Elsevier

Gene

Volume 277, Issues 1–2, 17 October 2001, Pages 1-14
Gene

Review
Function and regulation of the transcription factors of the Myc/Max/Mad network

https://doi.org/10.1016/S0378-1119(01)00697-7Get rights and content

Abstract

The members of the Myc/Max/Mad network function as transcriptional regulators. Substantial evidence has been accumulated over the last years that support the model that Myc/Max/Mad proteins affect different aspects of cell behavior, including proliferation, differentiation, and apoptosis, by modulating distinct target genes. The unbalanced expression of these genes, e.g. in response to deregulated Myc expression, is most likely an important aspect of Myc's ability to stimulate tumor formation. Myc and Mad proteins affect target gene expression by recruiting chromatin remodeling activities. In particular Myc interacts with a SWI/SNF-like complex that may contain ATPase activity. In addition Myc binds to TRRAP complexes that possess histone acetyl transferase activity. Mad proteins, that antagonize Myc function, recruit an mSin3 repressor complex with histone deacetylase activity. Thus the antagonism of Myc and Mad proteins is explained at the molecular level by the recruitment of opposing chromatin remodeling activities.

Introduction

The broad and lasting interest in myc genes and Myc proteins is based on the realization that they regulate decisively various aspects of cell behavior. Foremost is the large body of information that has been accumulated over the last twenty years which demonstrates a strong involvement of myc in tumorigenesis (Marcu et al., 1992; Henriksson and Lüscher, 1996; Nesbit et al., 1999). Soon after the initial identification of v-myc in several transforming chicken retroviruses, genetic alterations of myc genes were found in many different human malignancies, e.g. translocations of the c-myc gene in Burkitt's lymphoma and amplification of the N-myc gene in neuroblastoma. This results in general in a deregulated, enhanced expression of myc genes, a condition found in many if not most human tumors. In addition studies in animal models and in tissue culture systems have provided compelling evidence for a tumor promoting activity of Myc proteins (Morgenbesser and DePinho, 1994; Pelengaris et al., 2000). This appears to be the result of Myc's ability to stimulate cell proliferation and at the same time to inhibit cells to enter a resting state or to terminally differentiate. Thus a consequence of the constitutive presence of Myc is an increase in the respective cellular compartment which is thought to provide the base for additional genetic alterations that cooperate with Myc in tumor formation (Berns, 1991; Adams and Cory, 1992). Recently a more direct role of Myc in transforming cells has also been suggested. This is based on findings that overexpression of Myc causes genomic destabilization potentially due to its ability to induce endoreplication (Mai et al., 1999; Taylor and Mai, 1998; Felsher and Bishop, 1999; Felsher et al., 2000). Knock-out experiments provide further strong evidence for a critical role of Myc for normal cell behavior (Stanton et al., 1992; Charron et al., 1992; Davis et al., 1993; Moreno de Alboran et al., 2001). In addition Myc proteins can stimulate apoptosis, a finding that on first view does not seem to be compatible with the other functions mentioned above (Evan et al., 1992; Thompson, 1998; Prendergast, 1999). However it is now thought that the induction of apoptosis by Myc is part of a safeguard mechanism that helps restrict the dominant action of Myc proteins on cell proliferation. In support tumor cells with elevated Myc levels are frequently defective in apoptotic pathways. Another activity of Myc that has been demonstrated recently is the ability to induce cell growth independent of cell cycle progression in some systems (Johnston et al., 1999; Iritani and Eisenman, 1999; Schuhmacher et al., 1999). Together these findings define Myc as a highly versatile factor influencing many aspects of normal cell behavior.

For many years it was unclear how Myc proteins achieve the diverse biological effects summarized above. From various studies, including the nuclear localization of these proteins, it was inferred that Myc could play roles in nuclear architecture, replication, splicing, and/or transcription (Lüscher and Eisenman, 1990). A hypothesis supporting the latter was formulated towards the end of the 1980s mainly on the basis of sequence homologies to other transcription factors. The motif found in common is referred to as the basic region/helix-loop-helix/leucine zipper (bHLHZip) domain and functions as a DNA binding and protein-protein interaction domain (Lüscher and Larsson, 1999). A break-through observation in defining a role of Myc in transcription was the identification of the bHLHZip protein Max as a Myc dimerization partner 10 years ago (Blackwood and Eisenman, 1991; Prendergast et al., 1991; Blackwood et al., 1992). Max is the essential heterodimerization partner of Myc proteins for various biological activities, including transformation, apoptosis, and transcriptional activation (Amati et al., 1992, Amati et al., 1993a, Amati et al., 1993b). In agreement with these findings Max is essential for normal mouse development (Shen-Li et al., 2000). However whether Max is required for all molecular functions of Myc is presently not known. Nevertheless the identification of Max and the finding that Myc/Max heterodimers bind specifically to E box DNA sequences with the consensus core 5′-CACGTG and through such elements activate the expression of reporter genes supported the hypothesis that Myc functions as a transcriptional regulator (Dang, 1999; Grandori et al., 2000). In addition a transactivation domain (TAD) was identified near the N-terminus of Myc (Facchini and Penn, 1998; Dang, 1999). Both the bHLHZip and the TAD are important for Myc function in the control of cell behavior. Together these findings provide support for a role of Myc in gene transcription. Other activities, however, cannot be excluded.

How can Myc affect so profoundly different aspects of cell behavior? To answer this question it will be important to define the genes that are targeted by Myc and to understand how the expression and function of Myc is regulated. Of particular interest is the question how Myc causes cell transformation in view of identifying targets that might be accessible to therapeutic strategies. During the early days of the search for Myc target genes it was hoped, somewhat naively from toady's point of view, that one could identify ‘the Myc target gene for transformation’. It seems more realistic now that Myc will modulate the expression of many genes to control cell behavior. A continuously increasing number of target genes are being proposed some of which are thought to be relevant for transformation (Grandori and Eisenman, 1997; Facchini and Penn, 1998; Boyd and Farnham, 1999). In particular the use of DNA microarray technology has made it possible to probe a large number of genes and to define a set of potential target genes that reflect Myc's broad role in the control of cell behavior (Coller et al., 2000; Guo et al., 2000; O'Hagan et al., 2000b; Schuhmacher et al., 2001). The targets include genes involved in the control of the cell cycle, protein and DNA biosynthesis, cell growth, cell adhesion, apoptosis, and immortality (Grandori et al., 2000). It will now be challenging to sort out the hierarchy of the importance of these genes for specific Myc functions, particularly for transformation.

The regulation of Myc expression and function is still far from being understood. In particular the expression of myc genes appears highly complicated. While a large number of transcription factors have been defined that can regulate the c-myc promoter, no unifying concept exists that explains myc expression during the cell cycle and during the transitions from resting to cycling and from cycling to differentiating cells (Spencer and Groudine, 1991; Marcu et al., 1997). Recent findings however provide some insight into c-myc regulation during the transition from G0 into G1 (Nasi et al., 2001). The tyrosine kinase Src has been implicated in mediating growth factor signal-induced c-myc expression (Barone and Courtneidge, 1995; Blake et al., 2000). Src appears to signal through the Rho GTPases rather than Ras or mitogen-activated protein kinases to activate the c-myc promoter (Chiariello et al., 2001). Although the transcription factor targets of Src/Rho signaling remain undefined, these findings provide a starting point to unravel growth factor-dependent c-myc expression.

Regarding the regulation of Myc proteins, recent progress has been made in understanding the signals that impinge on Myc. In particular phosphorylation has been shown to affect the stability of Myc, a protein with a short half life in the order of 20–30 min. Furthermore numerous Myc interaction partners have been identified, the functions of some support a role of Myc in the regulation of chromatin structure (Dang, 1999; Grandori et al., 2000; Amati et al., 2001). One task for the coming years will be to determine the role of these different proteins for specific aspects of Myc function and to connect these interactions to signaling pathways. In addition the essential Myc dimerization partner Max binds also to several other bHLHZip factors, forming what is referred to as the Myc/Max/Mad network of transcriptional regulators (Fig. 1). To understand the biological functions of Myc proteins requires also that the other components of this network are studied and their regulation and function determined since they impinge in one way or another on Myc function.

Numerous excellent reviews have been published in recent years that describe various aspects of the function and the biology of the Myc/Max/Mad network. Several of these reviews are cited to summarize work that can not be discussed in detail here. This review concentrates on novel aspects regarding the molecular functions and regulations of Myc and its network partners in gene transcription and the control of cell proliferation.

Section snippets

The Myc/Max/Mad network: components and their basic functions

The identification of Max as binding partner of different Myc proteins, including c-Myc, N-Myc, and L-Myc, initiated a search for additional bHLHZip dimerization partners. This led to the description of the Mad proteins Mad1, Mxi1, Mad3, and Mad4, as interaction partners of Max and together these factors define the Myc/Max/Mad network (Fig. 1) (Henriksson and Lüscher, 1996; Grandori et al., 2000). Later two additional Max partners, Mnt and Mga, were found (Hurlin et al., 1997, 1999). Recently

The Myc–Mad antagonism: opposing regulation of chromatin structure and gene transcription

All the studies that have been reported today provide evidence for a functional antagonism of Myc and Mad proteins (Fig. 3). This has been proposed first from findings showing that Mad proteins are expressed preferentially in non-proliferating cells as opposed to Myc proteins which are present almost exclusively in proliferating cells. Furthermore Mad proteins inhibit reporter genes that are activated by Myc, interfere with transformation of rat embryo fibroblasts, block cell growth, and

Myc and gene repression through initiator elements

While evidence has steadily accumulated over the last years that Myc can function as transcriptional activator, the suggestion that Myc can also function as a transcriptional repressor has been ill-defined at the molecular level until recently (Facchini and Penn, 1998; Claassen and Hann, 1999; Grandori et al., 2000; Amati et al., 2001). A main problem has been the difficulty in identifying response elements that mediate Myc repression and in defining the mode of Myc function on such elements.

Myc and cell cycle control

The products of a substantial number of the proposed Myc target genes are involved in the control of cell proliferation, i.e. in the regulation of cell-matrix interaction, of protein and DNA synthesis and of the transition from G1 into S phase of the cell cycle (Coller et al., 2000; Guo et al., 2000; O'Hagan et al., 2000b; Schuhmacher et al., 2001). These genes and their products provide a mechanistic base to evaluate Myc's role in stimulating cell proliferation, particularly the G1-S

Myc and signal transduction

The conclusions that in normal cells the expression of Myc is highly regulated and that deregulated expression of Myc is important for tumor progression are well supported by experimental evidence. The regulation of Myc at the posttranslational level has become more transparent over the last several years. In particular regulation by phosphorylation and protein stability have obtained considerable interest, also in view of their relevance for tumorigenesis. Nevertheless some of the findings

Outlook

Recent years have brought to light novel aspects of Myc function and regulation. In particular a number of new interaction partners have been identified that suggest an important role of Myc in the control of chromatin remodeling. Both SWI/SNF and TRRAP complexes can be recruited by Myc, however, the order of events and many of the molecular details remain to be determined. As one example it is unclear which HAT activity is recruited to mediate the acetylation of core histones of Myc responsive

Acknowledgements

I apologize for the omission of many important and relevant papers due to space limitations. I thank many colleagues, in particular M. Eilers, L.-G. Larsson, and J. Lüscher-Firzlaff, and members of my laboratory for many helpful discussions. The work in my laboratory is supported by the Deutsche Forschungsgemeinschaft and by the Fonds der Chemischen Industrie.

References (162)

  • E.M. Blackwood et al.

    Myc and Max function as a nucleoprotein complex

    Curr. Opin. Genet. Dev.

    (1992)
  • L. Chen et al.

    Ubiquitylation and destruction of endogenous c-mycS by the proteasome: are myc boxes dispensable?

    Arch. Biochem. Biophys.

    (2000)
  • P. Cheung et al.

    Signaling to chromatin through histone modifications

    Cell

    (2000)
  • S.V. Ekholm et al.

    Regulation of G(1) cyclin-dependent kinases in the mammalian cell cycle

    Curr. Opin. Cell Biol.

    (2000)
  • G.I. Evan et al.

    Induction of apoptosis in fibroblasts by c-myc protein

    Cell

    (1992)
  • C.J. Fry et al.

    Chromatin remodeling enzymes: who's on first?

    Curr. Biol.

    (2001)
  • J.A. Goodrich et al.

    Contacts in context: promoter specificity and macromolecular interactions in transcription

    Cell

    (1996)
  • C. Grandori et al.

    Myc target genes

    Trends Biochem. Sci.

    (1997)
  • P.A. Grant et al.

    The ATM-related cofactor Tra1 is a component of the purified SAGA complex

    Mol. Cell

    (1998)
  • C.A. Hassig et al.

    Histone deacetylase activity is required for full transcriptional repression by mSin3A

    Cell

    (1997)
  • M. Henriksson et al.

    Proteins of the Myc network: essential regulators of cell growth and differentiation

    Adv. Cancer Res.

    (1996)
  • T. Ikura et al.

    Involvement of the TIP60 histone acetylase complex in DNA repair and apoptosis

    Cell

    (2000)
  • L.A. Johnston et al.

    Drosophila Myc regulates cellular growth during development

    Cell

    (1999)
  • J.T. Kadonaga

    Eukaryotic transcription: an interlaced network of transcription factors and chromatin-modifying machines

    Cell

    (1998)
  • A. Kiermaier et al.

    Transcriptional control: calling in histone deacetylase

    Curr. Biol.

    (1997)
  • P.S. Knoepfler et al.

    Sin meets NuRD and other tails of repression

    Cell

    (1999)
  • R.D. Kornberg et al.

    Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome

    Cell

    (1999)
  • C.D. Laherty et al.

    Histone deacetylases associated with the mSin3 corepressor mediate Mad transcriptional repression

    Cell

    (1997)
  • S. Mai et al.

    Chromosomal and extrachromosomal instability of the cyclin D2 gene is induced by Myc overexpression

    Neoplasia

    (1999)
  • J. Massague et al.

    TGFbeta signaling in growth control, cancer, and heritable disorders

    Cell

    (2000)
  • S.B. McMahon et al.

    The novel ATM-related protein TRRAP is an essential cofactor for the c- Myc and E2F oncoproteins

    Cell

    (1998)
  • J.M. Adams et al.

    Oncogene co-operation in leukaemogenesis

    Cancer Surv.

    (1992)
  • T. Albert et al.

    Ongoing mutations in the N-terminal domain of c-Myc affect transactivation in Burkitt's lymphoma cell lines

    Oncogene

    (1994)
  • L. Alland et al.

    Role for N-CoR and histone deacetylase in Sin3-mediated transcriptional repression

    Nature

    (1997)
  • B. Amati

    Integrating Myc and TGF-beta signalling in cell-cycle control

    Nat. Cell Biol.

    (2001)
  • B. Amati et al.

    Transcriptional activation by the human c-Myc oncoprotein in yeast requires interaction with Max

    Nature

    (1992)
  • B. Amati et al.

    The c-Myc protein induces cell cycle progression and apoptosis through dimerization with Max

    EMBO J.

    (1993)
  • P. Antonson et al.

    Myc inhibits CCAAT/enhancer-binding protein alpha-gene expression in HIB-1B hibernoma cells through interactions with the core promoter region

    Eur. J. Biochem.

    (1995)
  • D.E. Ayer et al.

    A switch from Myc:Max to Mad:Max heterocomplexes accompanies monocyte/macrophage differentiation

    Genes Dev.

    (1993)
  • M.V. Barone et al.

    Myc but not Fos rescue of PDGF signalling block caused by kinase- inactive Src

    Nature

    (1995)
  • T.A. Baudino et al.

    The Max network gone mad

    Mol. Cell Biol.

    (2001)
  • A. Berns

    Tumorigenesis in transgenic mice: identification and characterization of synergizing oncogenes

    J. Cell Biochem.

    (1991)
  • K. Bhatia et al.

    Point mutations in the c-Myc transactivation domain are common in Burkitt's lymphomas and mouse plasmacytomas

    Nat. Genet.

    (1993)
  • A.N. Billin et al.

    MondoA, a novel basic helix-loop-helix-leucine zipper transcriptional activator that constitutes a positive branch of a max-like network

    Mol. Cell Biol.

    (2000)
  • E.M. Blackwood et al.

    Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc

    Science

    (1991)
  • R.A. Blake et al.

    SU6656, a selective src family kinase inhibitor, used to probe growth factor signaling

    Mol. Cell Biol.

    (2000)
  • I. Blomberg et al.

    Ectopic expression of Cdc25A accelerates the G(1)/S transition and leads to premature activation of cyclin E- and cyclin A-dependent kinases

    Mol. Cell Biol.

    (1999)
  • C. Bouchard et al.

    Direct induction of cyclin D2 by Myc contributes to cell cycle progression and sequestration of p27

    EMBO J.

    (1999)
  • C. Bouchard et al.

    Regulation of cyclin D2 gene expression by the Myc/Max/Mad network: Myc-dependent TRRAP recruitment and histone acetylation at the cyclin D2 promoter

    Genes Dev.

    (2001)
  • K.E. Boyd et al.

    Identification of target genes of oncogenic transcription factors

    Proc. Soc. Exp. Biol. Med.

    (1999)
  • Cited by (208)

    View all citing articles on Scopus
    View full text