival signaling and reduces chemotherapymediated cell death in human malignant cholangiocytes (let-7a)

HMGA2, RAS, IGF2BP1, c-Myc, NF2 and LIN28Ba

miR-1

Involved in cardiogenic and myogenic differentiation; involved in maintaining proper cardiac depolarization Upregulated in primary brain tissue upon introduction of reactive oxygen species production Possible inhibitor of megakaryocytic differentiation

Pro-apoptotic effect in response to oxidative stress

HSP60, HSP70, KCNJ2 and GJA1

Akao et al. (2006); Boyerinas et al. (2008); Gramantieri et al. (2007); Guo et al. (2006); Hebert et al. (2007); Iorio et al. (2005); Johnson et al. (2005); Lee et al. (2006a); Lee and Dutta (2007); Lu et al. (2007a); Mayr et al. (2007); Meng et al. (2007b); Park et al. (2007); Pasquinelli et al. (2000); Reinhart et al. (2000); Sampson et al. (2007); Shell et al. (2007); Yanaihara et al. (2006) Chen et al. (2006); Xu et al. (2007); Yang et al. (2007)

miR-9 miR-10a miR-10b miR-15a

Regulation of pancreatic regeneration

miR-16

Involved in the regulation of pancreatic regeneration; negatively regulates cell cycle progression and cell growth

miR-21

miR-29

Suggested to participate in maintaining proper DNA methylation

miR-34a

p53 inducible; translationally regulates cell cycle, anti-apoptotic and check point genes; shown to induce growth arrest

miR-34b/c

p53 inducible; translational regulation of proliferation and adhesion-independent growth

miR-122

Hepatocellular-specific miRNA highly expressed in the normal liver and involved in fatty acid and cholesterol metabolism

Hypermethylated in breast cancer

Inversely correlates with prognosis and initiates invasion and metastasis in breast cancer cell lines Frequently deleted or downregulated in CLL patients Frequently deleted or downregulated in CLL patients

Granuphilin (Slp4)

Pro-apoptotic effects mediated through targeting BCL2 Pro-apoptotic effects mediated through BCL2

HOXA1

Bredenkamp et al. (2007); Lehmann et al. (2008); Lukiw and Pogue (2007); Plaisance et al. (2006) Garzon et al. (2006)

HOXD10

Ma et al. (2007)

BCL2 and NGN3a

Calin et al. (2005); Cimmino et al. (2005); Joglekar et al. (2007)

BCL2 and NGN3a

Calin et al. (2005); Cimmino et al. (2005); Joglekar et al. (2007); Linsley et al. (2007) Asangani et al. (2008); Chan et al. (2005); Frankel et al. (2007); Loffler et al. (2007); Meng et al. (2007a); Roldo et al. (2006); Si et al. (2007); Zhu et al. (2007) Fabbri et al. (2007); Mott et al. (2007); Pekarsky et al. (2006)

Widely overexpressed in cancer; promotes growth; shown to induce a metastatic phenotype

Shown to inhibit apoptosis

PTEN, TPM1 and PDCD4

Reduced levels in B-CLL and NSCLC, enforced expression restores normal methylation in NSCLC and reduces tumorigenicity in vitro and in vivo Decreased expression in lung and colon cancer, and neuroblastomas; shown to be highly expressed in CLL but not in ALL; gene locus is subject to heterozygous deletions in some tumors Decreased/loss of expression in p53 null tumors and described to inhibit neoplastic growth; often deleted in NSCLC

Increases apoptosis sensitivity to TRAIL

TCL1 and Mcl-1

Increases p53-mediated apoptotic sensitivity

CDK4, E2F3 and BCL2a

He et al. (2007); Raver-Shapira et al. (2007); Tazawa et al. (2007); Versteeg et al. (1995); Welch et al. (2007); Zanette et al. (2007)

Increases apoptotic sensitivity possibly through translational or indirect repression of anti-apoptotic factors

CDK6, c-Meta and c-Myca

Bommer et al. (2007); Corney et al. (2007); Raver-Shapira et al. (2007)

CCNG1 and CAT-1ba

Chang et al. (2004); Esau et al. (2006); Gramantieri et al. (2007); Kutay et al. (2006)

Commonly downregulated in HCC; loss suggested to facilitate genomic instability due to the loss of translational repression of CCNG1, a p53 pathway antagonist

Regulation by miRNAs R Schickel et al

miRNA

Table 1 Continued miRNA

Normal function

Cancer relevance

miR-126

Expressed in endothelial cells

miR-130a

Involved in megakaryocytic differentiation; suggested to inhibit differentiation by targeting transcription factors that mediate megakaryocyte differentiation in an ERK-dependent manner Promotes myoblast proliferation and inhibits cardiac hypertrophy

Downregulated in metastasizing breast cancer Inhibits pro-angiogenic factors, suppressing angiogenesis in vascular endothelial cells

miR-133

Highly expressed in hematopoietic tissues and fetal liver

miR-143

Required for adipocyte differentiation

miR-146

NF-kB inducible, attenuates the TLR4 signaling pathway Controls B-cell differentiation in vivo; highly expressed in mature B and T cells Regulates cytokine expression, optimizing germinal center T-cell response; JNK-induced expression during the macrophage response and in activated B cells; involved in maintaining erythroblasts B-cell linage; increases T-cell sensitivity to antigens and exhibits increased expression in mature T cells

miR-150 miR-155

miR-181a

miR-195

miR-196 miR-221/222

miR-223 miR-335

Stress-responsive miRNA involved in cardiac tissue growth and hypertrophy; mediates pancreatic postoperative regeneration Involved in developmental patterning and myeloid differentiation Maintains progenitor cells in erythropoiesis; suggested to promote proliferation Regulator of myelopoiesis; suggested involvement in chorioamniotic membrane development

Targets

References

VCAM-1

Harris et al. (2008); Tavazoie et al. (2008)

GAX, HOXA5 and MAFB

Chen and Gorski (2008); Garzon et al. (2006); Sevinsky et al. (2004)

Inhibits intrinsically SRF and caspase-9 activated apoptosis mediated through caspase-9 Locus is a common translocation site in aggressive B-cell leukemia generating a miR-142-myc fusion transcript, producing high levels of pre-miR-142 Downregulated in B-cell leukemias and colorectal neoplasia; reduced expression led to an increase in miR-21 expression; strongly downregulated in multiple tumor types Upregulated in papillary thyroid carcinoma

Overexpressed in multiple tumor types, particularly in leukemia; suspected oncogenic effect due to translational repression of a known tumor suppressor

Chen et al. (2004); Gauwerky et al. (1989)

Akao et al. (2007); Esau et al. (2004); Lui et al. (2007); Michael et al. (2003); Yanaihara et al. (2006) IRAK1 and TRAF6 c-Myb Inhibits TP53INP1mediated apoptosis

Induces polyclonal premalignant expansion; loss of expression noted in transition to aggressive B-cell chronic lymphoma Highly expressed in CLL

AGTR1 and TP53INP1

TCL1, HOXA11, SHP-2, DUSP5, DUSP6 and PTPN-22 NGN3a

HOXB8 Inhibits cell growth in erythroleukemic cell lines; upregulated in glioblastoma, papillary thyroid carcinoma, prostate and pancreatic tumors Epigenetically silenced by AML1/ ETO, an oncogenic protein; increased expression in bladder cancer Downregulated in metastasizing breast cancer; inhibits metastatic cell invasion

Care et al. (2007); Chen et al. (2006); Xu et al. (2007)

c-Kit and p27(Kip1)

Pro-apoptotic factor antagonizing miR-21

SOX4 and Tenascin C Jagged-1a

He et al. (2005a); Taganov et al. (2006) Xiao et al. (2007); Zhou et al. (2007) Regulation by miRNAs R Schickel et al

miR-142

Cell death relevance

Eis et al. (2005); Georgantas et al. (2007); Gironella et al. (2007); Lawrie et al. (2007); Masaki et al. (2007); Metzler et al. (2004); O’Connell et al. (2007); Ramkissoon et al. (2006); Sethupathy et al. (2007); Thai et al. (2007) Chen et al. (2004); Ciafre et al. (2005); Costinean et al. (2006); Li et al. (2007); Marton et al. (2008); Naguibneva et al. (2006); Pekarsky et al. (2006) Joglekar et al. (2007); van Rooij et al. (2006); Zanette et al. (2007) Hornstein et al. (2005); Kawasaki and Taira (2004); Yekta et al. (2004) Ciafre et al. (2005); Felli et al. (2005); Galardi et al. (2007); Gillies and Lorimer (2007); He et al. (2005a); le Sage et al. (2007); Lee et al. (2007) Fukao et al. (2007); Garzon et al. (2006); Gottardo et al. (2007); Ramkissoon et al. (2006) Sathyan et al. (2007); Tavazoie et al. (2008)

5963

Oncogene

Regulation by miRNAs R Schickel et al

Oncogene

Abbreviations: ALL, acute lymphocytic leukemia; CLL, chronic lymphocytic leukemia; ERK, extracellular signal-regulated kinase; HCC, hepatocellular carcinoma; IL, interleukin; JNK, Jun N-terminal kinase; miRNA, micro RNA; NSCLC, non-small-cell lung cancer. a Predicted target.

Dews et al. (2006); Hayashita et al. (2005); He et al. (2005b); Landais et al. (2007); Lu et al. (2007b); Matsubara et al. (2007); O’Donnell et al. (2005); Venturini et al. (2007); Zanette et al. (2007) miR-17-92 cluster 17-5p Shown to inhibit differentiation of lung 18 tissue; c-Myc inducible 19a 20a 92

AIB1, AML1, MCSFR and Rb2 Inhibition of miRNA cluster leads to an increase in apoptosis Limits tumor growth in breast cancer but promotes proliferation in lung epithelial progenitor cells; increased expression in B-cell lymphoma and facilitated cancer progression in a lymphoma murine model

Fontana et al. (2007); Hossain et al. (2006); Landais et al. (2007); Matsubara et al. (2007); Sylvestre et al. (2007); Volinia et al. (2006) AML1, M-CSFR, Rb, Mylip, Rbp1like and HIPK3a Implicated in inducing apoptosis

miR-372/373

miR-106-363 cluster 106a Key regulators of monocytopoiesis; 19b-2 expression induced under c-Myc 92-2 and N-Myc regulation 20b 363

Overexpressed in B50% of T-cell leukemias; high expression levels noted in multiple solid tumors; downregulated in CML; overexpression enhances blast proliferation

Voorhoeve et al. (2007) LATS2 Inhibits p53 directed CDK signaling, promoting tumorigenesis in testicular germ cells

Cell death relevance miRNA

Normal function

Cancer relevance

Table 1 Continued

Targets

References

5964

miRNAs in various human cancers clearly described the developmental origin of tumors based on miRNA profile, and it also identified a global downregulation of miRNAs in tumors regardless of cell type (Lu et al., 2005). It has also been found that the induction of miRNAs coincides with normal cellular differentiation, supporting the hypothesis that miRNA loss leads to a de-differentiation process and less-differentiated tumors (Lu et al., 2005). Interestingly, Lu et al. showed that the expression levels of the 217 miRNAs allowed for a better stratification of cancer material, when compared with information obtained from a gene array analysis of about 16 000 mRNAs. The prognostic power of these miRNA profiles is quite striking and strongly argues for the ultimate importance of miRNAs in developing and maintaining cellular fates. It is then quite easy to envisage how gain or loss of a single miRNA or a set of miRNAs in either differentiated adult tissues or more differentiated tumors could both induce and further neoplastic progression. Further support for the relevance of miRNAs in cancer came from studies that demonstrated that half of the known miRNAs are located in regions of chromosomal instability associated with cancer (Calin et al., 2004), and that a high proportion of genomic loci containing miRNA genes shows DNA copy number alterations (Zhang et al., 2006). As we have described above, specific roles of miRNAs in certain cancers have been established, and many miRNA targets are emerging that allow one to explain or predict some of the activities of miRNAs in cancer. Oncogenic miRNAs include miR-155, miR-17-5p and miR-21, whereas miRNAs with tumor-suppressing activities include miR-15a, miR-16–1 and let-7 (Table 1). Both oncogenic-and tumor-suppressing miRNAs are collectively called oncomiRs because of their respective roles in cancer progression (Esquela-Kerscher and Slack, 2006). One key pathway in which various miRNAs play the role of both tumor suppressor and oncogene is that of apoptosis. Apoptosis in immune system maintenance Apoptosis is of key importance in maintaining tissue homeostasis, developmental tissue remodeling, the defense against microbial and viral infections and in preventing tumor formation. Apoptosis also plays essential roles in various processes in the immune system, such as lymphocyte development, homeostatic control of immune response and also in the elimination of virally infected or tumor cells. During lymphocyte development, autoreactive T and B lymphocytes are eliminated by apoptosis in the primary lymphoid organs, the thymus and bone marrow, respectively, in a process termed negative selection. This process is important for preventing autoimmunity. Apoptosis induction is initiated intracellularly upon various cellular insults mediated through the mitochondria (the intrinsic pathway) or through transmembrane receptors (death receptors) that initiate apoptosis at the cell surface (the extrinsic pathway). In the intrinsic

Regulation by miRNAs R Schickel et al

5965

apoptosis pathway, sentinel proteins sense changes in the endoplasmic reticulum (ER stress), the nucleus (DNA damage), or directly at the mitochondria, which are the central command center of the apoptotic process. Key regulators of mitochondrial apoptotic activities are members of the BCL-2 protein family. They can be roughly divided into pro- and anti-apoptotic members and are characterized by the presence of BCL-2 homology (BH) domains. The multidomain-containing members that function directly on the outer mitochondrial membrane include pro-apoptotic BAX and BAK (containing BH1-BH3) and anti-apoptotic BCL-2 and BCL-XL (containing BH1–BH4). Upstream of these proteins are a number of very diverse pro-apoptotic sensor proteins containing only the BH3 domain (e.g., BIM, BID, BAD and NOXA). These proteins are activated by various pro-apoptotic stimuli and all act by either activating BAX/BAK or inhibiting BCL-2/ BCL-XL (Coultas and Strasser, 2003). Among the BH3-only proteins, BIM, in particular, has been well recognized to play a critical role in the apoptosis of autoreactive T cells during negative selection in the thymus. Bim knockout mice accumulate three- to five-fold excess numbers of lymphoid and myeloid cells, and activated T cells from these mice are abnormally resistant to cytokine withdrawal-induced apoptosis (Bouillet et al., 1999). BIM has also been demonstrated to be essential for the deletion of autoreactive B lymphocytes in both bone marrow and periphery (Enders et al., 2003). Once activated, mitochondria release factors, including cytochrome c and SMAC/Diablo, that act by triggering the formation of the apoptosome (comprising cytochrome c, APAF-1, the initiator caspase 9 and dATP/ATP) and by inhibiting apoptosis inhibitors of the IAP family (Salvesen and Duckett, 2002), respectively. Both of these events contribute to the activation of the executioner caspases 3 and 7. In the extrinsic apoptosis pathway, surface receptors of the death receptor family that include TNFRI, CD95 (Fas/APO-1) and the TNF-related apoptosis inducing ligand (TRAIL) receptors DR4 and DR5 (Schulze-Osthoff et al., 1998), once triggered by their cognate ligands, recruit adaptor proteins (FADD and TRADD) to form a high-order receptor signaling structure that is collectively called the death-inducing signaling complex (DISC) (Peter and Krammer, 2003). Additional components of the DISC are the initiator caspases 8 and 10, and the caspase-8/10 regulator c-FLIP. Once activated, caspase-8 cleaves a number of substrates including the executioner caspase 3 and the BH3 protein BID (Barry et al., 2000; Waterhouse et al., 2005). The involvement of death receptors in negative selection has not been consistent and is somewhat controversial. TRAIL-deficient mice displayed increased numbers of immature thymocytes and accelerated autoimmune disease (Lamhamedi-Cherradi et al., 2003), but no defects in lymphoid homeostasis or in T-cell function (Sedger et al., 2002; Cretney et al., 2003). Although negative selection is intact in CD95/CD95L (Singer and Abbas, 1994; Adachi et al., 1996), TNFR1

(Pfeffer et al., 1993), TNFR2 (Erickson et al., 1994) or TNFR1 and TNFR2 double-deficient mice (Page et al., 1998), there has been evidence supporting roles of TNF and CD95 in negative selection in some models (Castro et al., 1996; Kishimoto et al., 1998). Death receptormediated apoptosis does not appear to play a critical role in thymic central tolerance; however, genetic deletion of members of the BCL-2 family, especially BIM in mice, has shown BIM to be a critical mediator of apoptosis for negative selection in the thymus (Bouillet et al., 2002; Strasser et al., 2008). Recently, studies have shown that BIM functions together with the death receptor CD95 in the apoptotic process during T-cell clonal contraction or peripheral deletion during acute and chronic infection (Hughes et al., 2008; Hutcheson et al., 2008; Weant et al., 2008). Both intrinsic and extrinsic apoptotic pathways converge on the level of caspase-3 activation, which then goes on to cleave various intracellular substrates that cause the typical changes observed in apoptotic cells. The ability to evade the grips of the apoptotic pathway has been described as a hallmark of cancer, and all neoplasms contain disruptions of the apoptotic machinery. As >30% of the human genome is now believed to be under the control of miRNAs, and considering the fundamental role of miRNAs in important differentiation and homeostatic processes, it became an intriguing possibility that a relationship existed between miRNAs and the apoptotic machinery. Apoptosis-regulating miRNAs (‘apoptomiRs’) The first miRNA described as an apoptotic regulator was the Drosophila gene bantam (Brennecke et al., 2003). Bantam was found to directly regulate the pro-apoptotic factor hid, suppressing the apoptotic activity and allowing proliferation to occur. Furthermore, miR-14 was found to suppress pro-apoptotic activity of reaper during the eye formation (Xu et al., 2003). Although spatially and temporally regulated as observed in other cell-fate-determining miRNAs, both bantam and miR14 are described to function as anti-apoptotic miRNAs. In the following, we will give examples of miRNAs that have been shown to regulate cell death in mammalian cells. Chromosome 13q14 is a region deleted in more than half of B-cell chronic lymphocytic leukemia (CLL). The identification of miR-15 and miR-16 as being the tumor suppressors at this chromosomal location represented the first link between miRNAs and cancer (Calin et al., 2002). Groundbreaking studies by Cimmino et al. (2005) looking into tumorigenic effects of the miR-15a/miR-16-1 deletion demonstrated that the expression of miR-15a and miR-16-1 inversely correlated with BCL-2 expression in CLL cells, providing the first description of direct miRNA mediation of apoptotic activity in human tissue. The 30 -UTR of BCL2 contains a target site for each of these miRNAs, and miR-15a/16-1 controls the expression of BCL-2. Post-translational regulation of BCL-2 by miR-15a and miR-16-1 opens the door to potential Oncogene

Regulation by miRNAs R Schickel et al

5966

therapeutic development of miRNAs in BCL-2 overexpressing tumors. Another link between miRNAs and CLL is the finding that the TCL1 oncogene, which is often found in aggressive CLL, is regulated by miR-29 and miR-181b (Pekarsky et al., 2006). In contrast to miR-15/miR-16, whose expression is often reduced in cancers, the miR-17-92 cluster is overexpressed in many cancers including B-cell lymphoma (Ota et al., 2004; Hayashita et al., 2005; He et al., 2005b). Recently, it was demonstrated that the inhibition of two miRNAs in this cluster, miR-17-5p and miR-20a, sensitized lung cancer cells to apoptotic insults (Matsubara et al., 2007), which suggests that these miRNAs may be useful as a potential therapeutic modality. miR-20a negatively regulates E2F1 (O’Donnell et al., 2005), E2F2 and E2F3 (Sylvestre et al., 2007), and all three E2Fs regulate the expression of miR-17-92 through binding to its promoter, suggesting an autoregulatory feedback loop. The E2F transcriptional networks are believed to link cell cycle progression to apoptosis, and perturbation of their various levels can induce either proliferation or apoptosis depending on the cellular context. miR-20a overexpression protects cancer cell lines from apoptosis, suggesting that overexpression of the miR-17-92 cluster promotes oncogenesis, in part, by perturbing the E2F network in a way that induces proliferation and inhibits apoptosis (Sylvestre et al., 2007). Recently, the identification of MCL-1 regulation by miR-29a,b and c expanded what is known about miRNA regulation within the BCL-2 family (Mott et al., 2007). MCL-1, a member of the BCL-2 family of anti-apoptotic proteins, is upregulated in malignant cells, whereas miR29b is downregulated in cancer. The mechanism of sensitization of cancer cells to TRAIL-induced apoptosis differs among cell types and is regulated by multiple downstream factors. Enforced expression of miR-29b renders tumor cells more sensitive to the apoptosisinducing activity of TRAIL, suggesting that the miR-29b/ MCL-1 connection is functionally important and could be exploited for cancer therapy. Recently, it was demonstrated that acquired resistance to TRAIL therapy involves c-Myc-dependent upregulation of both MCL-1 and cIAP2 (Ricci et al., 2007), suggesting that targeting MCL-1 using miR-29 could therefore be a viable treatment option in this situation. It seems clear that tight regulation of anti-apoptotic function through miRNAs is critical in development and other cellular processes. Interestingly, a database search using TargetScan (http://www.targetscan.org) for potential miRNA regulators of BCL-2 family members found a notable lack of conserved miRNAs that could regulate the pro-apoptotic Bcl-2 members (with the exception of BIM), but revealed that a majority of the anti-apoptotic BCL-2 members maintain conserved potential miRNA-binding sites (Table 2). Pro-survival members have on average six miRNA target sites in their 30 -UTRs as compared with 2.2 sites found in pro-apoptosis members. BCL-2, BCL-W and MCL-1 have multiple conserved miRNA target sites in their 30 -UTR. BCL-XL only has three such sites, and this Oncogene

could be due to the fact that the alternative splice form BCL-XS, which shares the same 30 -UTR with BCL-XL, has pro-apoptotic activities. This may indicate that evolutionary pressure favored the fine-tuning of the anti-apoptotic process over the pro-apoptotic process as part of the regulation of development and tissue homeostasis. Consistent with this hypothesis is an analysis of the 30 -UTRs of all caspases that revealed that most caspases are not predicted miRNA targets with the exception of caspase-3 (Table 2). It is interesting to note that caspase-3 has been shown to have nonapoptotic functions as well (Algeciras-Schimnich et al., 2002), which could be regulated by miRNAs. With the goal of identifying miRNAs that are relevant for proliferation or cell death, Cheng et al. (2005) individually knocked down 90 human miRNAs in HeLa and A549 cells. MiR-21 was identified among miRNAs that were identified as regulators of apoptosis. Subsequently, miR-21 was shown to be highly overexpressed in breast tumors compared with normal breast tissue, and the inhibition of miR-21 was shown to inhibit tumor growth (Si et al., 2007). Further work identified PTEN and tropomyosin 1 as direct targets of miR-21 in cancer cell lines (Zhu et al., 2007; Meng et al., 2007a). In glioblastoma, miR-21 was described as an anti-apoptotic factor (Chan et al., 2005), and a combination treatment of anti-miR-21 and TRAIL was able to eradicate apoptosis-resistant glioma cells in vitro and in vivo (Corsten et al., 2007). A recent study linked miR-21 to the pro-apoptotic tumor suppressor p53 and identified programmed cell death 4 (PDCD4) as a direct target (Frankel et al., 2008). The fundamental significance of miR-21 is most likely best documented by the fact that it is the only miRNA that was consistently upregulated in six out of six solid human cancers (breast, colon, lung, pancreas, prostate and stomach), when compared with matching noncancerous tissues (Volinia et al., 2006). Hence, this single miRNA provides a significant survival advantage to cancer cells upon its upregulation, and this may be a fundamental mechanism utilized by cancers to elude apoptotic cell death. Other apoptosis regulators targeted by miRNAs include HSP60 and HSP70 (targeted by miR-1) and caspase-9 (targeted by miR-133), and both of these miRNAs are predominantly expressed in cardiac and skeletal muscles (Xu et al., 2007). miR-1 and miR-133 have opposing affects on apoptosis, with miR-1 being pro-apoptotic and miR-133 being anti-apoptotic. Interestingly, miR-1 and miR-133 are transcribed together from the same chromosomal locus, suggesting that their relative levels might be regulated at a post-transcriptional level, and that it is the post-transcriptional regulation of these miRNAs that determines the eventual physiological outcome within cardiac and skeletal muscle tissues. A key regulator in the activation of the intrinsic apoptosis pathway in response to DNA damage, cellular stress and/or improper mitotic stimulation is p53. As a transcriptional regulator, p53 both activates and represses gene expression, and is the most frequently dysregulated protein in cancer with a majority of

Regulation by miRNAs R Schickel et al

5967 Table 2 Predicted miRNA target sites in Bcl-2 family members and caspases Gene

Size of 30 -UTR (kb)

Pro-survival Bcl-2 genes BCL-2 BCL-XL BCL-W

No. of conserved TargetScan sites

5.2 1.5 2.7

10 3 12

A1 MCL-1

0.2 2.8

0 11

BOO

0.2

0

Pro-apoptotic Bcl-2 genes Bax/Bak-like BAX 0.2 BOK 1.7 1.5 BCL-XS BAK 1.3

0 0 3 3

BFK BCL-GL BH3-only proteins BAD BIK BID HRK BIM NOXA PUMA BMF Caspases CASP1 CASP2 CASP3 CASP4 CASP5 CASP6 CASP7 CASP8 CASP9 CASP10 CASP11-14

4.3 0.7

15/16/195/424/497, 21, 30-5p, 34/449, 153, 181, 204/211, 205, 217, 365 let-7/98, 133, 142-3p 10, 15/16/195/424/497 (2x), 17-5p/20/93/106/519, 24, 29, 30-3p, 103/107, 122, 133, 140, 148/152 17-5p/20/93/106/519, 25/32/92/363/367, 26, 29, 125/351, 133, 142-5p, 153, 181, 193, 19/191-3p/294/295/302/372/37

let-7/98, 133, 142-3p miR-26, miR-29, miR-125/351

No information in TargetScan 0

0.4 0.4 1.4 0.3 4.2

0 0 0 0 19

1.5 1 3.9

1 0 4

0.1 2.6 1.6 0.1 0.1 0.7 1.4 1.1 0.6 3.9

Conserved sites for miRNA families

9, 10, 17-5p/20/93/106/519, 19, 24 (2x), 25/32/9,25/32/92/363/367, 30, 93/291-3p/294/295/ 302/372/373/520, 101, 124.1, 124.2/506 (2x), 130/301, 148/152 (2x), 181, 221/222 142-3p 29, 125/351 (2x), 221/222

0 No information in TargetScan 3 let-7/98, 30-5p/138 0 0 0 1 23 0 0 0 No information in TargetScan

Analyses were performed using TargetScan 4.2 (http://www.targetscan.org).

cancers harboring functional loss of p53. Five groups independently identified miR-34 as a target for p53 (Bommer et al., 2007; Chang et al., 2007; He et al., 2007; Raver-Shapira et al., 2007; Tarasov et al., 2007). He et al. (2007) described that miR-34 expression correlates with p53 expression. Using a p53-inducible system, they reported that the miR-34 family of miRNAs is directly regulated by p53, and that miR-34 mediates growth arrest in multiple cell lines via direct 30 -UTR regulation of cell cycle regulatory factors, such as cyclin-E2 (CCNE2), cyclin-dependent kinase 4 (CDK4) and the hepatocyte growth factor receptor (c-Met). Ectopic expression of miR-34 also resulted in a decrease of phospho-Rb, supporting the hypothesis that miR-34 regulates CDK4 and CCNE2. Furthermore, miR-34a was shown to directly target E2F3. In addition, miR-34a resulted in an increase in caspase-dependent death when introduced into two cell lines (Welch et al., 2007) and contributed to an increase in p53-mediated apoptosis

(Chang et al., 2007; Raver-Shapira et al., 2007). miR-34 was therefore described as a general sensitizer to apoptosis mainly through its link to p53, although its targets are still speculative. Interestingly, the other main activity of p53 to induce growth arrest, and cellular senescence also seems to involve miR-34a, as it was recently demonstrated that HCT116 colon cancer cells that were suppressed in their growth upon the introduction of miR-34a acquired a senescence-like phenotype (Tazawa et al., 2007). It remains to be determined how many of the activities of p53 depend on regulation by miR-34a. For instance, tumor protein p53-induced nuclear protein (TP53INP1) is a stress-induced p53 target gene that induces caspase-3-dependent apoptosis. TP53INP1 was shown to be dramatically reduced in its expression in pancreatic ductal adenocarcinoma (Gironella et al., 2007). TP53INP1 was demonstrated to be a direct target of miR-155, and consistently miR-155 is often Oncogene

Regulation by miRNAs R Schickel et al

5968

overexpressed in pancreatic cancer (Lee et al., 2007; Szafranska et al., 2007). Other miRNAs functionally connected to p53 are miR-372 and miR-373, which were identified as oncogenes in testicular germ cell tumors, and target the tumor-suppressor LATS2 and neutralize p53-mediated CDK inhibition (Voorhoeve et al., 2006). Taken together, this body of evidence demonstrates that multiple miRNA families target p53, which is universally known as one of the most important tumor suppressors with apoptosis-inducing activities. As with the processes of differentiation and cell fate determination, it has become quite clear that miRNAs play a fundamental role in regulating key cogs of the apoptotic machinery. OncomiRs not (yet) directly connected to apoptosis Transcripts of miR-155 and its host gene BIC accumulate in B-cell malignancies. Elevated miR-155 levels are found in diffuse large B-cell lymphoma (Eis et al., 2005), Hodgkin’s lymphoma (Kluiver et al., 2005) and certain Burkitt’s lymphomas (Kluiver et al., 2006), supporting its role in B-cell development and lymphomagenesis. Consistently, transgenic mice expressing mmu-miR-155 driven by the pre-B-cell-specific immunoglobulin transcriptional enhancer element (Em-mmu-miR-155) exhibit preleukemic pre-B-cell proliferation that result in B-cell malignancy (Costinean et al., 2006). miRNAs have also been directly linked to metastasis formation. miR-10b was one of a few miRNAs whose expression was found to be elevated in metastatic breast cancer (Ma et al., 2007). Exogenous miR-10b increased both in vitro and in vivo invasion of established breast cancer cells combined with increased proliferation, whereas inhibition of miR-10b led to a 10-fold reduction in invasion. The bHLH transcription factor TWIST, a known inducer of epithelial-to-mesenchymal transition (EMT) and driving force of breast metastases (Yang et al., 2004), induces expression of miR-10b, and this induction indirectly upregulates levels of the prometastatic gene RHOC because miR-10b directly targets HOXD10 (Ma et al., 2007). These studies are important because they were the first to establish a connection between a particular miRNA and metastasis formation of a human cancer. Additional miRNAs such as miR335 and miR-126 have since been identified as key metastasis suppressors in vivo (Tavazoie et al., 2008). Recently, the miR-200 family of miRNAs (which includes miR-200a, b, c, miR-141 and miR-429) was identified as both a marker for epithelial cells and a powerful master regulator of EMT. Its activity was found to be mainly mediated through targeting two E-box-binding E-cadherin repressors, ZEB1 and ZEB2 (Gregory et al., 2008; Park et al., 2008). Interestingly, ZEB1 has been shown to regulate T-cell differentiation, to repress IL-2 production and to regulate the expression of CD4 (Yasui et al., 1998; Brabletz et al., 1999; Postigo and Dean, 1999). It will be interesting to determine whether miR-200 also regulates T-cell development or activation. Oncogene

The case of let-7 Although much of the research on the role of miRNAs in apoptosis regulation focuses on direct relationships between a miRNA and its putative apoptosis-regulating target and because miRNAs can regulate global differentiation pathways, it is likely that activities exerted by miRNAs on apoptosis pathways may often be indirect. Cells with gains or losses of specific miRNAs may be altered in a way that changes entire genetic programs and results in new phenotypes that have different apoptotic properties. In the following, we will review such a differentiation process that is regulated by miRNAs and affects apoptosis sensitivity of cells. The receptor-mediated activation of apoptosis is well defined in the TNF receptor superfamily. CD95 receptor is the best-characterized member of the death receptors in this superfamily, and it induces apoptosis by two distinct mechanisms in Type I or Type II cells, the latter discriminated by the requirement of mitochondrial involvement. Interestingly, T-cell activation was shown to represent another case of a conversion from Type II to Type I (Scaffidi et al., 1999; Schmitz et al., 2003). Upon binding of its ligand (CD95L), CD95 forms the DISC. The characteristics of the Type I pathway are receptor internalization followed by the formation of large amount of DISC on endosomes and the activation of caspase-8 that directly cleaves the executioner caspase 3 (Scaffidi et al., 1998; Barnhart et al., 2003; Lee et al., 2006b). In contrast, the mitochondrial-dependent Type II pathway does not involve internalization of the receptor to the same extent, and only a small amount of active caspase-8 is generated, which is sufficient to cleave Bid causing it to translocate to the mitochondria where it induces the release of mitochondrial factors that amplify the apoptotic signal (Li et al., 1998; Luo et al., 1998). Another difference between Type I and Type II cells is their response to stimulation with CD95L and to Taxanes. Type II cells were found to be much more sensitive to apoptosis induction by soluble CD95L and growth inhibition by Taxanes than Type I cells (Algeciras-Schimnich et al., 2003). The difference in CD95 apoptosis sensitivity was used to classify the 60 cell lines of the drug-screening panel at the NCI (NCI60). Of 58 cell lines tested, 22 cell lines were found to be sensitive to CD95-mediated apoptosis—11 of them were classified as Type I and 11 as Type II (AlgecirasSchimnich et al., 2003). In a previous study, the NCI60 cells had been subjected to a gene chip analysis (Ross et al., 2000), revealing that the cell lines genetically fell into two major superclusters (SCs). SC1 cells were found to express genes that expressed a less differentiated and stromal phenotype, when compared with SC2 cells that expressed a gene signature consistent with a more differentiated phenotype. We recently identified the let-7 family as being preferentially expressed in Type II cells. Because let-7 is a marker for more differentiated tissues, this suggests that let-7 levels could directly affect the differentiation state of cancer cells and hence their sensitivity to death ligands and Taxanes.

Regulation by miRNAs R Schickel et al

5969

Let-7 is downregulated in a number of human cancers such as lung, colon, or ovarian cancer, and it serves as a prognostic marker for disease outcome (Takamizawa et al., 2004; Johnson et al., 2005; Akao et al., 2006; Yanaihara et al., 2006; Shell et al., 2007). It was also suggested to be a tumor suppressor for lung cancer through targeting RAS (Johnson et al., 2005). More recently, high-mobility group A2 (HMGA2) was identified as a major target for let-7 (Hebert et al., 2007; Lee and Dutta, 2007; Mayr et al., 2007; Shell et al., 2007), and indeed HMGA2, which is not expressed in most adult tissues, is upregulated in various cancers, such as neuroblastoma (Giannini et al., 2000), pancreatic cancer (Abe et al., 2003), thyroid neoplasms (Chiappetta et al., 1995), squamous carcinoma (Miyazawa et al., 2004) and lung cancer (Sarhadi et al., 2006). Interestingly, HMGA2 is an early embryonic gene. Consistently, let-7 is one of the main miRNAs that is upregulated late during embryonic development (Lagos-Quintana et al., 2001; Abbott et al., 2005; Schulman et al., 2005), and this is conserved from C. elegans to humans (Pasquinelli et al., 2000). The physiological targets for let-7 are therefore most likely embryonic genes. We have recently demonstrated that let-7 is selectively expressed by Type II human cancer cells that are at an early stage of dedifferentiation (Shell et al., 2007), and we proposed that let-7 controls a number of oncofetal genes to prevent the de-differentiation process of reverse embryogenesis that might give tumor cells properties of embryonic cells (Park et al., 2007). Recently, a set of 12 let-7-regulated oncofetal genes (LOGs) was identified using a genomewide bioinformatics approach (Boyerinas et al., 2008). Overexpression of let-7 inhibits cancer growth (Takamizawa et al., 2004; Akao et al., 2006; Johnson et al., 2007) and its inhibition promotes growth (Kumar et al., 2007). Recently, it was shown that let-7 effectively suppresses cancer development in a mouse model of spontaneous lung cancer (Esquela-Kerscher et al., 2008; Kumar et al., 2008), giving hope that it could be useful for cancer therapy. Another target for cancer therapy are the recently discovered regulators of let-7 processing LIN28 and LIN28B (which happens to be LOG3) (Newman et al., 2008; Viswanathan et al., 2008). The stem cell factor LIN28 (Yu et al., 2007) has been shown to selectively block the processing of most let-7 family members at the Drosha level and it is likely to be upregulated in many different cancers. So far, this has been shown for hepatocellular carcinoma in which LIN28B was found to be upregulated in more advanced cancer (Guo et al., 2006). Although let-7 is downregulated in many human tumors, it cannot be viewed as a classical tumor suppressor gene. First, it consists of 12 individual genes transcribed from 8 chromosomal loci (Park et al., 2007),

and although all 12 miRNAs are predicted to have a similar set of targets, it is not clear whether all 12 miRNAs have the same function. Secondly, miRNAs exert their function through targeting other genes, and it is therefore conceivable that, in certain tissues under certain conditions, let-7 could have a set of targets that have tumor-suppressing functions, whereas in other contexts it may not. In fact, let-7a-3 was described to be epigenetically silenced in a human lung cancer cell line and was suggested to have oncogenic function (Brueckner et al., 2007). In another study, it was shown that the pro-inflammatory cytokine IL-6 can contribute to tumor growth and drug resistance through the upregulation of let-7a (Meng et al., 2007b). Other examples of miRNAs with no clearly defined activity with respect to tumorigenesis are miR-21 and miR-24, which, depending on the cell type, can either promote or inhibit cell growth (Cheng et al., 2005). As such, a miRNA can only be considered a tumor suppressor in particular tissues where its expression inhibits the expression of genes considered to have oncogenic properties. It is therefore most appropriate to refer to miRNAs that are involved in cancer development in more general terms as oncomiRs. Concluding remarks It has become quite clear that microRNAs regulate a diverse set of cellular processes in both the immune system and solid tissues. From proper immune function, to hematopoietic lineage commitment, to embryonic development, to apoptosis and cell cycle regulation, many miRNAs act as master regulators that enforce particular patterns of gene expression by both directly and indirectly modulating the expression of target genes. The field of miRNA studies is no longer in its infancy, yet we are many years from developing a comprehensive understanding of the subject. In many cases, there is a direct relationship between a particular miRNA and expression of a target gene—miR15a and BCL2. However, what has become clear is the importance of context when attempting to determine the functional outcome of gain or loss of any particular miRNA. The relationship between a miRNA and a particular target may be more important in some situations than others— mir-21, for instance, can play the part of both tumor suppressor and oncogene depending on cellular context. As the current evidence suggests that many miRNAs play an essential role in differentiation and cell fate determination, we will most likely find that many miRNAs function similar to let-7, which seems to control a program of differentiation, and whose loss contributes to carcinogenesis by modulating both apoptotic and cell cycle pathways.

References Abbott AL, Alvarez-Saavedra E, Miska EA, Lau NC, Bartel DP, Horvitz HR et al. (2005). The let-7 MicroRNA family members

mir-48, mir-84, and mir-241 function together to regulate developmental timing in Caenorhabditis elegans. Dev Cell 9: 403–414. Oncogene

Regulation by miRNAs R Schickel et al

5970 Abe N, Watanabe T, Suzuki Y, Matsumoto N, Masaki T, Mori T et al. (2003). An increased high-mobility group A2 expression level is associated with malignant phenotype in pancreatic exocrine tissue. Br J Cancer 89: 2104–2109. Adachi M, Suematsu S, Suda T, Watanabe D, Fukuyama H, Ogasawara J et al. (1996). Enhanced and accelerated lymphoproliferation in Fas-null mice. Proc Natl Acad Sci USA 93: 2131–2136. Akao Y, Nakagawa Y, Kitade Y, Kinoshita T, Naoe T. (2007). Downregulation of microRNAs-143 and -145 in B-cell malignancies. Cancer Sci 98: 1914–1920. Akao Y, Nakagawa Y, Naoe T. (2006). let-7 MicroRNA functions as a potential growth suppressor in human colon cancer cells. Biol Pharm Bull 29: 903–906. Algeciras-Schimnich A, Barnhart BC, Peter ME. (2002). Apoptosisindependent functions of killer caspases. Curr Opin Cell Biol 14: 721–726. Algeciras-Schimnich A, Pietras EM, Barnhart BC, Legembre P, Vijayan S, Holbeck SL et al. (2003). Two CD95 tumor classes with different sensitivities to antitumor drugs. Proc Natl Acad Sci USA 100: 11445–11450. Asangani IA, Rasheed SA, Nikolova DA, Leupold JH, Colburn NH, Post S et al. (2008). MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 27: 2128–2136. Baehrecke EH. (2003). miRNAs: micro managers of programmed cell death. Curr Biol 13: R473–R475. Barnhart BC, Alappat EC, Peter ME. (2003). The CD95 type I/type II model. Semin Immunol 15: 185–193. Barry M, Heibein JA, Pinkoski MJ, Lee SF, Moyer RW, Green DR et al. (2000). Granzyme B short-circuits the need for caspase 8 activity during granule-mediated cytotoxic T-lymphocyte killing by directly cleaving Bid. Mol Cell Biol 20: 3781–3794. Bartel DP. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116: 281–297. Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ et al. (2003). Dicer is essential for mouse development. Nat Genet 35: 215–217. Bommer GT, Gerin I, Feng Y, Kaczorowski AJ, Kuick R, Love RE et al. (2007). p53-Mediated activation of miRNA34 candidate tumor-suppressor genes. Curr Biol 17: 1298–1307. Bouillet P, Metcalf D, Huang DC, Tarlinton DM, Kay TW, Kontgen F et al. (1999). Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science 286: 1735–1738. Bouillet P, Purton JF, Godfrey DI, Zhang LC, Coultas L, Puthalakath H et al. (2002). BH3-only Bcl-2 family member Bim is required for apoptosis of autoreactive thymocytes. Nature 415: 922–926. Boyerinas B, Park SM, Shomron N, Hedegaard MM, Vinther J, Andersen JA et al. (2008). Identification of Let-7-regulated oncofetal genes. Cancer Res 68: 2587–2591. Brabletz T, Jung A, Hlubek F, Lohberg C, Meiler J, Suchy U et al. (1999). Negative regulation of CD4 expression in T cells by the transcriptional repressor ZEB. Int Immunol 11: 1701–1708. Bredenkamp N, Seoighe C, Illing N. (2007). Comparative evolutionary analysis of the FoxG1 transcription factor from diverse vertebrates identifies conserved recognition sites for microRNA regulation. Dev Genes Evol 217: 227–233. Brennecke J, Hipfner DR, Stark A, Russell RB, Cohen SM. (2003). bantam encodes a developmentally regulated microRNA that controls cell proliferation and regulates the proapoptotic gene hid in Drosophila. Cell 113: 25–36. Brueckner B, Stresemann C, Kuner R, Mund C, Musch T, Meister M et al. (2007). The human let-7a-3 locus contains an epigenetically regulated microRNA gene with oncogenic function. Cancer Res 67: 1419–1423. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E et al. (2002). 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. Oncogene

Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, Wojcik SE et al. (2005). A microRNA signature associated with prognosis and progression in chronic lymphocytic leukemia. N Engl J Med 353: 1793–1801. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S et al. (2004). Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA 101: 2999–3004. Care A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P et al. (2007). MicroRNA-133 controls cardiac hypertrophy. Nat Med 13: 613–618. Castro JE, Listman JA, Jacobson BA, Wang Y, Lopez PA, Ju S et al. (1996). Fas modulation of apoptosis during negative selection of thymocytes. Immunity 5: 617–627. Chalfie M, Horvitz HR, Sulston JE. (1981). Mutations that lead to reiterations in the cell lineages of C.elegans. Cell 24: 59–69. Chan JA, Krichevsky AM, Kosik KS. (2005). MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res 65: 6029–6033. Chang J, Nicolas E, Marks D, Sander C, Lerro A, Buendia MA et al. (2004). miR-122, a mammalian liver-specific microRNA, is processed from hcr mRNA and may downregulate the high affinity cationic amino acid transporter CAT-1. RNA Biol 1: 106–113. Chang TC, Wentzel EA, Kent OA, Ramachandran K, Mullendore M, Lee KH et al. (2007). Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol Cell 26: 745–752. Chang TC, Yu D, Lee YS, Wentzel EA, Arking DE, West KM et al. (2008). Widespread microRNA repression by Myc contributes to tumorigenesis. Nat Genet 40: 43–50. Chen CZ, Li L, Lodish HF, Bartel DP. (2004). MicroRNAs modulate hematopoietic lineage differentiation. Science 303: 83–86. Chen JF, Mandel EM, Thomson JM, Wu Q, Callis TE, Hammond SM et al. (2006). The role of microRNA-1 and microRNA-133 in skeletal muscle proliferation and differentiation. Nat Genet 38: 228–233. Chen Y, Gorski DH. (2008). Regulation of angiogenesis through a microRNA (miR-130a) that downregulates antiangiogenic homeobox genes GAX and HOXA5. Blood 111: 1217–1226. Cheng AM, Byrom MW, Shelton J, Ford LP. (2005). Antisense inhibition of human miRNAs and indications for an involvement of miRNA in cell growth and apoptosis. Nucleic Acids Res 33: 1290–1297. Chiappetta G, Bandiera A, Berlingieri MT, Visconti R, Manfioletti G, Battista S et al. (1995). The expression of the high mobility group HMGI (Y) proteins correlates with the malignant phenotype of human thyroid neoplasias. Oncogene 10: 1307–1314. Ciafre SA, Galardi S, Mangiola A, Ferracin M, Liu CG, Sabatino G et al. (2005). Extensive modulation of a set of microRNAs in primary glioblastoma. Biochem Biophys Res Commun 334: 1351–1358. Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M et al. (2005). miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA 102: 13944–13949. Corney DC, Flesken-Nikitin A, Godwin AK, Wang W, Nikitin AY. (2007). MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell proliferation and adhesion-independent growth. Cancer Res 67: 8433–8438. Corsten MF, Miranda R, Kasmieh R, Krichevsky AM, Weissleder R, Shah K. (2007). MicroRNA-21 knockdown disrupts glioma growth in vivo and displays synergistic cytotoxicity with neural precursor cell delivered S-TRAIL in human gliomas. Cancer Res 67: 8994–9000. Costinean S, Zanesi N, Pekarsky Y, Tili E, Volinia S, Heerema N et al. (2006). Pre-B cell proliferation and lymphoblastic leukemia/highgrade lymphoma in E(mu)-miR155 transgenic mice. Proc Natl Acad Sci USA 103: 7024–7029. Coultas L, Strasser A. (2003). The role of the Bcl-2 protein family in cancer. Semin Cancer Biol 13: 115–123.

Regulation by miRNAs R Schickel et al

5971 Cretney E, Uldrich AP, Berzins SP, Strasser A, Godfrey DI, Smyth MJ. (2003). Normal thymocyte negative selection in TRAIL-deficient mice. J Exp Med 198: 491–496. Cui Q, Yu Z, Purisima EO, Wang E. (2006). Principles of microRNA regulation of a human cellular signaling network. Mol Syst Biol 2: 46. Cullen BR. (2004). Transcription and processing of human microRNA precursors. Mol Cell 16: 861–865. Dews M, Homayouni A, Yu D, Murphy D, Sevignani C, Wentzel E et al. (2006). Augmentation of tumor angiogenesis by a Mycactivated microRNA cluster. Nat Genet 38: 1060–1065. Eis PS, Tam W, Sun L, Chadburn A, Li Z, Gomez MF et al. (2005). Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proc Natl Acad Sci USA 102: 3627–3632. Enders A, Bouillet P, Puthalakath H, Xu Y, Tarlinton DM, Strasser A. (2003). Loss of the pro-apoptotic BH3-only Bcl-2 family member Bim inhibits BCR stimulation-induced apoptosis and deletion of autoreactive B cells. J Exp Med 198: 1119–1126. Engels BM, Hutvagner G. (2006). Principles and effects of microRNAmediated post-transcriptional gene regulation. Oncogene 25: 6163–6169. Erickson SL, de Sauvage FJ, Kikly K, Carver-Moore K, Pitts-Meek S, Gillett N et al. (1994). Decreased sensitivity to tumour-necrosis factor but normal T-cell development in TNF receptor-2-deficient mice. Nature 372: 560–563. Esau C, Davis S, Murray SF, Yu XX, Pandey SK, Pear M et al. (2006). miR-122 Regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab 3: 87–98. Esau C, Kang X, Peralta E, Hanson E, Marcusson EG, Ravichandran LV et al. (2004). MicroRNA-143 regulates adipocyte differentiation. J Biol Chem 279: 52361–52365. Esquela-Kerscher A, Slack FJ. (2006). Oncomirs—microRNAs with a role in cancer. Nat Rev Cancer 6: 259–269. Esquela-Kerscher A, Trang P, Wiggins JF, Patrawala L, Cheng A, Ford L et al. (2008). The let-7 microRNA reduces tumor growth in mouse models of lung cancer. Cell Cycle 7: 759–764. Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E et al. (2007). MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci USA 104: 15805–15810. Fazi F, Rosa A, Fatica A, Gelmetti V, De Marchis ML, Nervi C et al. (2005). A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPalpha regulates human granulopoiesis. Cell 123: 819–831. Felli N, Fontana L, Pelosi E, Botta R, Bonci D, Facchiano F et al. (2005). MicroRNAs 221 and 222 inhibit normal erythropoiesis and erythroleukemic cell growth via kit receptor down-modulation. Proc Natl Acad Sci USA 102: 18081–18086. Fontana L, Pelosi E, Greco P, Racanicchi S, Testa U, Liuzzi F et al. (2007). MicroRNAs 17-5p-20a-106a control monocytopoiesis through AML1 targeting and M-CSF receptor upregulation. Nat Cell Biol 9: 775–787. Frankel LB, Christoffersen NR, Jacobsen A, Lindow M, Krogh A, Lund AH. (2008). Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J Biol Chem 283: 1026–1033. Fukao T, Fukuda Y, Kiga K, Sharif J, Hino K, Enomoto Y et al. (2007). An evolutionarily conserved mechanism for microRNA-223 expression revealed by microRNA gene profiling. Cell 129: 617–631. Galardi S, Mercatelli N, Giorda E, Massalini S, Frajese GV, Ciafre SA et al. (2007). miR-221 and miR-222 expression affects the proliferation potential of human prostate carcinoma cell lines by targeting p27Kip1. J Biol Chem 282: 23716–23724. Garzon R, Pichiorri F, Palumbo T, Iuliano R, Cimmino A, Aqeilan R et al. (2006). MicroRNA fingerprints during human megakaryocytopoiesis. Proc Natl Acad Sci USA 103: 5078–5083. Gauwerky CE, Huebner K, Isobe M, Nowell PC, Croce CM. (1989). Activation of MYC in a masked t(8;17) translocation results in an aggressive B-cell leukemia. Proc Natl Acad Sci USA 86: 8867–8871.

Georgantas III RW, Hildreth R, Morisot S, Alder J, Liu CG, Heimfeld S et al. (2007). CD34+ hematopoietic stem-progenitor cell microRNA expression and function: a circuit diagram of differentiation control. Proc Natl Acad Sci USA 104: 2750–2755. Giannini G, Kim CJ, Di Marcotullio L, Manfioletti G, Cardinali B, Cerignoli F et al. (2000). Expression of the HMGI(Y) gene products in human neuroblastic tumours correlates with differentiation status. Br J Cancer 83: 1503–1509. Gillies JK, Lorimer IA. (2007). Regulation of p27Kip1 by miRNA 221/222 in glioblastoma. Cell Cycle 6: 2005–2009. Gironella M, Seux M, Xie MJ, Cano C, Tomasini R, Gommeaux J et al. (2007). Tumor protein 53-induced nuclear protein 1 expression is repressed by miR-155, and its restoration inhibits pancreatic tumor development. Proc Natl Acad Sci USA 104: 16170–16175. Gottardo F, Liu CG, Ferracin M, Calin GA, Fassan M, Bassi P et al. (2007). Micro-RNA profiling in kidney and bladder cancers. Urol Oncol 25: 387–392. Gramantieri L, Ferracin M, Fornari F, Veronese A, Sabbioni S, Liu CG et al. (2007). Cyclin G1 is a target of miR-122a, a microRNA frequently down-regulated in human hepatocellular carcinoma. Cancer Res 67: 6092–6099. Grandori C, Cowley SM, James LP, Eisenman RN. (2000). The Myc/ Max/Mad network and the transcriptional control of cell behavior. Annu Rev Cell Dev Biol 16: 653–699. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshild G et al. (2008). The microRNA-200 family and miR-205 regulate epithelial–mesenchymal transition by targeting the E-cadherin repressors, ZEB1 and SIP1. Nature Cell Biol 10: 593–601. Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP. (2007). MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 27: 91–105. Guo Y, Chen Y, Ito H, Watanabe A, Ge X, Kodama T et al. (2006). Identification and characterization of lin-28 homolog B (LIN28B) in human hepatocellular carcinoma. Gene 384: 51–61. Harris TA, Yamakuchi M, Ferlito M, Mendell JT, Lowenstein CJ. (2008). MicroRNA-126 regulates endothelial expression of vascular cell adhesion molecule 1. Proc Natl Acad Sci USA 105: 1516–1521. Hayashita Y, Osada H, Tatematsu Y, Yamada H, Yanagisawa K, Tomida S et al. (2005). A polycistronic microRNA cluster, miR-1792, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Res 65: 9628–9632. He H, Jazdzewski K, Li W, Liyanarachchi S, Nagy R, Volinia S et al. (2005a). The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci USA 102: 19075–19080. He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y et al. (2007). A microRNA component of the p53 tumour suppressor network. Nature 447: 1130–1134. He L, Thomson JM, Hemann MT, Hernando-Monge E, Mu D, Goodson S et al. (2005b). A microRNA polycistron as a potential human oncogene. Nature 435: 828–833. Hebert C, Norris K, Scheper MA, Nikitakis N, Sauk JJ. (2007). High mobility group A2 is a target for miRNA-98 in head and neck squamous cell carcinoma. Mol Cancer 6: 5. Hornstein E, Mansfield JH, Yekta S, Hu JK, Harfe BD, McManus MT et al. (2005). The microRNA miR-196 acts upstream of Hoxb8 and Shh in limb development. Nature 438: 671–674. Hossain A, Kuo MT, Saunders GF. (2006). Mir-17-5p regulates breast cancer cell proliferation by inhibiting translation of AIB1 mRNA. Mol Cell Biol 26: 8191–8201. Hughes PD, Belz GT, Fortner KA, Budd RC, Strasser A, Bouillet P. (2008). Apoptosis regulators Fas and Bim cooperate in shutdown of chronic immune responses and prevention of autoimmunity. Immunity 28: 197–205. Hutcheson J, Scatizzi JC, Siddiqui AM, Haines III GK, Wu T, Li QZ et al. (2008). Combined deficiency of proapoptotic regulators Bim and Fas results in the early onset of systemic autoimmunity. Immunity 28: 206–217. Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S et al. (2005). MicroRNA gene expression deregulation in human breast cancer. Cancer Res 65: 7065–7070. Oncogene

Regulation by miRNAs R Schickel et al

5972 Janz S. (2006). Myc translocations in B cell and plasma cell neoplasms. DNA Repair (Amst) 5: 1213–1224. Joglekar MV, Parekh VS, Mehta S, Bhonde RR, Hardikar AA. (2007). MicroRNA profiling of developing and regenerating pancreas reveal post-transcriptional regulation of neurogenin3. Dev Biol 311: 603–612. Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko D et al. (2007). The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res 67: 7713–7722. Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A et al. (2005). RAS is regulated by the let-7 microRNA family. Cell 120: 635–647. Johnston RJ, Hobert O. (2003). A microRNA controlling left/right neuronal asymmetry in Caenorhabditis elegans. Nature 426: 845–849. Karin M, Lawrence T, Nizet V. (2006). Innate immunity gone awry: linking microbial infections to chronic inflammation and cancer. Cell 124: 823–835. Kawasaki H, Taira K. (2004). MicroRNA-196 inhibits HOXB8 expression in myeloid differentiation of HL60 cells. Nucleic Acids Symp Ser (Oxf) 48: 211–212. Kim VN. (2005). MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol 6: 376–385. Kishimoto H, Surh CD, Sprent J. (1998). A role for Fas in negative selection of thymocytes in vivo. J Exp Med 187: 1427–1438. Kluiver J, Haralambieva E, de Jong D, Blokzijl T, Jacobs S, Kroesen BJ et al. (2006). Lack of BIC and microRNA miR-155 expression in primary cases of Burkitt lymphoma. Genes Chromosomes Cancer 45: 147–153. Kluiver J, Poppema S, de Jong D, Blokzijl T, Harms G, Jacobs S et al. (2005). BIC and miR-155 are highly expressed in Hodgkin, primary mediastinal and diffuse large B cell lymphomas. J Pathol 207: 243–249. Kumar MS, Erkeland SJ, Pester RE, Chen CY, Ebert MS, Sharp PA et al. (2008). Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proc Natl Acad Sci USA 105: 3903–3908. Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. (2007). Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat Genet 39: 673–677. Kutay H, Bai S, Datta J, Motiwala T, Pogribny I, Frankel W et al. (2006). Downregulation of miR-122 in the rodent and human hepatocellular carcinomas. J Cell Biochem 99: 671–678. Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. (2001). Identification of novel genes coding for small expressed RNAs. Science 294: 853–858. Lai EC. (2002). Micro RNAs are complementary to 30 UTR sequence motifs that mediate negative post-transcriptional regulation. Nat Genet 30: 363–364. Lamhamedi-Cherradi SE, Zheng SJ, Maguschak KA, Peschon J, Chen YH. (2003). Defective thymocyte apoptosis and accelerated autoimmune diseases in TRAIL/ mice. Nat Immunol 4: 255–260. Landais S, Landry S, Legault P, Rassart E. (2007). Oncogenic potential of the miR-106-363 cluster and its implication in human T-cell leukemia. Cancer Res 67: 5699–5707. Lawrie CH, Soneji S, Marafioti T, Cooper CD, Palazzo S, Paterson JC et al. (2007). MicroRNA expression distinguishes between germinal center B cell-like and activated B cell-like subtypes of diffuse large B cell lymphoma. Int J Cancer 121: 1156–1161. le Sage C, Nagel R, Egan DA, Schrier M, Mesman E, Mangiola A et al. (2007). Regulation of the p27(Kip1) tumor suppressor by miR-221 and miR-222 promotes cancer cell proliferation. EMBO J 26: 3699–3708. Lee EJ, Gusev Y, Jiang J, Nuovo GJ, Lerner MR, Frankel WL et al. (2007). Expression profiling identifies microRNA signature in pancreatic cancer. Int J Cancer 120: 1046–1054. Lee HJ, Palkovits M, Young III WS. (2006a). miR-7b, a microRNA up-regulated in the hypothalamus after chronic hyperosmolar stimulation, inhibits Fos translation. Proc Natl Acad Sci USA 103: 15669–15674. Oncogene

Lee KH, Feig C, Tchikov V, Schickel R, Hallas C, Schutze S et al. (2006b). The role of receptor internalization in CD95 signaling. EMBO J 25: 1009–1023. Lee RC, Feinbaum RL, Ambros V. (1993). The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75: 843–854. Lee YS, Dutta A. (2007). The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes Dev 21: 1025–1030. Lehmann U, Hasemeier B, Christgen M, Muller M, Romermann D, Langer F et al. (2008). Epigenetic inactivation of microRNA gene hsa-mir-9-1 in human breast cancer. J Pathol 214: 17–24. Lewis BP, Burge CB, Bartel DP. (2005). Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120: 15–20. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB. (2003). Prediction of mammalian microRNA targets. Cell 115: 787–798. Li H, Zhu H, Xu CJ, Yuan J. (1998). Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 94: 491–501. Li QJ, Chau J, Ebert PJ, Sylvester G, Min H, Liu G et al. (2007). miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell 129: 147–161. Linsley PS, Schelter J, Burchard J, Kibukawa M, Martin MM, Bartz SR et al. (2007). Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol Cell Biol 27: 2240–2252. Liu CG, Calin GA, Meloon B, Gamliel N, Sevignani C, Ferracin M et al. (2004). An oligonucleotide microchip for genome-wide microRNA profiling in human and mouse tissues. Proc Natl Acad Sci USA 101: 9740–9744. Loffler D, Brocke-Heidrich K, Pfeifer G, Stocsits C, Hackermuller J, Kretzschmar AK et al. (2007). Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood 110: 1330–1333. Lu J, Getz G, Miska EA, varez-Saavedra E, Lamb J, Peck D et al. (2005). MicroRNA expression profiles classify human cancers. Nature 435: 834–838. Lu L, Katsaros D, de la Longrais IA, Sochirca O, Yu H. (2007a). Hypermethylation of let-7a-3 in epithelial ovarian cancer is associated with low insulin-like growth factor-II expression and favorable prognosis. Cancer Res 67: 10117–10122. Lu Y, Thomson JM, Wong HY, Hammond SM, Hogan BL. (2007b). Transgenic over-expression of the microRNA miR-17-92 cluster promotes proliferation and inhibits differentiation of lung epithelial progenitor cells. Dev Biol 310: 442–453. Lui WO, Pourmand N, Patterson BK, Fire A. (2007). Patterns of known and novel small RNAs in human cervical cancer. Cancer Res 67: 6031–6043. Lukiw WJ, Pogue AI. (2007). Induction of specific micro RNA (miRNA) species by ROS-generating metal sulfates in primary human brain cells. J Inorg Biochem 101: 1265–1269. Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. (1998). Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 94: 481–490. Ma L, Teruya-Feldstein J, Weinberg RA. (2007). Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature 449: 682–688. Marton S, Garcia MR, Robello C, Persson H, Trajtenberg F, Pritsch O et al. (2008). Small RNAs analysis in CLL reveals a deregulation of miRNA expression and novel miRNA candidates of putative relevance in CLL pathogenesis. Leukemia 22: 330–338. Masaki S, Ohtsuka R, Abe Y, Muta K, Umemura T. (2007). Expression patterns of microRNAs 155 and 451 during normal human erythropoiesis. Biochem Biophys Res Commun 364: 509–514. Matsubara H, Takeuchi T, Nishikawa E, Yanagisawa K, Hayashita Y, Ebi H et al. (2007). Apoptosis induction by antisense oligonucleotides against miR-17-5p and miR-20a in lung cancers overexpressing miR-17-92. Oncogene 26: 6099–6105.

Regulation by miRNAs R Schickel et al

5973 Mayr C, Hemann MT, Bartel DP. (2007). Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 315: 1576–1579. Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST, Patel T. (2007a). MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 133: 647–658. Meng F, Henson R, Wehbe-Janek H, Smith H, Ueno Y, Patel T. (2007b). The microRNA let-7a modulates interleukin-6-dependent STAT-3 survival signaling in malignant human cholangiocytes. J Biol Chem 282: 8256–8264. Metzler M, Wilda M, Busch K, Viehmann S, Borkhardt A. (2004). High expression of precursor microRNA-155/BIC RNA in children with Burkitt lymphoma. Genes Chromosomes Cancer 39: 167–169. Michael MZ, SM OC, van Holst Pellekaan NG, Young GP, James RJ. (2003). Reduced accumulation of specific microRNAs in colorectal neoplasia. Mol Cancer Res 1: 882–891. Miyazawa J, Mitoro A, Kawashiri S, Chada KK, Imai K. (2004). Expression of mesenchyme-specific gene HMGA2 in squamous cell carcinomas of the oral cavity. Cancer Res 64: 2024–2029. Mott JL, Kobayashi S, Bronk SF, Gores GJ. (2007). mir-29 Regulates Mcl-1 protein expression and apoptosis. Oncogene 26: 6133–6140. Naguibneva I, Ameyar-Zazoua M, Polesskaya A, Ait-Si-Ali S, Groisman R, Souidi M et al. (2006). The microRNA miR-181 targets the homeobox protein Hox-A11 during mammalian myoblast differentiation. Nat Cell Biol 8: 278–284. Newman MA, Thomson JM, Hammond S. (2008). Lin-28 interaction with the Let-7 precursor loop mediates regulated microRNA processing. RNA; 19 January 2008, e-pub ahead of print. Nielsen CB, Shomron N, Sandberg R, Hornstein E, Kitzman J, Burge CB. (2007). Determinants of targeting by endogenous and exogenous microRNA and siRNAs. RNA 13: 1894–1910. O’Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D. (2007). MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci USA 104: 1604–1609. O’Donnell KA, Wentzel EA, Zeller KI, Dang CV, Mendell JT. (2005). c-Myc-regulated microRNAs modulate E2F1 expression. Nature 435: 839–843. O’Rourke JR, Georges SA, Seay HR, Tapscott SJ, McManus MT, Goldhamer DJ et al. (2007). Essential role for Dicer during skeletal muscle development. Dev Biol 311: 359–368. Ota A, Tagawa H, Karnan S, Tsuzuki S, Karpas A, Kira S et al. (2004). Identification and characterization of a novel gene, C13orf25, as a target for 13q31–q32 amplification in malignant lymphoma. Cancer Res 64: 3087–3095. Page DM, Roberts EM, Peschon JJ, Hedrick SM. (1998). TNF receptor-deficient mice reveal striking differences between several models of thymocyte negative selection. J Immunol 160: 120–133. Park SM, Gaur AB, Lengyel E, Peter ME. (2008). The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors, ZEB1 and ZEB2. Genes Dev 22: 894–907. Park SM, Shell S, Radjabi AR, Schickel R, Feig C, Boyerinas B et al. (2007). Let-7 prevents early cancer progression by suppressing expression of the embryonic gene HMGA2. Cell Cycle 6: 2585–2590. Pasquinelli AE, Reinhart BJ, Slack F, Martindale MQ, Kuroda MI, Maller B et al. (2000). Conservation of the sequence and temporal expression of let-7 heterochronic regulatory RNA. Nature 408: 86–89. Pekarsky Y, Santanam U, Cimmino A, Palamarchuk A, Efanov A, Maximov V et al. (2006). Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res 66: 11590–11593. Peter ME, Krammer PH. (2003). The CD95(APO-1/Fas) DISC and beyond. Cell Death Differ 10: 26–35. Pfeffer K, Matsuyama T, Kundig TM, Wakeham A, Kishihara K, Shahinian A et al. (1993). Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection. Cell 73: 457–467.

Plaisance V, Abderrahmani A, Perret-Menoud V, Jacquemin P, Lemaigre F, Regazzi R. (2006). MicroRNA-9 controls the expression of Granuphilin/Slp4 and the secretory response of insulinproducing cells. J Biol Chem 281: 26932–26942. Postigo AA, Dean DC. (1999). Independent repressor domains in ZEB regulate muscle and T-cell differentiation. Mol Cell Biol 19: 7961–7971. Ramkissoon SH, Mainwaring LA, Ogasawara Y, Keyvanfar K, McCoy Jr JP, Sloand EM et al. (2006). Hematopoietic-specific microRNA expression in human cells. Leuk Res 30: 643–647. Raver-Shapira N, Marciano E, Meiri E, Spector Y, Rosenfeld N, Moskovits N et al. (2007). Transcriptional activation of miR-34a contributes to p53-mediated apoptosis. Mol Cell 26: 731–743. Reinhart BJ, Slack FJ, Basson M, Pasquinelli AE, Bettinger JC, Rougvie AE et al. (2000). The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans. Nature 403: 901–906. Ricci MS, Kim SH, Ogi K, Plastaras JP, Ling J, Wang W et al. (2007). Reduction of TRAIL-induced Mcl-1 and cIAP2 by c-Myc or sorafenib sensitizes resistant human cancer cells to TRAIL-induced death. Cancer Cell 12: 66–80. Rodriguez A, Vigorito E, Clare S, Warren MV, Couttet P, Soond DR et al. (2007). Requirement of bic/microRNA-155 for normal immune function. Science 316: 608–611. Roldo C, Missiaglia E, Hagan JP, Falconi M, Capelli P, Bersani S et al. (2006). MicroRNA expression abnormalities in pancreatic endocrine and acinar tumors are associated with distinctive pathologic features and clinical behavior. J Clin Oncol 24: 4677–4684. Ross DT, Scherf U, Eisen MB, Perou CM, Rees C, Spellman P et al. (2000). Systematic variation in gene expression patterns in human cancer cell lines. Nat Genet 24: 227–235. Salvesen GS, Duckett CS. (2002). IAP proteins: blocking the road to death’s door. Nat Rev Mol Cell Biol 3: 401–410. Sampson VB, Rong NH, Han J, Yang Q, Aris V, Soteropoulos P et al. (2007). MicroRNA let-7a down-regulates MYC and reverts MYC-induced growth in Burkitt lymphoma cells. Cancer Res 67: 9762–9770. Sarhadi VK, Wikman H, Salmenkivi K, Kuosma E, Sioris T, Salo J et al. (2006). Increased expression of high mobility group A proteins in lung cancer. J Pathol 209: 206–212. Sathyan P, Golden HB, Miranda RC. (2007). Competing interactions between micro-RNAs determine neural progenitor survival and proliferation after ethanol exposure: evidence from an ex vivo model of the fetal cerebral cortical neuroepithelium. J Neurosci 27: 8546–8557. Scaffidi C, Fulda S, Srinivasan A, Friesen C, Li F, Tomaselli KJ et al. (1998). Two CD95 (APO-1/Fas) signaling pathways. EMBO J 17: 1675–1687. Scaffidi C, Schmitz I, Krammer PH, Peter ME. (1999). The role of c-FLIP in modulation of CD95-induced apoptosis. J Biol Chem 274: 1541–1548. Schmitz I, Krueger A, Baumann S, Schulze-Bergkamen H, Krammer PH, Kirchhoff S. (2003). An IL-2-dependent switch between CD95 signaling pathways sensitizes primary human T cells toward CD95mediated activation-induced cell death. J Immunol 171: 2930–2936. Schulman BR, Esquela-Kerscher A, Slack FJ. (2005). Reciprocal expression of lin-41 and the microRNAs let-7 and mir-125 during mouse embryogenesis. Dev Dyn 234: 1046–1054. Schulze-Osthoff K, Ferrari D, Los M, Wesselborg S, Peter ME. (1998). Apoptosis signaling by death receptors. Eur J Biochem 254: 439–459. Sedger LM, Glaccum MB, Schuh JC, Kanaly ST, Williamson E, Kayagaki N et al. (2002). Characterization of the in vivo function of TNF-alpha-related apoptosis-inducing ligand, TRAIL/Apo2L, using TRAIL/Apo2L gene-deficient mice. Eur J Immunol 32: 2246–2254. Sethupathy P, Borel C, Gagnebin M, Grant GR, Deutsch S, Elton TS et al. (2007). Human microRNA-155 on chromosome 21 differentially interacts with its polymorphic target in the AGTR1 Oncogene

Regulation by miRNAs R Schickel et al

5974 30 untranslated region: a mechanism for functional single-nucleotide polymorphisms related to phenotypes. Am J Hum Genet 81: 405–413. Sevinsky JR, Whalen AM, Ahn NG. (2004). Extracellular signalregulated kinase induces the megakaryocyte GPIIb/CD41 gene through MafB/Kreisler. Mol Cell Biol 24: 4534–4545. Shell S, Park SM, Radjabi AR, Schickel R, Kistner EO, Jewell DA et al. (2007). Let-7 expression defines two differentiation stages of cancer. Proc Natl Acad Sci USA 104: 11400–11405. Si ML, Zhu S, Wu H, Lu Z, Wu F, Mo YY. (2007). miR-21-Mediated tumor growth. Oncogene 26: 2799–2803. Singer GG, Abbas AK. (1994). The fas antigen is involved in peripheral but not thymic deletion of T lymphocytes in T cell receptor transgenic mice. Immunity 1: 365–371. Strasser A, Puthalakath H, O’Reilly LA, Bouillet P. (2008). What do we know about the mechanisms of elimination of autoreactive T and B cells and what challenges remain. Immunol Cell Biol 86: 57–66. Sylvestre Y, De Guire V, Querido E, Mukhopadhyay UK, Bourdeau V, Major F et al. (2007). An E2F/miR-20a autoregulatory feedback loop. J Biol Chem 282: 2135–2143. Szafranska AE, Davison TS, John J, Cannon T, Sipos B, Maghnouj A et al. (2007). MicroRNA expression alterations are linked to tumorigenesis and non-neoplastic processes in pancreatic ductal adenocarcinoma. Oncogene 26: 4442–4452. Taganov KD, Boldin MP, Chang KJ, Baltimore D. (2006). NFkappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc Natl Acad Sci USA 103: 12481–12486. Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H et al. (2004). Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Res 64: 3753–3756. Tarasov V, Jung P, Verdoodt B, Lodygin D, Epanchintsev A, Menssen A et al. (2007). Differential regulation of microRNAs by p53 revealed by massively parallel sequencing: miR-34a is a p53 target that induces apoptosis and G1-arrest. Cell Cycle 6: 1586–1593. Tavazoie SF, Alarcon C, Oskarsson T, Padua D, Wang Q, Bos PD et al. (2008). Endogenous human microRNAs that suppress breast cancer metastasis. Nature 451: 147–152. Tazawa H, Tsuchiya N, Izumiya M, Nakagama H. (2007). Tumorsuppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc Natl Acad Sci USA 104: 15472–15477. Thai TH, Calado DP, Casola S, Ansel KM, Xiao C, Xue Y et al. (2007). Regulation of the germinal center response by microRNA155. Science 316: 604–608. Tili E, Michaille JJ, Cimino A, Costinean S, Dumitru CD, Adair B et al. (2007). Modulation of miR-155 and miR-125b levels following lipopolysaccharide/TNF-alpha stimulation and their possible roles in regulating the response to endotoxin shock. J Immunol 179: 5082–5089. van Rooij E, Sutherland LB, Liu N, Williams AH, McAnally J, Gerard RD et al. (2006). A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure. Proc Natl Acad Sci USA 103: 18255–18260. Venturini L, Battmer K, Castoldi M, Schultheis B, Hochhaus A, Muckenthaler MU et al. (2007). Expression of the miR-17-92 polycistron in chronic myeloid leukemia (CML) CD34+ cells. Blood 109: 4399–4405. Versteeg R, Caron H, Cheng NC, van der Drift P, Slater R, Westerveld A et al. (1995). 1p36: every subband a suppressor? Eur J Cancer 31A: 538–541. Viswanathan SR, Daley GQ, Gregory RI. (2008). Selective blockade of microRNA processing by Lin-28. Science 320: 97–100. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F et al. (2006). A microRNA expression signature of human solid tumors

Oncogene

defines cancer gene targets. Proc Natl Acad Sci USA 103: 2257–2261. Voorhoeve PM, le Sage C, Schrier M, Gillis AJ, Stoop H, Nagel R et al. (2006). A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell 124: 1169–1181. Voorhoeve PM, le Sage C, Schrier M, Gillis AJ, Stoop H, Nagel R et al. (2007). A genetic screen implicates miRNA-372 and miRNA373 as oncogenes in testicular germ cell tumors. Adv Exp Med Biol 604: 17–46. Waterhouse NJ, Sedelies KA, Browne KA, Wowk ME, Newbold A, Sutton VR et al. (2005). A central role for Bid in granzyme B-induced apoptosis. J Biol Chem 280: 4476–4482. Weant AE, Michalek RD, Khan IU, Holbrook BC, Willingham MC, Grayson JM. (2008). Apoptosis regulators Bim and Fas function concurrently to control autoimmunity and CD8+ T cell contraction. Immunity 28: 218–230. Welch C, Chen Y, Stallings RL. (2007). MicroRNA-34a functions as a potential tumor suppressor by inducing apoptosis in neuroblastoma cells. Oncogene 26: 5017–5022. Wightman B, Ha I, Ruvkun G. (1993). Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 75: 855–862. Xiao C, Calado DP, Galler G, Thai TH, Patterson HC, Wang J et al. (2007). MiR-150 controls B cell differentiation by targeting the transcription factor c-Myb. Cell 131: 146–159. Xu C, Lu Y, Pan Z, Chu W, Luo X, Lin H et al. (2007). The musclespecific microRNAs miR-1 and miR-133 produce opposing effects on apoptosis by targeting HSP60, HSP70 and caspase-9 in cardiomyocytes. J Cell Sci 120: 3045–3052. Xu P, Vernooy SY, Guo M, Hay BA. (2003). The Drosophila microRNA Mir-14 suppresses cell death and is required for normal fat metabolism. Curr Biol 13: 790–795. Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M et al. (2006). Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 9: 189–198. Yang B, Lin H, Xiao J, Lu Y, Luo X, Li B et al. (2007). The musclespecific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat Med 13: 486–491. Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C et al. (2004). Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117: 927–939. Yasui DH, Genetta T, Kadesch T, Williams TM, Swain SL, Tsui LV et al. (1998). Transcriptional repression of the IL-2 gene in Th cells by ZEB. J Immunol 160: 4433–4440. Yekta S, Shih IH, Bartel DP. (2004). MicroRNA-directed cleavage of HOXB8 mRNA. Science 304: 594–596. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S et al. (2007). Induced pluripotent stem cell lines derived from human somatic cells. Science 318: 1917–1920. Zanette DL, Rivadavia F, Molfetta GA, Barbuzano FG, Proto-Siqueira R, Silva-Jr WA. (2007). miRNA Expression profiles in chronic lymphocytic and acute lymphocytic leukemia. Braz J Med Biol Res 40: 1435–1440. Zeng Y. (2006). Principles of micro-RNA production and maturation. Oncogene 25: 6156–6162. Zeng Y, Cullen BR. (2006). Recognition and cleavage of primary microRNA transcripts. Methods Mol Biol 342: 49–56. Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A et al. (2006). microRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci USA 103: 9136–9141. Zhou B, Wang S, Mayr C, Bartel DP, Lodish HF. (2007). miR-150, a microRNA expressed in mature B and T cells, blocks early B cell development when expressed prematurely. Proc Natl Acad Sci USA 104: 7080–7085. Zhu S, Si ML, Wu H, Mo YY. (2007). MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem 282: 14328–14336.