Skip to main content
Genes & Cancer logoLink to Genes & Cancer
. 2010 Jun;1(6):532–541. doi: 10.1177/1947601910378024

Myc

The Beauty and the Beast

Amanda R Wasylishen 1,2, Linda Z Penn 1,2,
Editor: Chi Van Dang
PMCID: PMC3092215  PMID: 21779457

Abstract

The iconic history of the Myc oncoprotein encompasses 3 decades of intense scientific discovery. There is no question that Myc has been a pioneer, advancing insight into the molecular basis of cancer as well as functioning as a critical control center for several diverse biological processes and regulatory mechanisms. This narrative chronicles the journey and milestones that have defined the understanding of Myc, and it provides an opportunity to consider future directions in this challenging yet rewarding field.

Keywords: Myc, oncogene, cancer, transcription, cell cycle


As one of the first oncogenes identified, Myc has enjoyed an illustrious history (see Figure 1). In reflection of 30 years of research and in excess of 20,000 published articles, 2 major themes emerge. First, the fine-tuned regulation and numerous activities of Myc in normal cells are unparalleled and a beauty to behold. Second, when this regulation is lost, Myc rears its beastly head and takes the role of one of the most prominent and intriguing oncoproteins. This review focuses on the history and early work that has defined Myc. It also provides perspective for the articles that follow, which will comprehensively discuss Myc in its current context.

Figure 1.

Figure 1.

Snapshots from Myc’s history.

MYC Is Established as a Prominent Oncogene

Identification of MYC

In the late 1970s, an avian acute leukemia virus (MC29) was shown to promote a spectrum of malignancies, including myelocytomas, sarcomas, and importantly, carcinomas. This ability to induce carcinomas was of particular interest because this represented the most common form of human malignancy. The transforming sequence of MC29 was identified as v-myc and named myelocytomatosis for a resultant leukemia. v-myc was found to present in the cells as a 110-kDa v-gag-myc fusion. Consistent with the notion that oncogenes could be stolen by retroviruses, the cellular homologue was identified soon after in normal cells from many species. The discovery of MYC further reinforced the startling realization that oncogenic transformation could be caused by the activation of a cellular gene.14

Mechanisms of Deregulation in Cancer

Building on the awareness that, unlike other proto-oncogenes, MYC activation was not a consequence of mutations in the coding sequence, research focused on identifying and understanding other modes of oncogenic activation. This led to the discovery of 3 novel mechanisms through which Myc and, in turn, other oncogenes could be deregulated and promote transformation: insertional mutagenesis, chromosomal translocation, and gene amplification. Combined, these findings led the way for the discovery and understanding of oncogenes and provided new paradigms for the genetic basis of cancers.

In addition to the acutely transforming retrovirus mentioned previously, a second class of retroviruses had been shown to induce leukemias and lymphomas. These viruses, often referred to as non–acutely transforming retroviruses, induced tumors with a much longer latency and were unable to transform cells in culture. Although these initial observations were puzzling, the mechanisms through which these viruses promoted tumorigenesis were soon realized when retroviruses were shown to activate the expression of oncogenes through proviral insertion.58 Specifically, viruses such as the acute leukosis virus (ALV) integrated into the host genome at or near proto-oncogenes, resulting in a high level of expression driven by the viral promoter. Myc was the first oncogene found to be activated by this mechanism, with 80% of B-cell lymphomas induced by ALV owing to activated MYC. These groundbreaking studies laid the foundation for the discovery of several other oncogenes.9

Nonrandom chromosomal translocations had been observed in a number of malignancies, including Burkitt lymphoma and chronic myeloid leukemia. It was tempting to speculate that these translocations resulted in aberrant expression of the same proto-oncogenes identified in the acutely transforming retroviruses. The mapping of MYC to the long arm of chromosome 8 gave creed to this hypothesis.10,11 Specifically, Burkitt lymphomas had been characterized to contain reciprocal translocations between chromosome 8 and chromosome 14, 2, or 22, which harbor immunoglobulin (Ig) heavy and light chain genes.12 It was then discovered that these cancers were driven by activated expression of MYC resulting from the translocation. The first MYC transgenic mouse, Eµ-Myc, was developed to model Burkitt lymphoma, with activated MYC expression driving a clonal B-cell lymphoma.13 Mouse plasmacytomas were similarly found to be a consequence of MYC translocation with the Ig heavy chain locus.14,15

It was well established that cancer cells contained a number of chromosomal abnormalities, including the presence of double minute chromosomes and homogeneously staining regions. The contributions of these aberrations to cellular transformation were largely appreciated through the study of MYC. Human colon cancer (COLO-320) and acute promyelocytic leukemia (HL-60) cell lines were shown to express multiple copies of MYC.1618 Importantly, gene amplifications were observed in primary patient material, including uncultured samples from the patient whose tumor was the source for HL-60 cells.17 Overall, these findings further supported the understanding that deregulated expression of a proto-oncogene could promote neoplastic transformation.

A Family Affair

Through these studies, it became clear that MYC was a member of a larger family of oncogenes. When v-myc was used as a probe in hybridization experiments, additional restriction fragments were consistently and reproducibly identified, which suggested that the human genome contained a series of sequences that were similar or related to the MYC oncogene. Amplification of a new MYC family member was soon identified in a panel of neuroblastoma samples and cell lines.19,20 This new oncogene was named MYCN, for the neuroblastomas in which it was identified. Importantly, MYCN was quickly associated with aggressive disease and poor outcomes in this pediatric cancer, and amplifications were assigned great prognostic value.21,22 MYCN amplification in neuroblastoma provided one of the earliest examples of how basic research involving oncogenes could be translated into a clinically relevant prognostic marker, a principle that remains at the forefront of cancer research today.

Within an additional couple of years, a third family member was identified through gene amplifications in small-cell lung cancer and was fittingly named MYCL1.23 Further research identified amplifications of these 3 transforming members of the Myc family in a number of other tumor types.24 These amplification studies expanded the role for the Myc family in the pathogenesis of a large number and wide variety of human malignancies, beyond leukemias and lymphomas.

With a Little Help from MYC’s Friends

MYC was also fundamental to the understanding of oncogene cooperation and how cellular oncogenes could promote tumorigenesis. These foundations were built through studies in primary rat embryo fibroblasts.25 Ectopic expression of activated RAS (EJ-RAS) alone was insufficient to promote transformation; however, cotransfection of EJ-RAS and v-Myc or c-Myc promoted foci formation in vitro, which provided the first evidence that cooperation between cellular oncogenes was essential for cellular transformation. This cooperation model was supported by observations from MYC transgenic models. The tumors that arose in Eµ-Myc mice were clonal in origin and thereby suggested a need to acquire a secondary mutation for tumor development.13 When Eµ-Myc mice were crossed with Eµ-Bcl2 mice, driving constitutive overexpression of the antiapoptotic BCL2 oncogene, tumors formed more rapidly than the single transgenic.26,27 The model of oncogene cooperation and the need for multiple collaborating alterations to promote tumorigenesis have become fundamental to our current understanding of the neoplastic process.28

Myc Regulation in Nontransformed Cells

It was clear that there were multiple modes through which deregulated Myc expression could be achieved, which shifted the focus to understanding the normal regulation of Myc. Serum stimulation of quiescent cells in culture resulted in a rapid upregulation of Myc mRNA.29 The induction peaked at 2 hours poststimulation, was cycloheximide independent, and established MYC as an intermediate–early response gene. The converse was also true that antiproliferative signals resulted in rapid downregulation of Myc.3033 Additionally, Myc mRNA and protein demonstrated short half-lives.34,35 As such, an appreciation was developed for the tight regulation of Myc expression in non-transformed cells, which was exceptionally responsive to extracellular cues. Because Myc is one of the most highly regulated proteins in the cell, many important regulatory mechanisms have been discovered through studies of Myc.

Transcriptional Control and mRNA Turnover

From early on, considerable research efforts were directed at understanding the regulation of MYC at the transcriptional level. The gene was shown to have rather unusual topology, with a large noncoding first exon and 2 additional coding exons (reviewed in References 36 and 37). Within the 5′ region of exon 1, transcription can be initiated from 2 promoters (P1 and P2) separated by 150 base pairs, with the majority of transcripts arising from P2. The complex array of regulatory mechanisms and the multitude of signaling cascades that converge on MYC have made this a challenging area of research. MYC, however, has maintained its role as a pioneer and was the first eukaryotic gene found to be regulated by transcriptional elongation control.3841 There is clearly much remaining to be discovered, and recent studies suggest that we are now in a strong position to better delineate some of these mechanisms. For example, through their interactions with the far upstream element (FUSE; 1.7 kb upstream of P2), FUSE-binding protein (FBP) and FBP-interacting repressor have recently been identified as important players in regulating transcription of MYC.42,43 Additionally, genomewide association studies have linked single-nucleotide polymorphisms on chromosome 8q24, upstream of MYC, to an increased risk of some cancers.4449 Further studies will continue to advance our understanding of the transcriptional regulation of MYC.

Protein Expression and Regulation

Myc has 2 primary protein products that arise from an AUG codon at the 5′ end of exon II and a CUG initiation codon at the 3′ end of exon I that can be visualized as 64-kDa and 67-kDa proteins by immunoblot analysis.37,50,51 An additional short isoform, MycS, that lacks the first 100 amino acids has been reported52; however, the functional and/or regulatory role of this molecule is not fully understood. Tryptic phosphopeptide mapping has revealed a number of serine and threonine sites within Myc that are phosphorylated in vivo.5355 Of these, serine-62 (S62) and threonine-58 (T58) have dominated our understanding of Myc posttranslational modifications.56,57 Much of the interest in these residues has been driven by the identification of point mutations in the homologous threonine of v-Myc (T61) and in the detection of point mutations at S62 and T58 and surrounding residues in some cases of Burkitt and AIDS-associated lymphomas.5861 Collectively, research has shown that S62 is phosphorylated in response to proliferative signals, which are thought to stabilize and promote c-Myc activation. Phosphorylated S62 then allows GSK3 to bind and phosphorylate T58, which subsequently promotes binding of FBXW7 and recruits the SCF complex, leading to the ubiquitylation and proteosomal degradation of c-Myc. Interestingly, many regulators of Myc are themselves oncogenes and tumor suppressor genes, and targeting these signaling pathways provides an indirect approach to inhibit Myc activity. In addition, c-Myc has been shown to be O-glycosylated and acetylated, although our understanding of the biological role of Myc posttranslational modifications remains in its infancy. This is remarkable for a protein like Myc, which has been at the forefront of cancer research for decades, and it is clear that this will be an important area of future work.

The Biological Activities of Myc

Cell Cycle and Differentiation

That the deregulation of proto-oncogenes was fundamental in tumorigenesis made it natural to speculate that they functioned in normal cell proliferation. There were several lines of evidence to suggest that Myc might play a role in cell cycle progression. As mentioned previously, MYC was established as an intermediate–early response gene when serum stimulation of quiescent cells rapidly induced Myc mRNA.29 Additionally, ectopic expression of Myc promoted growth factor–independent proliferation of mouse fibroblasts in culture.62 More definitive evidence came several years later through the use of conditional Myc–estrogen receptor (Myc-ER) chimeras, in which Myc was fused to the hormone-binding domain of the ER.63 Activation of Myc-ER by estradiol promoted cell cycle entry of quiescent cells in tissue culture.64 Therefore, it was clear that Myc functioned in promoting cell cycle progression.

Not unrelated to the ability to promote cell cycle progression, Myc expression was shown to block differentiation. It was demonstrated by multiple groups and through a number of models that Myc downregulation was essential for cells to exit the cell cycle and undergo differentiation. For example, ectopic expression of Myc was shown to block the differentiation of 3T3-L1 preadipocyte,65 MEL murine erythroleukemia,6668 and F9 teratocarcinoma69 cell lines. These abilities to promote cell proliferation and block differentiation have natural associations with tumorigenesis and are features of aggressive disease.

Apoptosis

With well-established contributions to tumorigenesis and convincing evidence to suggest that Myc played a role in cell cycle and proliferation, the field was met with much surprise in the early 1990s when ectopic expression of Myc was shown to sensitize cells to undergo apoptosis.7072 It was initially perplexing—why and how could a single molecule drive such disparate functions? As was the case previously, this groundbreaking observation for Myc soon held true for other oncogenes, such as E2F1 and E1A.73 With this, a dual-signal model emerged. Myc was able to encourage both proliferation and apoptosis. Apoptosis, however, could be averted by survival signals, including those from insulin- and platelet-derived growth factors.72,74 Myc’s ability to promote apoptosis provided a built-in safety mechanism to protect against inappropriate proliferation as a consequence of deregulated Myc. This finding shed considerable light on the model of oncogene cooperation and how an antiapoptotic protein such as Bcl2 could cooperate with Myc to promote oncogenesis.27,75,76 Specifically, it suggested that abrogating Myc-induced apoptosis was a critical component of cellular transformation.

Within a few years, the field had a better appreciation for some of the mechanisms through which Myc could drive apoptosis. Deregulation of Myc was shown to activate the tumor suppressor p53 through the upregulation of ARF.7779 A loss of either of these tumor suppressors accelerated tumorigenesis in mouse models of oncogenic Myc.8084 Other regulators of the ARF-MDM2-p53 axis have been shown to cooperate with Myc to promote cellular transformation.8587 Furthermore, Myc is able to promote apoptosis through p53-independent mechanisms by influencing the balance between proapoptotic and antiapoptotic proteins in the cell.73,8894 Combined, there is much interest in improving our understanding of the mechanisms through which Myc promotes apoptosis in the cell and how this activity is abrogated in cancer.

Molecular Mechanisms

Although it was vital to determine Myc’s biological activities in the cell, it was equally important to understand its mechanisms of action. Early work studying v-gag-myc using antibodies against gag demonstrated that, unlike many other oncogenes at that time, Myc was localized to the nucleus and able to bind double-stranded DNA.95,96 Soon after, this observation was confirmed for the native protein.97 From this, 2 models emerged. First, Myc was hypothesized to play a role in DNA replication. Second, Myc was believed to regulate gene transcription, either directly or indirectly. It was the latter model that soon took center stage.

Transcriptional Activation by Myc

Unlike in the first decade of research—in which Myc was at the leading edge, forging new territory and serving as a model for the understanding of other oncoproteins—the field relied on previous work on other proteins with similar domains when it came to understanding the mechanisms of Myc activity. The C-terminus of Myc was shown to contain both a helix-loop-helix98 domain and a leucine zipper domain.99 At the time, it was unusual for a protein to contain both these domains, but it encouraged the field to focus on potential protein and DNA interactions. Importantly, through deletion and insertion mutants, these regions were found to be essential for all functional readouts of Myc activity: REF focus formation and Rat1A colony formation, negative autoregulation, and the inhibition of differentiation.100

The parallel discoveries of a specific DNA-binding sequence for Myc provided a much-needed boost to researchers in the field.101,102 The first report showed that homodimers of Myc C-terminal domains could bind to CACGTG sequences in vitro. This work and the low affinity with which these dimers formed suggested that Myc must have a partner. The search was on, and soon after Max (Myc-associated factor X) was identified.103 Max was shown to be a constant and obligate partner for Myc, with consistent and abundant expression in proliferating and quiescent cells, which was not altered in response to extracellular stimuli. It is important to note that Max also forms heterodimers with members of the Mxd family, thereby providing an additional mechanism to regulate Myc activity in the cell.104,105

The first direct evidence of Myc as a transcriptional activator came from fusion of the Myc N-terminal domain to the yeast Gal4 DNA-binding domain.106 These fusions were able to activate expression from a reporter construct. The ability of Myc to function as a transcriptional regulator was advanced with the discoveries of other interacting proteins, which were in turn components of chromatin-remodeling complexes. For example, the N-terminal domain of Myc was shown to interact with TRRAP and subsequently recruit histone acetyl transferase activity.107,108

Transcriptional Repression by Myc

Some of the initial evidence showing that Myc was able to function as a transcriptional repressor emerged from studies of Myc expression that suggested negative autoregulation. Specifically, several reports demonstrated that Myc was not expressed from the nontranslocated allele in Burkitt lymphoma.109,110 It was further shown that expression of v-myc was able to downregulate expression of endogenous Myc, and the same was true for ectopic Myc expression.111,112 Structure–function studies then revealed that the regions essential for transformation were the same as those required for this negative autoregulation.113,114 Despite these early observations, the mechanisms of transcriptional repression by Myc remained elusive for some time. In due course, repression was shown to be independent of binding to E-boxes.115 Additionally, bona fide repressed targets and key interactors were identified that developed our current understanding of transcriptional repression by Myc. For example, MYC-interacting zinc finger 1 (Miz1) was identified as an interacting protein and important mediator of Myc function.116,117 As such, the current model is that Myc–Max heterodimers interact with transcriptional activators, such as nuclear factor Y and Miz1, which are directly bound to DNA through enhancer or initiator elements.118,119 This binding displaces coactivators and recruits corepressors, thereby preventing transcriptional activation. Even though the understanding of Myc as a transcriptional repressor lagged behind its role in activating gene transcription, it is clear that this activity is important for Myc function.

Target Genes

Not only was the development of the Myc-ER fusion instrumental in defining Myc’s role in driving the cell cycle, but it also provided a tool for the discovery of the first Myc target genes.64 The activation of Myc in the presence of cycloheximide to block protein synthesis identified α-prothymosin as a transcriptional target of Myc. This same approach was used to identity a number of other target genes, including ornithine decarboxylase 1.120,121 Another valuable approach used to identify Myc targets employed mitogen stimulation of quiescent Myc-null rat fibroblast cells to determine if transcriptional regulation was dependent on Myc.122 This method subsequently demonstrated CAD and GADD45A as activated and repressed Myc targets, respectively.123125 Although gene-by-gene approaches were successful overall, identification of Myc targets was slow and laborious.126

Genomewide approaches and expression microarrays transformed our ability to identify Myc target genes. This was met with its own challenges, however, in that consistency between studies was quite poor. One of the major explanations was that Myc induced only small changes in the mRNA abundance of targets, which made their identification difficult.127 This prompted the field to establish criteria for designating Myc target genes and to develop a database to catalog and manage genes based on these criteria (http://myc-cancer-gene.org/).

More recently, new technologies have been used to survey Myc binding to chromatin in cells, including DNA adenine methyltransferase identification and chromatin immunoprecipitation (ChIP). These techniques have revolutionized our ability to evaluate protein–DNA interactions in vivo, and they have further improved our ability to identify direct Myc target genes. Additionally, the specificity afforded by ChIP assays, combined with the high-throughput capabilities of microarrays (ChIP-chip) or high-throughput nucleotide sequencing (ChIP-PET and ChIP-seq), has provided the field with the ability to interrogate genomewide binding of Myc. Data from these studies suggest an expanded role for Myc. Binding 10% to 15% of the genome, Myc has been shown to regulate the expression of genes encoding noncoding RNAs, as well as proteins.127,128 The identification of the direct target genes has started to shed some light on how Myc is able to promote such an array of cellular processes. For example, Myc promotes the cell cycle by activation of cyclins D1, D2, E1, and A1, as well as the repression of p21, p27, p15, and GADD45. However, it has become clear that Myc does not act solely as a classic transcription factor, regulating a small number of target genes to promote a given biological function. Rather, Myc can globally influence chromatin structure and affect genetic programs.129 We direct you to several excellent reviews that recently discussed Myc regulation of target genes and chromatin (including articles in this issue by McMahon130 and Bui & Mendell131).2,127,129,132,133

New Biological Activities Are Added to Myc’s Arsenal

By the late 1990s, Myc had added several different biological activities to its collection and was shown to regulate cell size, affect genomic stability, and trigger the angiogenic switch. First, Myc demonstrated an ability to promote cell growth both in vitro and in vivo by supplying the cell with several classes of basic building blocks and increasing protein synthesis and cell metabolism.134,135 Many of the universally regulated Myc target genes supported this activity, including those involved in ribosomal and mitochondrial biogenesis, cellular metabolism, and protein and nucleic acid synthesis.136,137 Second, cells with deregulated Myc were shown to have a higher rate of specific gene amplifications, and further studies indicated that Myc could promote chromosomal instability.138141 Although this activity seems to be context dependent, several mechanisms have been proposed, including alterations in chromosome structure, overcoming the p53 checkpoint, and increased levels of reactive oxygen species.142,143 The third of these novel activities is the ability of Myc to stimulate neovascularization, which has been evident in a number of in vivo models.144147 The downregulation of the angiogenesis suppressor thrombospondin and release of interleukin 1β have been shown to play a role in this process. As such, there is no question that Myc’s ability to influence these hallmarks would contribute to tumorigenesis, and we recommend several recent review articles for thorough discussions of these activities.2,132,143,148

In recent years, additional biological activities of Myc have been characterized. The renewed interest in the Warburg effect and tumor cell metabolism has highlighted a new role for Myc. In addition to stimulating mitrochondrial biogenesis, oncogenic levels of Myc have been shown to promote glutaminolysis.136,149-151 This increased glutamine metabolism fuels cell growth and proliferation, which are essential for tumor cells to thrive. There have also been suggestions that tumor cells become addicted to glutamine, which may provide opportunities for therapeutic intervention.136

Although oncogene-induced senescence had been widely recognized as a mechanism through which normal cells protect themselves against transformation, it was only recently that this function was assigned to Myc. Unlike Ras, overexpression of Myc alone does not induce senescence. Myc can, however, induce senescence in the context of the loss of genes such as WRN or CDK2.152-156 Future research will inevitably explore if Myc-induced senescence can be harnessed to promote tumor regression.

The ability of Myc to block differentiation perhaps foreshadowed the recently uncovered role of Myc in regulating stemness. Conditional knockout mice have demonstrated an essential role for Myc in the normal developmental control of hematopoetic and neuronal stem cells.157,158 Myc has recently been identified as 1 of the 4 genes whose overexpression could reprogram normal terminally differentiated fibroblasts into induced pluripotent stem cells.159,160 Although it was later shown that Myc was dispensable for this process,161 it did underscore important implications for deregulated Myc in initiating and maintaining tumor stem cells. This speaks to the observation that the stem cell signature of undifferentiated and aggressive tumors carries great similarity with the phenotypes of Myc-activated tumors. With such an impressive array of biological activities, the profound impact of Myc deregulation in cancer is even more apparent.

Prospects for Taming the Beast

Given the well-established and widespread role of Myc in promoting tumorigenesis, the question becomes, can we exploit this potent oncoprotein for the diagnosis and treatment of cancers? Inducible transgenic models have demonstrated that even a brief inactivation of Myc can have a profound and cell type–specific effect on tumor cell fate.162 Importantly, these models have demonstrated that targeting Myc activity is associated with tumor regression. Despite this, translating our understanding of Myc for clinical application has been met by several hurdles. First, the diverse mechanisms through which the widespread deregulation of Myc occurs in cancers preclude traditional molecular diagnostics. As our understanding of the molecular mechanisms of Myc activities continues to advance, it may become possible for molecular pathologists to score deregulation based on activity. Second, the Myc protein is not a classically “druggable” molecule, because it is localized in the nucleus and does not possess enzymatic activity that can be readily blocked. Approaches to target the regulation of Myc or the effectors of Myc activity may provide alternative means to inhibit this oncoprotein. There have been additional concerns about a therapeutic window, given that Myc is essential in all cells. Recent work has directly addressed this important issue and established Myc as a genuine molecular target in cancer. Briefly, a mutant basic helix-loop-helix leucine zipper domain referred to as Omomyc was developed and shown to bind to and inhibit transcriptional activation by Myc.163,164 In preclinical studies, Omomyc eradicated tumors in a mouse model of Ras-induced lung adenocarcinoma. Moreover, the systemic inhibition of Myc was well tolerated by normal tissues, and all side effects were rapidly reversible. We direct readers to several review articles for further discussions of Myc as a therapeutic target in cancer,165,166 including those in this issue by Frenzel, Lovén, & Henriksson167 as well as by Prochownick & Vogt.168 In sum, targeting Myc activity in cancer is an exciting avenue of current research and has great potential to impact patient care.

Concluding Thoughts

In reflection of just over 30 years of research, there is no question that Myc has been and will continue to be an influential and significant molecule. Many seminal discoveries have been made through studying Myc, and we hope that this review highlights both the beauty and the beast that is Myc.

Acknowledgments

We would like to thank all of our colleagues who have shared in the fascinating and challenging study of Myc, and we apologize to those whose work we were unable to include because of space constraints. We additionally thank the members of the Penn Lab for helpful discussions and critical review of this article.

Footnotes

This work was supported by grants from the Canadian Cancer Society Research Institute, the Natural Sciences and Engineering Research Council of Canada, and the Canadian Institutes of Health Research (FRN CPG99377) (L.Z.P.), as well as a Canadian Breast Cancer Foundation–Ontario Region Doctoral Fellowship (A.R.W.). Additional support was provided by the Ontario Ministry of Health and Long Term Care. The views expressed do not necessarily reflect those of the ministry.

The authors declare no conflicts of interest with respect to the publication of this article.

References

  • 1. Varmus HE. The molecular genetics of cellular oncogenes. Annu Rev Genet 1984;18:553-612 [DOI] [PubMed] [Google Scholar]
  • 2. Meyer N, Penn LZ. Reflecting on 25 years with MYC. Nat Rev Cancer 2008;8:976-90 [DOI] [PubMed] [Google Scholar]
  • 3. Sheiness D, Bishop JM. DNA and RNA from uninfected vertebrate cells contain nucleotide sequences related to the putative transforming gene of avian myelocytomatosis virus. J Virol 1979;31:514-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Roussel M, Saule S, Lagrou C, et al. Three new types of viral oncogene of cellular origin specific for haematopoietic cell transformation. Nature 1979;281:452-5 [DOI] [PubMed] [Google Scholar]
  • 5. Neel BG, Hayward WS, Robinson HL, Fang J, Astrin SM. Avian leukosis virus-induced tumors have common proviral integration sites and synthesize discrete new RNAs: oncogenesis by promoter insertion. Cell 1981;23:323-34 [DOI] [PubMed] [Google Scholar]
  • 6. Payne GS, Courtneidge SA, Crittenden LB, Fadly AM, Bishop JM, Varmus HE. Analysis of avian leukosis virus DNA and RNA in bursal tumours: viral gene expression is not required for maintenance of the tumor state. Cell 1981;23:311-22 [DOI] [PubMed] [Google Scholar]
  • 7. Payne GS, Bishop JM, Varmus HE. Multiple arrangements of viral DNA and an activated host oncogene in bursal lymphomas. Nature 1982;295:209-14 [DOI] [PubMed] [Google Scholar]
  • 8. Hayward WS, Neel BG, Astrin SM. Activation of a cellular onc gene by promoter insertion in ALV-induced lymphoid leukosis. Nature 1981;290:475-80 [DOI] [PubMed] [Google Scholar]
  • 9. Peters G. Oncogenes at viral integration sites. Cell Growth Differ 1990;1:503-10 [PubMed] [Google Scholar]
  • 10. Dalla-Favera R, Bregni M, Erikson J, Patterson D, Gallo RC, Croce CM. Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells. Proc Natl Acad Sci U S A 1982;79:7824-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Neel BG, Jhanwar SC, Chaganti RS, Hayward WS. Two human c-onc genes are located on the long arm of chromosome 8. Proc Natl Acad Sci U S A 1982;79:7842-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Taub R, Kirsch I, Morton C, et al. Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells. Proc Natl Acad Sci U S A 1982;79:7837-41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Adams JM, Harris AW, Pinkert CA, et al. The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nature 1985;318:533. [DOI] [PubMed] [Google Scholar]
  • 14. Shen-Ong GL, Keath EJ, Piccoli SP, Cole MD. Novel myc oncogene RNA from abortive immunoglobulin-gene recombination in mouse plasmacytomas. Cell 1982;31:443-52 [DOI] [PubMed] [Google Scholar]
  • 15. Crews S, Barth R, Hood L, Prehn J, Calame K. Mouse c-myc oncogene is located on chromosome 15 and translocated to chromosome 12 in plasmacytomas. Science 1982;218:1319-21 [DOI] [PubMed] [Google Scholar]
  • 16. Alitalo K, Schwab M, Lin CC, Varmus HE, Bishop JM. Homogeneously staining chromosomal regions contain amplified copies of an abundantly expressed cellular oncogene (c-myc) in malignant neuroendocrine cells from a human colon carcinoma. Proc Natl Acad Sci U S A 1983;80:1707-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Dalla-Favera R, Wong-Staal F, Gallo RC. Onc gene amplification in promyelocytic leukaemia cell line HL-60 and primary leukaemic cells of the same patient. Nature 1982;299:61-3 [DOI] [PubMed] [Google Scholar]
  • 18. Collins S, Groudine M. Amplification of endogenous myc-related DNA sequences in a human myeloid leukaemia cell line. Nature 1982;298:679-81 [DOI] [PubMed] [Google Scholar]
  • 19. Schwab M, Alitalo K, Klempnauer KH, et al. Amplified DNA with limited homology to myc cellular oncogene is shared by human neuroblastoma cell lines and a neuroblastoma tumour. Nature 1983;305:245-8 [DOI] [PubMed] [Google Scholar]
  • 20. Kohl NE, Kanda N, Schreck RR, et al. Transposition and amplification of oncogene-related sequences in human neuroblastomas. Cell 1983;35:359-67 [DOI] [PubMed] [Google Scholar]
  • 21. Schwab M, Ellison J, Busch M, Rosenau W, Varmus HE, Bishop JM. Enhanced expression of the human gene N-myc consequent to amplification of DNA may contribute to malignant progression of neuroblastoma. Proc Natl Acad Sci U S A 1984;81:4940-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Brodeur GM, Seeger RC, Schwab M, Varmus HE, Bishop JM. Amplification of N-myc in untreated human neuroblastomas correlates with advanced disease stage. Science 1984;224:1121-4 [DOI] [PubMed] [Google Scholar]
  • 23. Nau MM, Brooks BJ, Battey J, et al. L-myc, a new myc-related gene amplified and expressed in human small cell lung cancer. Nature 1985;318:69-73 [DOI] [PubMed] [Google Scholar]
  • 24. Zimmerman K, Alt FW. Expression and function of myc family genes. Crit Rev Oncog 1990;2:75-95 [PubMed] [Google Scholar]
  • 25. Land H, Parada LF, Weinberg RA. Tumorigenic conversion of primary embryo fibroblasts requires at least two cooperating oncogenes. Nature 1983;304:596-602 [DOI] [PubMed] [Google Scholar]
  • 26. Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature 1988;335:440-2 [DOI] [PubMed] [Google Scholar]
  • 27. Strasser A, Harris AW, Bath ML, Cory S. Novel primitive lymphoid tumours induced in transgenic mice by cooperation between myc and bcl-2. Nature 1990;348:331-3 [DOI] [PubMed] [Google Scholar]
  • 28. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70 [DOI] [PubMed] [Google Scholar]
  • 29. Kelly K, Cochran BH, Stiles CD, Leder P. Cell-specific regulation of the c-myc gene by lymphocyte mitogens and platelet-derived growth factor. Cell 1983;35:603-10 [DOI] [PubMed] [Google Scholar]
  • 30. Lachman HM, Skoultchi AI. Expression of c-myc changes during differentiation of mouse erythroleukaemia cells. Nature 1984;310:592-4 [DOI] [PubMed] [Google Scholar]
  • 31. Gonda TJ, Metcalf D. Expression of myb, myc and fos proto-oncogenes during the differentiation of a murine myeloid leukaemia. Nature 1984;310:249-51 [DOI] [PubMed] [Google Scholar]
  • 32. Campisi J, Gray HE, Pardee AB, Dean M, Sonenshein GE. Cell-cycle control of c-myc but not c-ras expression is lost following chemical transformation. Cell 1984;36:241-7 [DOI] [PubMed] [Google Scholar]
  • 33. Dean M, Levine RA, Ran W, Kindy MS, Sonenshein GE, Campisi J. Regulation of c-myc transcription and mRNA abundance by serum growth factors and cell contact. J Biol Chem 1986;261:9161-6 [PubMed] [Google Scholar]
  • 34. Dani C, Blanchard JM, Piechaczyk M, El Sabouty S, Marty L, Jeanteur P. Extreme instability of myc mRNA in normal and transformed human cells. Proc Natl Acad Sci U S A 1984;81:7046-50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Hann SR, Eisenman RN. Proteins encoded by the human c-myc oncogene: differential expression in neoplastic cells. Mol Cell Biol 1984;4:2486-97 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Marcu KB, Bossone SA, Patel AJ. myc function and regulation. Annu Rev Biochem 1992;61:809-60 [DOI] [PubMed] [Google Scholar]
  • 37. Spencer CA, Groudine M. Control of c-myc regulation in normal and neoplastic cells. Adv Cancer Res 1991;56:1-48 [DOI] [PubMed] [Google Scholar]
  • 38. Bentley DL, Groudine M. A block to elongation is largely responsible for decreased transcription of c-myc in differentiated HL60 cells. Nature 1986;321:702-6 [DOI] [PubMed] [Google Scholar]
  • 39. Bentley DL, Groudine M. Sequence requirements for premature termination of transcription in the human c-myc gene. Cell 1988;53:245-56 [DOI] [PubMed] [Google Scholar]
  • 40. Nepveu A, Marcu KB. Intragenic pausing and anti-sense transcription within the murine c-myc locus. EMBO J 1986;5:2859-65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Eick D, Bornkamm GW. Transcriptional arrest within the first exon is a fast control mechanism in c-myc gene expression. Nucleic Acids Res 1986;14:8331-46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Levens D. How the c-myc promoter works and why it sometimes does not. J Natl Cancer Inst Monogr 2008;39:41-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Chung HJ, Levens D. c-myc expression: keep the noise down! Mol Cells 2005;20:157-66 [PubMed] [Google Scholar]
  • 44. Kiemeney LA, Thorlacius S, Sulem P, et al. Sequence variant on 8q24 confers susceptibility to urinary bladder cancer. Nat Genet 2008;40:1307-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Tomlinson I, Webb E, Carvajal-Carmona L, et al. Genome-wide association scan of tag SNPs identifies a susceptibility variant for colorectal cancer at 8q24.21. Nat Genet 2007;39:984-8 [DOI] [PubMed] [Google Scholar]
  • 46. Ahmadiyeh N, Pomerantz MM, Grisanzio C, et al. 8q24 prostate, breast, and colon cancer risk loci show tissue-specific long-range interaction with MYC. Proc Natl Acad Sci U S A 2010;107:9742-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Pomerantz MM, Beckwith CA, Regan MM, et al. Evaluation of the 8q24 prostate cancer risk locus and MYC expression. Cancer Res 2009;69:5568-74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Pomerantz MM, Ahmadiyeh N, Jia L, et al. The 8q24 cancer risk variant rs6983267 shows long-range interaction with MYC in colorectal cancer. Nat Genet 2009;41:882-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Haiman CA, Patterson N, Freedman ML, et al. Multiple regions within 8q24 independently affect risk for prostate cancer. Nat Genet 2007;39:638-44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Cole MD. The myc oncogene: its role in transformation and differentiation. Annu Rev Genet 1986;20:361-84 [DOI] [PubMed] [Google Scholar]
  • 51. Littlewood TD, Evan GI. The role of myc oncogenes in cell growth and differentiation. Adv Dent Res 1990;4:69-79 [DOI] [PubMed] [Google Scholar]
  • 52. Spotts GD, Patel SV, Xiao Q, Hann SR. Identification of downstream-initiated c-Myc proteins which are dominant-negative inhibitors of transactivation by full-length c-Myc proteins. Mol Cell Biol 1997;17:1459-68 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Luscher B, Kuenzel EA, Krebs EG, Eisenman RN. Myc oncoproteins are phosphorylated by casein kinase II. EMBO J 1989;8:1111-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Lutterbach B, Hann SR. Hierarchical phosphorylation at N-terminal transformation-sensitive sites in c-Myc protein is regulated by mitogens and in mitosis. Mol Cell Biol 1994;14:5510-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Lutterbach B, Hann SR. Overexpression of c-Myc and cell immortalization alters c-Myc phosphorylation. Oncogene 1997;14:967-75 [DOI] [PubMed] [Google Scholar]
  • 56. Hann SR. Role of post-translational modifications in regulating c-Myc proteolysis, transcriptional activity and biological function. Semin Cancer Biol 2006;16:288-302 [DOI] [PubMed] [Google Scholar]
  • 57. Vervoorts J, Luscher-Firzlaff J, Luscher B. The ins and outs of MYC regulation by posttranslational mechanisms. J Biol Chem 2006;281:34725-9 [DOI] [PubMed] [Google Scholar]
  • 58. Albert T, Urlbauer B, Kohlhuber F, Hammersen B, Eick D. Ongoing mutations in the N-terminal domain of c-Myc affect transactivation in Burkitt’s lymphoma cell lines. Oncogene 1994;9:759-63 [PubMed] [Google Scholar]
  • 59. Bhatia K, Spangler G, Gaidano G, Hamdy N, Dalla-Favera R, Magrath I. Mutations in the coding region of c-myc occur frequently in acquired immunodeficiency syndrome-associated lymphomas. Blood 1994;84:883-8 [PubMed] [Google Scholar]
  • 60. Clark HM, Yano T, Otsuki T, Jaffe ES, Shibata D, Raffeld M. Mutations in the coding region of c-MYC in AIDS-associated and other aggressive lymphomas. Cancer Res 1994;54:3383-6 [PubMed] [Google Scholar]
  • 61. Yano T, Sander CA, Clark HM, Dolezal MV, Jaffe ES, Raffeld M. Clustered mutations in the second exon of the MYC gene in sporadic Burkitt’s lymphoma. Oncogene 1993;8:2741-8 [PubMed] [Google Scholar]
  • 62. Armelin HA, Armelin MC, Kelly K, et al. Functional role for c-myc in mitogenic response to platelet-derived growth factor. Nature 1984;310:655-60 [DOI] [PubMed] [Google Scholar]
  • 63. Eilers M, Picard D, Yamamoto KR, Bishop JM. Chimaeras of myc oncoprotein and steroid receptors cause hormone-dependent transformation of cells. Nature 1989;340:66-8 [DOI] [PubMed] [Google Scholar]
  • 64. Eilers M, Schirm S, Bishop JM. The MYC protein activates transcription of the alpha-prothymosin gene. EMBO J 1991;10:133-41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Freytag SO. Enforced expression of the c-myc oncogene inhibits cell differentiation by precluding entry into a distinct predifferentiation state in G0/G1. Mol Cell Biol 1988;8:1614-24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Dmitrovsky E, Kuehl WM, Hollis GF, Kirsch IR, Bender TP, Segal S. Expression of a transfected human c-myc oncogene inhibits differentiation of a mouse erythroleukaemia cell line. Nature 1986;322:748-50 [DOI] [PubMed] [Google Scholar]
  • 67. Prochownik EV, Kukowska J. Deregulated expression of c-myc by murine erythroleukaemia cells prevents differentiation. Nature 1986;322:848-50 [DOI] [PubMed] [Google Scholar]
  • 68. Coppola JA, Cole MD. Constitutive c-myc oncogene expression blocks mouse erythroleukaemia cell differentiation but not commitment. Nature 1986;320:760-3 [DOI] [PubMed] [Google Scholar]
  • 69. Griep AE, Westphal H. Antisense Myc sequences induce differentiation of F9 cells. Proc Natl Acad Sci U S A 1988;85:6806-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Shi Y, Glynn JM, Guilbert LJ, Cotter TG, Bissonnette RP, Green DR. Role for c-myc in activation-induced apoptotic cell death in T cell hybridomas. Science 1992;257:212-4 [DOI] [PubMed] [Google Scholar]
  • 71. Evan GI, Wyllie AH, Gilbert CS, et al. Induction of apoptosis in fibroblasts by c-myc protein. Cell 1992;69:119-28 [DOI] [PubMed] [Google Scholar]
  • 72. Askew DS, Ashmun RA, Simmons BC, Cleveland JL. Constitutive c-myc expression in an IL-3-dependent myeloid cell line suppresses cell cycle arrest and accelerates apoptosis. Oncogene 1991;6:1915-22 [PubMed] [Google Scholar]
  • 73. Meyer N, Kim SS, Penn LZ. The Oscar-worthy role of Myc in apoptosis. Semin Cancer Biol 2006;16:275-87 [DOI] [PubMed] [Google Scholar]
  • 74. Harrington EA, Bennett MR, Fanidi A, Evan GI. c-Myc-induced apoptosis in fibroblasts is inhibited by specific cytokines. EMBO J 1994;13:3286-95 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Fanidi A, Harrington EA, Evan GI. Cooperative interaction between c-myc and bcl-2 protooncogenes. Nature 1992;359:554-6 [DOI] [PubMed] [Google Scholar]
  • 76. Bissonnette RP, Echeverri F, Mahboubi A, Green DR. Apoptotic cell death induced by c-myc is inhibited by bcl-2. Nature 1992;359:552-4 [DOI] [PubMed] [Google Scholar]
  • 77. Hermeking H, Eick D. Mediation of c-Myc-induced apoptosis by p53. Science 1994;265:2091-3 [DOI] [PubMed] [Google Scholar]
  • 78. Wagner AJ, Kokontis JM, Hay N. Myc-mediated apoptosis requires wild-type p53 in a manner independent of cell cycle arrest and the ability of p53 to induce p21waf1/cip1. Genes Dev 1994;8:2817-30 [DOI] [PubMed] [Google Scholar]
  • 79. Zindy F, Eischen CM, Randle DH, et al. Myc signaling via the ARF tumor suppressor regulates p53-dependent apoptosis and immortalization. Genes Dev 1998;12:2424-33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Finch A, Prescott J, Shchors K, et al. Bcl-xL gain of function and p19 ARF loss of function cooperate oncogenically with Myc in vivo by distinct mechanisms. Cancer Cell 2006;10:113-20 [DOI] [PubMed] [Google Scholar]
  • 81. Eischen CM, Weber JD, Roussel MF, Sherr CJ, Cleveland JL. Disruption of the ARF-Mdm2-p53 tumor suppressor pathway in Myc-induced lymphomagenesis. Genes Dev 1999;13:2658-69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Schmitt CA, McCurrach ME, de Stanchina E, Wallace-Brodeur RR, Lowe SW. INK4a/ARF mutations accelerate lymphomagenesis and promote chemoresistance by disabling p53. Genes Dev 1999;13:2670-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Alt JR, Greiner TC, Cleveland JL, Eischen CM. Mdm2 haplo-insufficiency profoundly inhibits Myc-induced lymphomagenesis. EMBO J 2003;22:1442-50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van Lohuizen M. Bmi-1 collaborates with c-Myc in tumorigenesis by inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev 1999;13:2678-90 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Kim SS, Shago M, Kaustov L, et al. CUL7 is a novel antiapoptotic oncogene. Cancer Res 2007;67:9616-22 [DOI] [PubMed] [Google Scholar]
  • 86. Maestro R, Dei Tos AP, Hamamori Y, et al. Twist is a potential oncogene that inhibits apoptosis. Genes Dev 1999;13:2207-17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. van Lohuizen M, Verbeek S, Scheijen B, Wientjens E, van der Gulden H, Berns A. Identification of cooperating oncogenes in E mu-myc transgenic mice by provirus tagging. Cell 1991;65:737-52 [DOI] [PubMed] [Google Scholar]
  • 88. Eischen CM, Packham G, Nip J, et al. Bcl-2 is an apoptotic target suppressed by both c-Myc and E2F-1. Oncogene 2001;20:6983-93 [DOI] [PubMed] [Google Scholar]
  • 89. Eischen CM, Woo D, Roussel MF, Cleveland JL. Apoptosis triggered by Myc-induced suppression of Bcl-XL or Bcl-2 is bypassed during lymphomagenesis. Mol Cell Biol 2001;21:5063-70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Maclean KH, Keller UB, Rodriguez-Galindo C, Nilsson JA, Cleveland JL. c-Myc augments [gamma] irradiation-induced apoptosis by suppressing Bcl-XL. Mol Cell Biol 2003;23:7256-70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Dansen TB, Whitfield J, Rostker F, Brown-Swigart L, Evan GI. Specific requirement for Bax, not Bak, in Myc-induced apoptosis and tumor suppression in vivo. J Biol Chem 2006;281:10890-5 [DOI] [PubMed] [Google Scholar]
  • 92. Juin P, Hunt A, Littlewood T, et al. c-Myc functionally cooperates with Bax to induce apoptosis. Mol Cell Biol 2002;22:6158-69 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Annis MG, Soucie EL, Dlugosz PJ, et al. Bax forms multispanning monomers that oligomerize to permeabilize membranes during apoptosis. [DOI] [PMC free article] [PubMed]
  • 94. Soucie EL, Annis MG, Sedivy J, et al. Myc potentiates apoptosis by stimulating Bax activity at the mitochondria. Mol Cell Biol 2001;21:4725-36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Abrams HD, Rohrschneider LR, Eisenman RN. Nuclear location of the putative transforming protein of avian myelocytomatosis virus. Cell 1982;29:427-39 [DOI] [PubMed] [Google Scholar]
  • 96. Donner P, Greiser-Wilke I, Moelling K. Nuclear localization and DNA binding of the transforming gene product of avian myelocytomatosis virus. Nature 1982;296:262-9 [DOI] [PubMed] [Google Scholar]
  • 97. Hann SR, Abrams HD, Rohrschneider LR, Eisenman RN. Proteins encoded by v-myc and c-myc oncogenes: identification and localization in acute leukemia virus transformants and bursal lymphoma cell lines. Cell 1983;34:789-98 [DOI] [PubMed] [Google Scholar]
  • 98. Murre C, McCaw PS, Baltimore D. A new DNA binding and dimerization motif in immunoglobulin enhancer binding, daughterless, MyoD, and myc proteins. Cell 1989;56:777-83 [DOI] [PubMed] [Google Scholar]
  • 99. Landschulz WH, Johnson PF, McKnight SL. The leucine zipper: a hypothetical structure common to a new class of DNA binding proteins. Science 1988;240:1759-64 [DOI] [PubMed] [Google Scholar]
  • 100. Facchini LM, Penn LZ. The molecular role of Myc in growth and transformation: recent discoveries lead to new insights. FASEB J 1998;12:633-51 [PubMed] [Google Scholar]
  • 101. Blackwell TK, Kretzner L, Blackwood EM, Eisenman RN, Weintraub H. Sequence-specific DNA binding by the c-Myc protein. Science 1990;250:1149-51 [DOI] [PubMed] [Google Scholar]
  • 102. Prendergast GC, Ziff EB. Methylation-sensitive sequence-specific DNA binding by the c-Myc basic region. Science 1991;251:186-9 [DOI] [PubMed] [Google Scholar]
  • 103. Blackwood EM, Eisenman RN. Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc. Science 1991;251:1211-7 [DOI] [PubMed] [Google Scholar]
  • 104. Ayer DE, Kretzner L, Eisenman RN. Mad: a heterodimeric partner for Max that antagonizes Myc transcriptional activity. Cell 1993;72:211-22 [DOI] [PubMed] [Google Scholar]
  • 105. Rottmann S, Luscher B. The Mad side of the Max network: antagonizing the function of Myc and more. Curr Top Microbiol Immunol 2006;302:63-122 [DOI] [PubMed] [Google Scholar]
  • 106. Kato GJ, Barrett J, Villa-Garcia M, Dang CV. An amino-terminal c-myc domain required for neoplastic transformation activates transcription. Mol Cell Biol 1990;10:5914-20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. McMahon SB, Van Buskirk HA, Dugan KA, Copeland TD, Cole MD. The novel ATM-related protein TRRAP is an essential cofactor for the c-Myc and E2F oncoproteins. Cell 1998;94:363-74 [DOI] [PubMed] [Google Scholar]
  • 108. McMahon SB, Wood MA, Cole MD. The essential cofactor TRRAP recruits the histone acetyltransferase hGCN5 to c-Myc. Mol Cell Biol 2000;20:556-62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Nishikura K, ar-Rushdi A, Erikson J, Watt R, Rovera G, Croce CM. Differential expression of the normal and of the translocated human c-myc oncogenes in B cells. Proc Natl Acad Sci U S A 1983;80:4822-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. ar-Rushdi A, Nishikura K, Erikson J, Watt R, Rovera G, Croce CM. Differential expression of the translocated and the untranslocated c-myc oncogene in Burkitt lymphoma. Science 1983;222:390-3 [DOI] [PubMed] [Google Scholar]
  • 111. Cleveland JL, Huleihel M, Bressler P, et al. Negative regulation of c-myc transcription involves myc family proteins. Oncogene Res 1988;3:357-75 [PubMed] [Google Scholar]
  • 112. Penn LJ, Brooks MW, Laufer EM, Land H. Negative autoregulation of c-myc transcription. EMBO J 1990;9:1113-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Stone J, de Lange T, Ramsay G, et al. Definition of regions in human c-myc that are involved in transformation and nuclear localization. Mol Cell Biol 1987;7:1697-709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Penn LJ, Brooks MW, Laufer EM, et al. Domains of human c-myc protein required for autosuppression and cooperation with ras oncogenes are overlapping. Mol Cell Biol 1990;10:4961-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Li LH, Nerlov C, Prendergast G, MacGregor D, Ziff EB. c-Myc represses transcription in vivo by a novel mechanism dependent on the initiator element and Myc box II. EMBO J 1994;13:4070-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Schneider A, Peukert K, Eilers M, Hanel F. Association of Myc with the zinc-finger protein Miz-1 defines a novel pathway for gene regulation by Myc. Curr Top Microbiol Immunol 1997;224:137-46 [DOI] [PubMed] [Google Scholar]
  • 117. Peukert K, Staller P, Schneider A, Carmichael G, Hanel F, Eilers M. An alternative pathway for gene regulation by Myc. EMBO J 1997;16:5672-86 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Mao DY, Watson JD, Yan PS, et al. Analysis of Myc bound loci identified by CpG island arrays shows that Max is essential for Myc-dependent repression. Curr Biol 2003;13:882-6 [DOI] [PubMed] [Google Scholar]
  • 119. Kleine-Kohlbrecher D, Adhikary S, Eilers M. Mechanisms of transcriptional repression by Myc. Curr Top Microbiol Immunol 2006;302:51-62 [DOI] [PubMed] [Google Scholar]
  • 120. Wagner AJ, Meyers C, Laimins LA, Hay N. c-Myc induces the expression and activity of ornithine decarboxylase. Cell Growth Differ 1993;4:879-83 [PubMed] [Google Scholar]
  • 121. Bello-Fernandez C, Packham G, Cleveland JL. The ornithine decarboxylase gene is a transcriptional target of c-Myc. Proc Natl Acad Sci U S A 1993;90:7804-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Mateyak MK, Obaya AJ, Adachi S, Sedivy JM. Phenotypes of c-Myc-deficient rat fibroblasts isolated by targeted homologous recombination. Cell Growth Differ 1997;8:1039-48 [PubMed] [Google Scholar]
  • 123. Bush A, Mateyak M, Dugan K, et al. c-myc null cells misregulate cad and gadd45 but not other proposed c-Myc targets. Genes Dev 1998;12:3797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Marhin WW, Chen S, Facchini LM, Fornace AJ, Penn LZ. Myc represses the growth arrest gene gadd45. Oncogene 1997;14:2825-34 [DOI] [PubMed] [Google Scholar]
  • 125. Miltenberger RJ, Sukow KA, Farnham PJ. An E-box-mediated increase in cad transcription at the G1/S-phase boundary is suppressed by inhibitory c-Myc mutants. Mol Cell Biol 1995;15:2527-35 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Watson JD, Oster SK, Shago M, Khosravi F, Penn LZ. Identifying genes regulated in a Myc-dependent manner. J Biol Chem 2002;277:36921-30 [DOI] [PubMed] [Google Scholar]
  • 127. Dang CV, O’Donnell KA, Zeller KI, Nguyen T, Osthus RC, Li F. The c-Myc target gene network. Semin Cancer Biol 2006;16:253-64 [DOI] [PubMed] [Google Scholar]
  • 128. Patel JH, Loboda AP, Showe MK, Showe LC, McMahon SB. Analysis of genomic targets reveals complex functions of MYC. Nat Rev Cancer 2004;4:562-8 [DOI] [PubMed] [Google Scholar]
  • 129. Knoepfler PS. Myc goes global: new tricks for an old oncogene. Cancer Res 2007;67:5061-3 [DOI] [PubMed] [Google Scholar]
  • 130. McMahon SB. Emerging concepts in the analysis of transcriptional targets of the MYC oncoprotein: are the targets targetable? Genes & Cancer 2010;1:560-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Bui TV, Mendell JT. Myc: maestro of microRNAs. Genes & Cancer 2010;1:568-75 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Dang CV. c-Myc target genes involved in cell growth, apoptosis, and metabolism. Mol Cell Biol 1999;19:1-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Eilers M, Eisenman RN. Myc’s broad reach. Genes Dev 2008;22:2755-66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Schuhmacher M. Control of cell growth by c-Myc in the absence of cell division. Curr Biol 1999;9:1255-66 [DOI] [PubMed] [Google Scholar]
  • 135. Iritani BM, Eisenman RN. c-Myc enhances protein synthesis and cell size during B lymphocyte development. Proc Natl Acad Sci U S A 1999;96:13180-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Dang CV, Le A, Gao P. MYC-induced cancer cell energy metabolism and therapeutic opportunities. Clin Cancer Res 2009;15:6479-83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. van Riggelen J, Yetil A, Felsher DW. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat Rev Cancer 2010;10:301-9 [DOI] [PubMed] [Google Scholar]
  • 138. Mai S, Fluri M, Siwarski D, Huppi K. Genomic instability in MycER-activated Rat1A-MycER cells. Chromosome Res 1996;4:365-71 [DOI] [PubMed] [Google Scholar]
  • 139. Li Q, Dang CV. c-Myc overexpression uncouples DNA replication from mitosis. Mol Cell Biol 1999;19:5339-51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Felsher DW, Bishop JM. Transient excess of MYC activity can elicit genomic instability and tumorigenesis. Proc Natl Acad Sci U S A 1999;96:3940-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Yin XY, Grove L, Datta NS, Long MW, Prochownik EV. C-myc overexpression and p53 loss cooperate to promote genomic instability. Oncogene 1999;18:1177-84 [DOI] [PubMed] [Google Scholar]
  • 142. Prochownik EV. c-Myc: linking transformation and genomic instability. Curr Mol Med 2008;8:446-58 [DOI] [PubMed] [Google Scholar]
  • 143. Prochownik EV, Li Y. The ever expanding role for c-Myc in promoting genomic instability. Cell Cycle 2007;6:1024-9 [DOI] [PubMed] [Google Scholar]
  • 144. Ngo CV, Gee M, Akhtar N, et al. An in vivo function for the transforming Myc protein: elicitation of the angiogenic phenotype. Cell Growth Differ 2000;11:201-10 [PMC free article] [PubMed] [Google Scholar]
  • 145. Dews M. Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat Genet 2006;38:1060-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Watnick RS, Cheng YN, Rangarajan A, Ince TA, Weinberg RA. Ras modulates Myc activity to repress thrombospondin-1 expression and increase tumor angiogenesis. Cancer Cell 2003;3:219-31 [DOI] [PubMed] [Google Scholar]
  • 147. Pelengaris S, Khan M, Evan GI. Suppression of Myc-induced apoptosis in [beta] cells exposes multiple oncogenic properties of Myc and triggers carcinogenic progression. Cell 2002;109:321-4 [DOI] [PubMed] [Google Scholar]
  • 148. Soucek L, Evan G. Myc: Is this the oncogene from hell? Cancer Cell 2002;1:406-8 [DOI] [PubMed] [Google Scholar]
  • 149. Dang CV. Rethinking the Warburg effect with Myc micromanaging glutamine metabolism. Cancer Res 2010;70:859-62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CB. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab 2008;7:11-20 [DOI] [PubMed] [Google Scholar]
  • 151. Wise DR, DeBerardinis RJ, Mancuso AS, et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc Natl Acad Sci U S A 2008;105:18782-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Drayton S, Rowe J, Jones R, et al. Tumor suppressor p16INK4a determines sensitivity of human cells to transformation by cooperating cellular oncogenes. Cancer Cell 2003;4:301-10 [DOI] [PubMed] [Google Scholar]
  • 153. Campaner S, Doni M, Hydbring P, et al. Cdk2 suppresses cellular senescence induced by the c-myc oncogene. Nat Cell Biol 2010;12:54-9 [DOI] [PubMed] [Google Scholar]
  • 154. Hydbring P, Larsson LG. Cdk2: a key regulator of the senescence control function of Myc. Aging (Albany NY) 2010;2:244-50 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. van Riggelen J, Felsher DW. Myc and a Cdk2 senescence switch. Nat Cell Biol 2010;12:7-9 [DOI] [PubMed] [Google Scholar]
  • 156. Grandori C, Wu KJ, Fernandez P, et al. Werner syndrome protein limits MYC-induced cellular senescence. Genes Dev 2003;17:1569-74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Wilson A, Murphy MJ, Oskarsson T, et al. c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Genes Dev 2004;18:2747-63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Knoepfler PS, Cheng PF, Eisenman RN. N-myc is essential during neurogenesis for the rapid expansion of progenitor cell populations and the inhibition of neuronal differentiation. Genes Dev 2002;16:2699-712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature 2007;448:313-7 [DOI] [PubMed] [Google Scholar]
  • 160. Wernig M, Meissner A, Foreman R, et al. In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 2007;448:318-24 [DOI] [PubMed] [Google Scholar]
  • 161. Nakagawa M. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nature Biotechnol 2008;26:101. [DOI] [PubMed] [Google Scholar]
  • 162. Arvanitis C, Felsher DW. Conditional transgenic models define how MYC initiates and maintains tumorigenesis. Semin Cancer Biol 2006;16:313-7 [DOI] [PubMed] [Google Scholar]
  • 163. Soucek L, Whitfield J, Martins CP, et al. Modelling Myc inhibition as a cancer therapy. Nature 2008;455:679-83 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Soucek L, Jucker R, Panacchia L, Ricordy R, Tato F, Nasi S. Omomyc: a potential Myc dominant negative, enhances Myc-induced apoptosis. Cancer Res 2002;62:3507-10 [PubMed] [Google Scholar]
  • 165. Albihn A, Johnsen JI, Henriksson MA. MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res 2010;107:163-224 [DOI] [PubMed] [Google Scholar]
  • 166. Ponzielli R, Katz S, Barsyte-Lovejoy D, Penn LZ. Cancer therapeutics: targeting the dark side of Myc. Eur J Cancer 2005;41:2485-501 [DOI] [PubMed] [Google Scholar]
  • 167. Frenzel A, Lovén J, Henriksson MA. Targeting MYC-regulated miRNAs to combat cancer. Genes & Cancer 2010;1:660-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Prochownik EV, Vogt PK. Therapeutic targeting of Myc. Genes & Cancer 2010;1:650-9 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Genes & Cancer are provided here courtesy of Impact Journals, LLC

RESOURCES