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

Page created by Ian Greene
 
CONTINUE READING
Function and regulation of the transcription factors of the Myc/Max/Mad network
Gene 277 (2001) 1–14
                                                                                                                         www.elsevier.com/locate/gene
                                                                        Review

                Function and regulation of the transcription factors of the
                                Myc/Max/Mad network
                                                              Bernhard Lüscher*
       Abt. Biochemie und Molekularbiologie, Institut für Biochemie, Universitätsklinikum der RWTH, Pauwelstrasse 30, 52057 Aachen, Germany
                               Received 18 June 2001; received in revised form 17 August 2001; accepted 27 August 2001
                                                             Received by A.J. van Wijnen

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 expres-
sion, 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. q 2001 Published by Elsevier Science B.V.
Keywords: Cell cycle; Chromatin; Differentiation; Oncogene; Signaling; transformation

1. Introduction                                                                activity of Myc proteins (Morgenbesser and DePinho, 1994;
                                                                               Pelengaris et al., 2000). This appears to be the result of
   The broad and lasting interest in myc genes and Myc                         Myc’s ability to stimulate cell proliferation and at the
proteins is based on the realization that they regulate deci-                  same time to inhibit cells to enter a resting state or to term-
sively various aspects of cell behavior. Foremost is the large                 inally differentiate. Thus a consequence of the constitutive
body of information that has been accumulated over the last                    presence of Myc is an increase in the respective cellular
twenty years which demonstrates a strong involvement of                        compartment which is thought to provide the base for addi-
myc in tumorigenesis (Marcu et al., 1992; Henriksson and                       tional genetic alterations that cooperate with Myc in tumor
Lüscher, 1996; Nesbit et al., 1999). Soon after the initial                   formation (Berns, 1991; Adams and Cory, 1992). Recently a
identification of v-myc in several transforming chicken                        more direct role of Myc in transforming cells has also been
retroviruses, genetic alterations of myc genes were found                      suggested. This is based on findings that overexpression of
in many different human malignancies, e.g. translocations                      Myc causes genomic destabilization potentially due to its
of the c-myc gene in Burkitt’s lymphoma and amplification                      ability to induce endoreplication (Mai et al., 1999; Taylor
of the N-myc gene in neuroblastoma. This results in general                    and Mai, 1998; Felsher and Bishop, 1999; Felsher et al.,
in a deregulated, enhanced expression of myc genes, a                          2000). Knock-out experiments provide further strong
condition found in many if not most human tumors. In addi-                     evidence for a critical role of Myc for normal cell behavior
tion studies in animal models and in tissue culture systems                    (Stanton et al., 1992; Charron et al., 1992; Davis et al., 1993;
have provided compelling evidence for a tumor promoting                        Moreno de Alboran et al., 2001). In addition Myc proteins
                                                                               can stimulate apoptosis, a finding that on first view does not
   Abbreviations: bHLHZip, basic region/helix4oop-helix/leucine zipper;        seem to be compatible with the other functions mentioned
CDK, cyclin-dependent kinase; ChIP, chromatin immunoprecipitation;             above (Evan et al., 1992; Thompson, 1998; Prendergast,
CKI, cyclin-dependent kinase inhibitor; GSK, glycogen synthase kinase;         1999). However it is now thought that the induction of
HAT, histone acetyl transferase; HDAC, histone deacetylase; INR, initiator     apoptosis by Myc is part of a safeguard mechanism that
element; MBII, Myc box II; RB, retinoblastoma tumor suppressor protein;
                                                                               helps restrict the dominant action of Myc proteins on cell
SID, mSin3 interaction domain; TAD, transactivation domain
 * Tel.: 149-241-808-8850; fax: 149-511-808-2427.                              proliferation. In support tumor cells with elevated Myc
    E-mail address: luescher@rwth-aachen.de (B. Lüscher).                     levels are frequently defective in apoptotic pathways.
0378-1119/01/$ - see front matter q 2001 Published by Elsevier Science B.V.
PII: S 0378-111 9(01)00697-7
2                                                 B. Lüscher / Gene 277 (2001) 1–14

Another activity of Myc that has been demonstrated recently           (Grandori and Eisenman, 1997; Facchini and Penn, 1998;
is the ability to induce cell growth independent of cell cycle        Boyd and Farnham, 1999). In particular the use of DNA
progression in some systems (Johnston et al., 1999; Iritani           microarray technology has made it possible to probe a
and Eisenman, 1999; Schuhmacher et al., 1999). Together               large number of genes and to define a set of potential target
these findings define Myc as a highly versatile factor influ-         genes that reflect Myc’s broad role in the control of cell
encing many aspects of normal cell behavior.                          behavior (Coller et al., 2000; Guo et al., 2000; O’Hagan et
   For many years it was unclear how Myc proteins achieve             al., 2000b; Schuhmacher et al., 2001). The targets include
the diverse biological effects summarized above. From                 genes involved in the control of the cell cycle, protein and
various studies, including the nuclear localization of these          DNA biosynthesis, cell growth, cell adhesion, apoptosis,
proteins, it was inferred that Myc could play roles in nuclear        and immortality (Grandori et al., 2000). It will now be chal-
architecture, replication, splicing, and/or transcription             lenging to sort out the hierarchy of the importance of these
(Lüscher and Eisenman, 1990). A hypothesis supporting                genes for specific Myc functions, particularly for transfor-
the latter was formulated towards the end of the 1980s                mation.
mainly on the basis of sequence homologies to other tran-                The regulation of Myc expression and function is still far
scription factors. The motif found in common is referred to           from being understood. In particular the expression of myc
as the basic region/helix-loop-helix/leucine zipper                   genes appears highly complicated. While a large number of
(bHLHZip) domain and functions as a DNA binding and                   transcription factors have been defined that can regulate the
protein-protein interaction domain (Lüscher and Larsson,             c-myc promoter, no unifying concept exists that explains
1999). A break-through observation in defining a role of              myc expression during the cell cycle and during the transi-
Myc in transcription was the identification of the bHLHZip            tions from resting to cycling and from cycling to differen-
protein Max as a Myc dimerization partner 10 years ago                tiating cells (Spencer and Groudine, 1991; Marcu et al.,
(Blackwood and Eisenman, 1991; Prendergast et al., 1991;              1997). Recent findings however provide some insight into
Blackwood et al., 1992). Max is the essential heterodimer-            c-myc regulation during the transition from G0 into G1
ization partner of Myc proteins for various biological activ-         (Nasi et al., 2001). The tyrosine kinase Src has been impli-
ities,    including      transformation,   apoptosis,     and         cated in mediating growth factor signal-induced c-myc
transcriptional activation (Amati et al., 1992, 1993a,b). In          expression (Barone and Courtneidge, 1995; Blake et al.,
agreement with these findings Max is essential for normal             2000). Src appears to signal through the Rho GTPases rather
mouse development (Shen-Li et al., 2000). However                     than Ras or mitogen-activated protein kinases to activate the
whether Max is required for all molecular functions of                c-myc promoter (Chiariello et al., 2001). Although the tran-
Myc is presently not known. Nevertheless the identification           scription factor targets of Src/Rho signaling remain unde-
of Max and the finding that Myc/Max heterodimers bind                 fined, these findings provide a starting point to unravel
specifically to E box DNA sequences with the consensus                growth factor-dependent c-myc expression.
core 5 0 -CACGTG and through such elements activate the                  Regarding the regulation of Myc proteins, recent progress
expression of reporter genes supported the hypothesis that            has been made in understanding the signals that impinge on
Myc functions as a transcriptional regulator (Dang, 1999;             Myc. In particular phosphorylation has been shown to affect
Grandori et al., 2000). In addition a transactivation domain          the stability of Myc, a protein with a short half life in the
(TAD) was identified near the N-terminus of Myc (Facchini             order of 20–30 min. Furthermore numerous Myc interaction
and Penn, 1998; Dang, 1999). Both the bHLHZip and the                 partners have been identified, the functions of some support
TAD are important for Myc function in the control of cell             a role of Myc in the regulation of chromatin structure (Dang,
behavior. Together these findings provide support for a role          1999; Grandori et al., 2000; Amati et al., 2001). One task for
of Myc in gene transcription. Other activities, however,              the coming years will be to determine the role of these
cannot be excluded.                                                   different proteins for specific aspects of Myc function and
   How can Myc affect so profoundly different aspects of              to connect these interactions to signaling pathways. In addi-
cell behavior? To answer this question it will be important           tion the essential Myc dimerization partner Max binds also
to define the genes that are targeted by Myc and to under-            to several other bHLHZip factors, forming what is referred
stand how the expression and function of Myc is regulated.            to as the Myc/Max/Mad network of transcriptional regula-
Of particular interest is the question how Myc causes cell            tors (Fig. 1). To understand the biological functions of Myc
transformation in view of identifying targets that might be           proteins requires also that the other components of this
accessible to therapeutic strategies. During the early days of        network are studied and their regulation and function deter-
the search for Myc target genes it was hoped, somewhat                mined since they impinge in one way or another on Myc
naively from toady’s point of view, that one could identify           function.
‘the Myc target gene for transformation’. It seems more                  Numerous excellent reviews have been published in
realistic now that Myc will modulate the expression of                recent years that describe various aspects of the function
many genes to control cell behavior. A continuously                   and the biology of the Myc/Max/Mad network. Several of
increasing number of target genes are being proposed                  these reviews are cited to summarize work that can not be
some of which are thought to be relevant for transformation           discussed in detail here. This review concentrates on novel
B. Lüscher / Gene 277 (2001) 1–14                                                      3

Fig. 1. The Myc/Max/Mad network. The components of the Myc/Max/Mad network are indicated. Arrows define the known interactions between different
network members. The interactions are specified by the HLHZip dimerization domains. As far as characterized the members of the network function as
regulators of different aspects of cell behavior as indicated. More detailed information about individual components is given in the text.

aspects regarding the molecular functions and regulations of                further expand the network laterally. Together with the
Myc and its network partners in gene transcription and the                  many proteins found associated with the Myc/Max/Mad
control of cell proliferation.                                              network (see also Sections 3 and 4), an assemblage of
                                                                            factors is being defined that is rapidly growing not only in
                                                                            number of components but also in functional complexity.
2. The Myc/Max/Mad network: components and their                               Comparing the structure of the different network compo-
basic functions                                                             nents reveals similarities but also differences (Fig. 2). As
                                                                            expected from the identification of these factors as dimer-
   The identification of Max as binding partner of different                ization partners, all components possess a bHLHZip motif
Myc proteins, including c-Myc, N-Myc, and L-Myc,                            as DNA binding/dimerization interphase. Max and Mlx are
initiated a search for additional bHLHZip dimerization part-                capable of forming homodimers but none of the other
ners. This led to the description of the Mad proteins Mad1,                 components. All these dimers function as DNA binding
Mxi1, Mad3, and Mad4, as interaction partners of Max and                    competent complexes that recognize E box DNA sequences
together these factors define the Myc/Max/Mad network                       and have been suggested to function at least in part as tran-
(Fig. 1) (Henriksson and Lüscher, 1996; Grandori et al.,                   scriptional regulators (Dang, 1999; Lüscher and Larsson,
2000). Later two additional Max partners, Mnt and Mga,                      1999; Grandori et al., 2000). The importance of the
were found (Hurlin et al., 1997, 1999). Recently the identi-                bHLHZip domains of both Myc and Mad proteins is
fication of Mlx, a Max-like protein that can heterodimerize                 supported by mutational analysis, demonstrating an essen-
with Mad1, Mad4, and Mnt, but neither with the other Mad                    tial role of this domain for all functions tested. Interestingly
proteins nor with Myc or Max, has been reported (Billin et                  as far as analyzed, only Myc proteins, MondoA, and
al., 1999; Meroni et al., 2000). The most recent extensions                 WBSCR14 carry a TAD (Fig. 2) and Myc proteins and
of the network are the cloning of MondoA as an Mlx bind-                    MondoA can transactivate reporter genes through E box
ing partner (Billin et al., 2000) and the identification of                 elements (Dang, 1999; Billin et al., 2000; Cairo et al., 2001).
WBSCR14 that also interacts with Mlx (Cairo et al.,                            In addition to gene activation, Myc can also function as a
2001). The gene encoding WBSCR14 lies in the commonly                       repressor (Claassen and Hann, 1999; Grandori et al., 2000;
deleted region associated with Williams-Beuren syndrome                     Amati et al., 2001). This activity of Myc has remained,
(Cairo et al., 2001). Thus both Max and Mlx are the central                 however, largely unexplained at the molecular level until
components of network subdomains that are interconnected                    recently. One mechanism of Myc-dependent repression
by common interaction partners (Fig. 1). It is possible that                functions through blocking of the positive acting, initiator
not all members of the network have been defined yet and it                 (INR) binding factor Miz-1 by Myc (Section 4) (Staller et
will be of interest to detail additional components that may                al., 2001; Seoane et al., 2001). From many different studies
4                                                           B. Lüscher / Gene 277 (2001) 1–14

Fig. 2. Structure-function schematic of components of the Myc/Max/Mad network. The known functional domains of different network components are
summarized. Information regarding the expression and the loss of function phenotypes, as far as defined, is indicated. TAD: transactivation domain; I: Myc box
I; II: Myc box II; b: basic region; R: region involved in repression; HLH: helix-loop-helix domain; Zip: leucine zipper domain; SID: mSin3-interaction domain;
T-Domain: T-box DNA binding domain; TRD: transrepression domain; P: position of known phosphorylation sites. The numbers refer to amino acids of the
human proteins.

it appears that both repression and activation of transcription                   for numerous additional functional domains (Fig. 2). Inter-
are essential activities of Myc proteins (for a recent discus-                    estingly Mga carries two distinct DNA binding domains
sion see Amati et al., 2001).                                                     suggesting that it can regulate genes not only through E
    In contrast to Myc, Mad proteins and Mnt possess an                           box elements but also through sequences recognized by its
mSin3 interaction domain (SID) which mediates transcrip-                          T-box DNA binding motif (Tada and Smith, 2001). It is
tional repression (Kiermaier and Eilers, 1997; Schreiber-                         likely that further analysis of the different components of
Agus and DePinho, 1998; Knoepfler and Eisenman, 1999).                            the network will reveal additional new and unexpected find-
Besides bHLHZip, the SID is critical for the function of                          ings.
Mad proteins. This is also true for Mnt, however this protein
has most likely additional domains relevant for function.
The former is suggested by the findings that Mad proteins                         3. The Myc–Mad antagonism: opposing regulation of
with an impaired SID show little biological activity. In                          chromatin structure and gene transcription
support of the latter is that MntDSID gains transforming
activity in cooperation with an activated Ras oncoprotein,                           All the studies that have been reported today provide
whereas Mnt and Mad proteins with functional SIDs inter-                          evidence for a functional antagonism of Myc and Mad
fere with transformation (Grandori et al., 2000). Together                        proteins (Fig. 3). This has been proposed first from findings
these studies define the TAD of Myc proteins, the SID of                          showing that Mad proteins are expressed preferentially in
Mad proteins, and the bHLHZip of all components as essen-                         non-proliferating cells as opposed to Myc proteins which
tial domains for the function of the respective network                           are present almost exclusively in proliferating cells. Further-
members. However detailed structure-function analyses of                          more Mad proteins inhibit reporter genes that are activated
the proteins that have been identified more recently have not                     by Myc, interfere with transformation of rat embryo fibro-
been performed yet. This is certainly true for Mga which, as                      blasts, block cell growth, and prevent apoptosis (Henriksson
its name indicates, is a very large protein of just over 3000                     and Lüscher, 1996; Grandori et al., 2000). The findings
amino acids (Hurlin et al., 1999). This already distinguishes                     summarized above, which are based mainly on overexpres-
Mga from the other members of the Myc/Max/Mad network                             sion studies, are supported by the analysis of mad knock-out
which in general are rather small molecules and offers room                       and mad transgenic mice (Schreiber-Agus et al., 1998;
B. Lüscher / Gene 277 (2001) 1–14                                                              5

Fig. 3. Model for the control of promoter activity by Myc/Max/Mad network members. Distinct dimeric complexes, i.e. Myc/Max, Max/Max, and Mad/Max,
are thought to recruit different cofactors to promoters through specific interaction with E box DNA elements. Myc binds to a component of the SWI/SNF
complex that contains an ATPase activity involved in nucleosome remodeling. In addition Myc interact with TRRAP complexes that possess HAT activity
important for modulating the acetylation status of core histones and possibly other components associated with promoters, including transcription factors and
subunits of the PolII complex. Max has not been reported to bind transcriptional cofactors. Thus Max/Max homodimers will most likely not influence directly
promoter activity. Nevertheless Max/Max dimers are potentially able to compete for DNA binding with other E box binding factors. Mad proteins recruit an
mSin3-dependent repressor complex that is associated with HDAC activity. This complex will deacetylate core histones and possibly other acetylated proteins.
The results of binding of Myc/Max, Max/Max, or Mad/Max dimers to a promoter are summarized and a possible order of events is indicated. Myc/Max binding
and recruitment of SWI/SNF may lead to alterations of a promoter’s chromatin structure by modulating nucleosomes. This may make binding sites available
for other transcriptional regulators that may or may not cooperate with Myc in promoter modulation. The recruitment of a cofactor complex with HAT activity
will further enhance promoter accessibility and in addition also provide acetylation-dependent binding sites on histone tails. It is not clear whether SWI/SNF
and TRRAP complexes work on the same promoters and in which order. Myc interacts with components of the PolII complex and may therefore also be
involved in PolII recruitment. The binding of Mad/Max and recruitment of the mSin3 repressor complex will lead to histone deacetylation and increased
chromatin compaction. This is thought to result in decreased accessibility of transcription factors to their binding sites and of the PolII complex to the core
promoter region. Besides HDAC other components of the mSin3 complex contribute to repression. This suggests that other functions, e.g. an activity that
antagonizes SWI/SNF function, may be associated with this repressor. E box: refers to a DNA element that is recognized by Myc/Max/Mad network members;
RE: response element for transcriptional regulators; Ac: acetylation of histones; TF: transcription factor; HAT: histone acetyl transferase; HDAC; histone
deacetylase;

Foley et al., 1998; Queva et al., 1999, 2001). However, in                         activators are important for the efficient recruitment and
particular the knock-out studies have not provided the                             activation of the polymerase complex. In addition activators
strong phenotypes originally expected, most likely due to                          may regulate the processivity of the polymerase complex.
overlapping and redundant functions of Mad proteins in                             More recently it has become evident that these activities are
different cell types. Indeed in all cells analyzed more than                       most likely downstream of effects on the chromatin struc-
one Mad protein is expressed (Grandori et al., 2000).                              ture of gene loci and more specifically of promoter regions.
   Transcriptional regulators are thought to affect the tran-                      The structure of the chromatin appears to be a major deter-
scription of specific genes at several different levels (Good-                     minant whether promoters are accessible to the polymerase
rich et al., 1996; Struhl, 1996). Early findings suggested that                    complex and thus whether a gene can be transcribed or not
6                                                      B. Lüscher / Gene 277 (2001) 1–14

(Kadonaga, 1998; Kornberg and Lorch, 1999; Wolffe and                      activity, one containing GCN5/PCAF the other Tip60/NuA4
Hansen, 2001). Repressors of transcription may also affect                 (Vassilev et al., 1998; Grant et al., 1998; Ikura et al., 2000).
the polymerase complex but it is thought that their primary                Indeed Myc recruits GCN5 and HAT activity, most likely
effects are on compacting chromatin and thus making it                     through TRRAP (McMahon et al., 2000; Park et al., 2001).
inaccessible for positive acting factors as well as for poly-              A potential involvement of Tip60 is also indicated by the
merases. How is chromatin affected? Two fundamentally                      identification of Tip48 and Tip49, both are part of the Tip60
distinct mechanisms have emerged. One involves the activ-                  complex, as Myc interaction partners (Wood et al., 2000).
ity of molecules that possess ATPase activity and possibly                 TRRAP interacts with the TAD of Myc, more precisely it
helicase activity and affect the structure and location of                 requires a conserved element in the TAD, the so called Myc
nucleosomes (Kingston and Narlikar, 1999; Peterson and                     box II (MBII) (McMahon et al., 1998). MBII is a small
Workman, 2000; Aalfs and Kingston, 2000; Fry and Peter-                    element that is essential for all biological activities of
son, 2001). The second mechanism targets histones. These                   Myc proteins. Although a role of MBII in gene transcription
proteins can be modified by, e.g. phosphorylation, acetyla-                has been questioned (Xiao et al., 1998), the binding of this
tion and methylation, leading to a multitude of differentially             Myc domain to HAT complexes suggests that MBII is
modified histones. Distinct modification patterns, referred to             involved in the regulation of gene transcription. Indeed
as histone code, are thought to affect compaction of chro-                 recent studies using chromatin immunoprecipitation
matin and to provide docking sites for proteins. At least in               (ChIP) assays, in which transcription factors and their cofac-
some cases certain histone modifications have been linked                  tors are crosslinked in vivo to their DNA binding sites and
to specific functional states of promoters (Strahl and Allis,              subsequently the associated DNA is analyzed by gene-
2000; Cheung et al., 2000).                                                specific PCR, have demonstrated that Myc recruits
    Evidence is accumulating that the antagonism of Myc and                TRRAP activity in a MBII-dependent manner to target
Mad proteins at the molecular level works at least in part by              genes. This results in histone acetylation of responsive
affecting chromatin structure (Fig. 3). The first evidence that            promoters and subsequently in gene expression (Bouchard
Myc/Max/Mad network proteins may indeed function in                        et al., 2001; Frank et al., 2001). The apparent conflict in
chromatin remodeling came from the findings that Mad                       regard of a role of MBII in transcription reflects most likely
proteins recruit through the SID a repressor complex that                  the difference between the analysis of transiently transfected
contains histone deacetylase (HDAC) activity (Hassig et al.,               reporter genes and endogenous, chromosome-embedded
1997; Alland et al., 1997; Sommer et al., 1997; Laherty et                 genes (Amati et al., 2001). The latter are dependent for
al., 1997). Several lines of evidence support the notion that              expression on chromatin remodeling and histone modifica-
the recruitment of the repressor complex is essential for the              tion activities, while reporter genes have more relaxed
biological effects of Mad1 described above. Deletion of the                requirements. Together these findings define an important
SID impairs the activity of Mad proteins in transformation,                role for MBII in the control of gene transcription by Myc
transactivation, cell growth inhibition, and apoptosis                     and thus support the concept that regulation of gene tran-
(Grandori et al., 2000; Baudino and Cleveland, 2001).                      scription is an essential aspect of Myc biology.
Since the bHLHZip is also important for these functions,                      Early on after the identification of Mad1 it was suggested
it is likely that these are, at least in part, the result of specific      that Myc/Max and Mad/Max complexes form a molecular
target gene regulation. Although the widely expressed Mnt                  switch involved in the regulation of transitions between
also possesses a SID domain that binds mSin3 in vitro and                  different cell states, in which Myc/Max complexes are
its deletion affects Mnt function, it remains to be determined             exchanged by antagonizing Mad/Max complexes, and vice
whether in vivo the SID of Mnt serves to recruit an HDAC-                  versa, on responsive E box DNA elements (Ayer et al.,
containing repressor complex. It is possible that the inter-               1993; Ayer and Eisenman, 1993). In support of this model
action of Mnt with the mSin3 complex is regulated in an as                 Myc/Max and Mad/Max complexes are expressed mainly in
yet unknown manner. Since Mnt is, in contrast to Mad                       growing and in resting and differentiated cells, respectively.
proteins, a stable protein expressed ubiquitously such a                   More recently the exchange of Myc/Max to Mad/Max
regulation might be desirable to modulate its function.                    complexes could be observed on two responsive promoters.
    The findings that Mad proteins recruit a repressor                     Using ChIP assays it has been demonstrated that Myc/Max
complex with HDAC activity led to the suggestion that                      complexes bound to the promoters of hTERT and human
Myc proteins might recruit histone acetyl transferase                      cyclin D2 genes in exponentially growing HL-60 myelo-
(HAT) activity, thus providing a molecular explanation                     blast cells are replaced by Mad/Max complexes during
for the antagonism between Myc and Mad proteins. Indeed                    differentiation (Xu et al., 2001; Bouchard et al., 2001).
recent findings demonstrated that Myc interacts with a large               Furthermore this switch from Myc/Max to Mad/Max
protein, TRRAP, that appears to function as coactivator                    complexes on cyclin D2 is associated with recruitment of
(McMahon et al., 1998; Saleh et al., 1998). Like mSin3                     HDAC1, a decrease in histone acetylation, and a loss of
for Mad proteins, TRRAP can function as a platform for                     polymerase II binding to the cyclin D2 promoter (Bouchard
additional proteins involved in gene transcription. TRRAP                  et al., 2001). Thus the findings discussed above provide
is part of at least two large complexes that possess HAT                   evidence for and define one consequence of the proposed
B. Lüscher / Gene 277 (2001) 1–14                                                 7

switch, i.e. the alternate recruitment of HAT and HDAC                 Miz-1 depends on the retinoblastoma tumor suppressor, the
activity to target genes. This suggest that the distinct regula-       expression of the Ink4 cyclin-dependent kinase inhibitors
tion of histone acetylation in response to Myc/Max/Mad                 was analyzed. Miz-1 binds to the Ink4b INR and activates
network members represents one level of the functional                 the expression of this gene. Importantly Myc is also asso-
antagonism of Myc and Mad proteins.                                    ciated with the INR thus providing the first direct evidence
   While at present there is little evidence for additional            for the physical interaction of Myc with a negatively regu-
Mad-interacting proteins that are involved in the regulation           lated promoter. Several genes are repressed by Myc through
of gene transcription, several other factors implicated in the         core promoter and INR elements, including c-myc, p21 CIP1,
regulation of transcription have been identified that bind to          C/EBPa, p27, and H-ferritin (Antonson et al., 1995;
Myc. The C-terminal domain of Myc interacts with INI1/                 Facchini et al., 1997; Wu et al., 1999; Claassen and Hann,
hSNF5, a component of the multiprotein SWI/SNF                         2000; Yang et al., 2001) suggesting an involvement of Miz-
complex, that is involved in chromatin remodeling in an                1. Other genes, including gadd45 and pdgf-b receptor, are
ATP-dependent manner (Cheng et al., 1999; Kingston and                 repressed in an INR-independent manner (Marhin et al.,
Narlikar, 1999; Peterson and Workman, 2000). Interestingly             1997; Oster et al., 2000; Izumi et al., 2001). Repression of
INI1 has been suggested to function as a tumor suppressor              the pdgf-b receptor is mediated by NF-Y, a factor that is
and the two SWI2/SNF2 homologues, BRG1 and hBRM,                       inhibited by Myc (Izumi et al., 2001).
can induce cell cycle arrest (Versteege et al., 1998;                     How does Myc inhibit the activation of the Ink4b promo-
Muchardt and Yaniv, 1999). Thus it appears, somewhat                   ter by Miz-1? Miz-1 recruits the coactivator p300, an inter-
puzzling, that the proto-oncoprotein c-Myc interacts with              action that is competed for by Myc (Staller et al., 2001).
a complex that has growth inhibitory activity. Although it             Since the C-terminal HLHZip region of Myc is sufficient for
has been reported that recruitment of the SWI/SNF complex              this inhibition, one would expect that Myc brings a func-
is necessary for Myc transactivation function in reporter              tional TAD into Miz-1/Myc complex. However the Myc
gene assays, it is not yet clear what the role of this complex         TAD appears not to function under these conditions, either
is for the regulation of chromosome embedded target genes.             coactivators cannot be recruited efficiently or the Ink4a
It will now be of interest to determine the relative contribu-         promoter is not responsive. Another possibility is that addi-
tion of the different complexes, including TRRAP and SWI/              tional domains in Myc are relevant for repression, including
SNF complexes, for Myc-specific gene regulation.                       MBII and the region spanning amino acids 96–106 (region
                                                                       R in Fig. 2) as suggested previously (Philipp et al., 1994; Li
                                                                       et al., 1994; Lee et al., 1997). As indicated above it will be
4. Myc and gene repression through initiator elements                  important to define whether other Myc-repressed promoters
                                                                       are regulated by a Miz-1 dependent mechanism or whether
   While evidence has steadily accumulated over the last               Myc can regulate INR elements through additional means.
years that Myc can function as transcriptional activator,
the suggestion that Myc can also function as a transcrip-
tional repressor has been ill-defined at the molecular level           5. Myc and cell cycle control
until recently (Facchini and Penn, 1998; Claassen and Hann,
1999; Grandori et al., 2000; Amati et al., 2001). A main                  The products of a substantial number of the proposed
problem has been the difficulty in identifying response                Myc target genes are involved in the control of cell prolif-
elements that mediate Myc repression and in defining the               eration, i.e. in the regulation of cell-matrix interaction, of
mode of Myc function on such elements. Recent findings                 protein and DNA synthesis and of the transition from G1
however have shed light on Myc-mediated repression. Early              into S phase of the cell cycle (Coller et al., 2000; Guo et al.,
on it has been suggested that the initiator element (INR),             2000; O’Hagan et al., 2000b; Schuhmacher et al., 2001).
which is part of some core promoters, responds to Myc (Li              These genes and their products provide a mechanistic base
et al., 1994; Philipp et al., 1994; Mai and Martensson, 1995;          to evaluate Myc’s role in stimulating cell proliferation,
Lee et al., 1996). At least three DNA binding proteins that            particularly the G1-S transition and the inactivation of the
can bind to INR sequences, TFII-I, YY1, and Miz-1, are also            G1-S checkpoint (or restriction point) (Fig. 4). Overcoming
interacting with Myc, leading to the proposal that these               the latter is essential for cell proliferation. This requires the
factors mediate Myc repression (Peukert et al., 1997; Roy              inactivation of pocket proteins, in particular the retinoblas-
et al., 1993; Hariharan et al., 1991; Seto et al., 1991; Shri-         toma tumor suppressor protein (RB), and the activation of
vastava et al., 1993). While a role for TFII-I and YY1 in              the cyclin E/cyclin-dependent kinase (CDK) 2 complex
Myc-dependent repression has not been firmly established,              (Adams, 2001; Sherr and Roberts, 1999; Ekholm and
recent evidence suggests that Miz-1 mediates Myc repres-               Reed, 2000). Early on it was shown that activation of Myc
sion. Miz-1 binds to and activates transcription from INR              results in the activation of cyclin/CDK complexes (Jansen-
elements and inhibits cell cycle progression, both functions           Dürr et al., 1993; Steiner et al., 1995; Perez-Roger et al.,
are antagonized by Myc (Peukert et al., 1997; Staller et al.,          1997; Müller et al., 1997), leading to the suggestion that one
2001; Seoane et al., 2001). Since the cell cycle arrest by             important activity of Myc in regulating cell proliferation is
8                                                             B. Lüscher / Gene 277 (2001) 1–14

                                                                                     With the identification of Myc as a regulator of the
                                                                                  expression and activity of cyclin/CDKs, one focus of subse-
                                                                                  quent research was the analysis of the mechanism of this
                                                                                  regulation. While some components of cyclin/CDK
                                                                                  complexes are direct targets of the Myc/Max/Mad network,
                                                                                  including cyclin D2 and possibly cyclin D1 and CDK4
                                                                                  (Bouchard et al., 1999; Bouchard et al., 2001; Perez-
                                                                                  Roger et al., 1999; Hermeking et al., 2000; Coller et al.,
                                                                                  2000), indirect effects of Myc also contribute to the activa-
                                                                                  tion of cyclin/CDK. Together the identification of these
                                                                                  target genes will now allow to define in more detail how
                                                                                  Myc proteins affect the activity of different cyclin D/CDK
                                                                                  and cyclin E/CDK2 complexes (Fig. 4). Both these cyclin/
                                                                                  CDK complexes are required to enter S phase (Sherr and
                                                                                  Roberts, 1999; Ekholm and Reed, 2000). Cyclin D/CDK
                                                                                  complexes are built from several different subunits includ-
                                                                                  ing cyclin D1, D2, and D3 and the two catalytic subunits
                                                                                  CDK4 and CDK6. The activation of cyclin D1 and D2 by
                                                                                  Myc is of particular importance since their expression not
                                                                                  only can positively influence cyclin D/CDK complexes but
                                                                                  these two cyclins have also been shown to sequester p27 KIP1,
                                                                                  an inhibitor of cyclin E/CDK2 (Bouchard et al., 1999;
                                                                                  Bouchard et al., 2001; Perez-Roger et al., 1999; Coller et
                                                                                  al., 2000; Dey et al., 2000). Furthermore the Ink4b gene,
                                                                                  encoding the cyclin-dependent kinase inhibitor (CKI)
                                                                                  p15 INK4b, is repressed by Myc as summarized above (Staller
Fig. 4. Regulation of G1–S phase progression by Myc. Accumulating                 et al., 2001; Seoane et al., 2001). The activation of different
evidence suggests that Myc regulates several target genes whose products          Ink4 genes has been recognized as an important mechanism
are involved in controlling the transition from G1 into S. These interactions     to negatively regulate cell proliferation by many different
are summarized (see Sections 4 and 5 for details). Myc can activate the           agents (Vidal and Koff, 2000). In particular the Ink4b gene
genes encoding Id2, CDK4, Cyclin D2, Cul1, and Cdc25A. Id2 is a negative
                                                                                  is activated in response to TGFb, a function that is antag-
regulator of pocket proteins, including RB, and thus can stimulate progres-
sion through the restriction point. Induction of CDK4 and cyclin D2 stimu-        onized by Myc (Massague et al., 2000; Amati, 2001).
lates both cyclin D/CDK and cyclin E/CDK complexes. Cul1 is a                     p15 INK4b is an inhibitor of cyclin D/CDK4/6 function.
component of the SCF complex that is responsible for signal-dependent             Together these findings demonstrate that Myc can stimulate,
ubiquitination of p27 KIP1 which leads to its degradation. The phosphatase        through the regulation of Ink4b, cyclin D2, and CDK4,
Cdc25A activates CDK2 by removing inhibitory phosphates. Furthermore
                                                                                  cyclin D/CDK function.
Myc represses genes that encode cyclin/CDK inhibitors, including p15 INK4b,
p21 CIP1, and p27 KIP1. These Myc-dependent regulations result in the stimu-         The targets of cyclin D/CDK4/6 are pocket proteins. In
lation of cyclin D- and cyclin E-dependent kinase activities and in the           particular the inactivation of the RB protein, a frequently
inhibition of pocket proteins. Together these effects enhance S phase             mutated and by this inactivated tumor suppressor, is critical
progression and explain at least in part the S phase-inducing activity of         for the regulation of the G1-S checkpoint (Bartek et al.,
Myc upon deregulated expression.
                                                                                  1997; Reed, 1997). Besides directly impinging on the activ-
                                                                                  ity of cyclin D/CDK complexes, Myc has also been shown
stimulating the G1-S transition. This transition is also a key                    to stimulate the expression of Id2, which encodes an HLH
target during oncogenesis (Bartek et al., 1997; Reed, 1997).                      protein that inhibits RB function (Lasorella et al., 2000).
Most if not all tumor cells have mutations that result in                         This provides an additional mechanism to release E2F tran-
altered control of the G1-S checkpoint and thus facilitate                        scription factor activity, which is negatively regulated by
the entry into the DNA replication mode. Once cells have                          RB, and thus to promote S phase progression (Harbour and
progressed into S phase, the remainder of the cell cycle                          Dean, 2000). The findings that Myc can interfere at multiple
proceeds independent of the presence of growth factors.                           levels with the function of RB proteins and the frequent
Thus under limiting amounts of such factors, advancing                            mutations in RB and in the upstream regulatory components
beyond the G1-S checkpoint is critical for proliferation.                         in human tumors suggest that this is an important function of
Myc is sufficient to overcome the restriction point and to                        Myc in tumorigenesis. Both gene activation and repression
induce S phase in the absence of growth factors (Eilers et al.,                   are important for this activity of Myc.
1991). Therefore it is attractive to suggest that the activation                     The second cyclin/CDK activity that is required for enter-
of cyclin/CDK complexes is critical for Myc’s ability to                          ing S phase is cyclin E/CDK2. Although neither of the two
induce S phase.                                                                   genes appear to be direct targets of Myc, multiple ways have
B. Lüscher / Gene 277 (2001) 1–14                                                9

evolved how Myc can influence cyclin E/CDK2 activity.                  the central acidic domain and one near the basic region, are
The cdc25A gene has been proposed to be a Myc target                   phosphorylated by protein kinase CK2 (Lüscher et al.,
(Galaktionov et al., 1996). Activation of Cdc25A, a dual               1989). Little is known about the regulation and function
specificity phosphatase, stimulates cyclin E/CDK2 activity             of these phosphorylation sites. The third cluster is located
(Blomberg and Hoffmann, 1999; Sexl et al., 1999). In addi-             in the transactivation domain, more specifically at and near
tion several pathways target the cyclin E/CDK2 inhibitor               MBI (Seth et al., 1992, 1993; Pulverer et al., 1994; Henriks-
p27 KIP1. This CKI is an important regulator of cell prolifera-        son et al., 1993; Lutterbach and Hann, 1994). Of particular
tion that is induced when growth factor levels are insuffi-            importance are two sites, Thr-58 and Ser-62, because this
cient to sustain proliferation. Consistently p27 KIP1 is highly        region is a mutational hot spot (Bhatia et al., 1993, 1994;
expressed in resting cells and downregulated upon serum                Yano et al., 1993; Albert et al., 1994; Axelson et al., 1995).
induction (Sherr and Roberts, 1999). Importantly p27 KIP1              These mutations that are prevalent in Burkitt’s lymphomas,
is also repressed in response to Myc activation. First                 a tumor characterized by a translocated c-myc gene, affect
p27 KIP1 is sequestered by Myc-induced cyclin D2 and subse-            predominantly phosphorylation of Thr-58. This site is phos-
quently degraded in response to cyclin E/CDK2-dependent                phorylated in vitro by glycogen synthase kinase (GSK) 3
phosphorylation by a ubiquitin/proteasome dependent path-              dependent on phosphorylation of the adjacent Ser-62
way (Bouchard et al., 1999; Bouchard et al., 2001; Perez-              (Henriksson et al., 1993; Sears et al., 2000). This latter
Roger et al., 1999; Pagano et al., 1995). Also the latter              site is targeted by MAP kinases and by cyclin/CDKs (Seth
process is activated by Myc (Steiner et al., 1995). A recently         et al., 1992; Hoang et al., 1995; Pulverer et al., 1994; Lutter-
identified target gene of Myc is cul1 that is involved in              bach and Hann, 1994; Henriksson et al., 1993; J. Vervoorts
p27 KIP1 degradation (O’Hagan et al., 2000a). Last but not             and B. Lüscher, unpublished findings). For some time the
least Myc can also directly repress the expression of the              functional relevance of these two phosphorylation sites and
p27 KIP1 gene (Yang et al., 2001). These findings are consis-          the observed mutations in this region remained unresolved.
tent with the observation that immortal myc 2/2 cells have             Early on it was suggested that phosphorylation at Ser-62
increased p27 levels (Moreno de Alboran et al., 2001). A               stimulated the transcriptional potential of Myc (Seth et al.,
third CKI gene that is targeted by Myc is p21 CIP1 (Claassen           1992, 1993). However this finding is controversial (Henriks-
and Hann, 2000). This molecule inhibits cyclin E/CDK2 and              son et al., 1993; Lutterbach and Hann, 1994). In in vitro
is upregulated in response to the activation of the tumor              transformation assays small differences between wt c-Myc
suppressor p53 and in differentiating as well as aging cells           and Thr-58 and Ser-62 mutants were detected (Pulverer et
as part of an antiproliferative program (Sherr and Roberts,            al., 1994; Henriksson et al., 1993). Thus these findings did
1999).                                                                 not allow to make strong conclusions regarding the function
   Together these studies have defined the cyclin D and                of the N-terminal phosphorylation sites.
cyclin E kinase complexes, that are essential regulators of               However more recently, evidence has been provided indi-
the decision process whether or not a cell proceeds through            cating that these phosphorylation sites are regulating Myc
the G1-S checkpoint and into a new round of cell division,             stability. Myc is degraded by ubiquitin-mediated proteolysis
as targets of Myc. The identification of multiple Myc regu-            (Salghetti et al., 1999; Bahram et al., 2000; Gregory and
lated genes whose products impinge on these kinases                    Hann, 2000), a process that involves covalent attachment
provide strong support for the notion that these two                   of ubiquitin to target proteins and subsequent proteolysis by
cyclin/CDK complexes are critical targets for Myc’s func-              the proteasome (Varshavsky, 1997). In Myc the TAD deter-
tion in the regulation of cell proliferation.                          mines the short half life, in particular MBI and II have been
                                                                       implicated in degradation (Flinn et al., 1998; Salghetti et al.,
                                                                       1999; Gregory and Hann, 2000; Chen et al., 2000). The
6. Myc and signal transduction                                         mutations at and around Thr-58 and Ser-62 found in
                                                                       Burkitt’s lymphoma have been shown to result in increased
   The conclusions that in normal cells the expression of              stability of Myc (Salghetti et al., 1999; Bahram et al., 2000;
Myc is highly regulated and that deregulated expression                Gregory and Hann, 2000). Consistent with this finding the
of Myc is important for tumor progression are well                     analysis of signaling that emanates from Ras and targets
supported by experimental evidence. The regulation of                  Myc has been shown to result in increased Myc stability
Myc at the posttranslational level has become more trans-              (Sears et al., 1999, 2000). This appears to be the conse-
parent over the last several years. In particular regulation by        quence of Ser-62 phosphorylation and at the same time
phosphorylation and protein stability have obtained consid-            inhibition of Thr-58 phosphorylation by inhibiting a PI3K/
erable interest, also in view of their relevance for tumori-           GSK3-dependent mechanism (Sears et al., 2000). This
genesis. Nevertheless some of the findings that have been              increase in Myc stability may be relevant during the early
reported are still controversial and will require more                 phases of the cell cycle in which Ras-dependent signaling
detailed analysis.                                                     contributes to entry of cells into G1 from a resting state.
   c-Myc is a highly phosphorylated protein with three clus-           Since these findings have been obtained from Myc over-
ters of phosphorylation sites. Two of these clusters, one in           expression studies, it will now be important to demonstrate
10                                                 B. Lüscher / Gene 277 (2001) 1–14

an increase in stability of endogenous Myc in response to              sive role Myc plays in the control of cell proliferation one
growth factors. Ser-62 phosphorylation is not only triggered           would suspect the former possibility.
by Ras signaling but also by cyclin/CDK complexes and                     The expression of the c-myc gene and the regulation of
possibly other kinases suggesting that changes in Myc turn-            the protein are highly regulated. Recent progress implicates
over may also occur in other phases of the cell cycle.                 the Ras pathway in the control of the half life of Myc. Also
However such effects may be difficult to document since                mutations in the TAD of c-Myc are associated with
at any given point in time only a subfraction of Myc may               increased protein stability. These findings suggest that the
respond. Indeed the half life of the total Myc pool changes            short half life is one way to interfere with Myc function by
little through the cell cycle with the exception of mitosis            limiting its presence. The regulation of protein stability
(Lüscher and Eisenman, 1987; Gregory and Hann, 2000). It              appears to be connected to signaling pathways that are
has also been suggested recently that transactivation and              controlled by Ras. Since the majority of studies have been
degradation may be coupled (Thomas and Tyers, 2000;                    performed by using overexpression systems, it will be criti-
Tansey, 2001). Thus this may provide yet another signal                cal in the future to establish whether similar events occur in
for Myc turnover, potentially through the same phosphory-              cells that are not manipulated. This will be important since
lation sites. The emerging picture indicates that Myc protein          overexpression of both, Myc and Ras, can have advert
turnover is a highly dynamic process we are just beginning             effects on cell behavior. Thus indirect effects will have to
to understand.                                                         be excluded.
    While more evidence regarding the regulation of Myc is                Our knowledge regarding other Myc/Max/Mad network
becoming available (see above), there is still almost nothing          members is still patchy. The prediction that Mad proteins
known about the regulation of Mad proteins. Nevertheless               would function as tumor suppressors did not come true,
the short half life of Mad and the fact that these proteins are        although the overexpression studies have indicated that
phosphorylated in vivo offers possibilities for regulation             these proteins are potent inhibitors of cell proliferation.
that will have to be explored in the future. In addition               However the analysis of knock out mice did not support
Max has been considered as the inert interaction partner               these original findings. This may be due to overlapping
for Myc and Mad proteins. Max is ubiquitously expressed                expression patterns and functions of different Mad proteins.
and constitutively phosphorylated by CK2 and little infor-             Thus combinations of knock outs will hopeful provide more
mation regarding its regulation has been obtained (Henriks-            insight into the function of these proteins.
son and Lüscher, 1996; Grandori et al., 2000). However                   We are witnessing an ever increasing list of Myc target
recent evidence indicates that during apoptosis several path-          genes, mainly due to the use of DNA microarray screens. It
ways target Max resulting in its dephosphorylation and clea-           will be a challenge to understand which of these target genes
vage by caspases (Krippner-Heidenreich et al., 2001). These            are critical for tumor formation and thus may provide targets
findings suggest that the central component of the Myc/                for therapy. In this respect it will also be interesting to see
Max/Mad network is regulated under specific circum-                    whether Myc that is overexpressed, as in many human
stances. Further studies are required to define whether that           tumors, regulates additional genes as compared to normal
affects the function of other components of the network, in            physiological levels of Myc, or whether the difference is
particular Myc and Mad regarding their involvement in                  merely quantitative. Defining these differences will require
apoptosis.                                                             the input and work, preferentially concerted, of many
                                                                       laboratories.

7. Outlook
                                                                       Acknowledgements
    Recent years have brought to light novel aspects of Myc
function and regulation. In particular a number of new inter-             I apologize for the omission of many important and rele-
action partners have been identified that suggest an impor-            vant papers due to space limitations. I thank many collea-
tant role of Myc in the control of chromatin remodeling.               gues, in particular M. Eilers, L.-G. Larsson, and J. Lüscher-
Both SWI/SNF and TRRAP complexes can be recruited                      Firzlaff, and members of my laboratory for many helpful
by Myc, however, the order of events and many of the                   discussions. The work in my laboratory is supported by the
molecular details remain to be determined. As one example              Deutsche Forschungsgemeinschaft and by the Fonds der
it is unclear which HAT activity is recruited to mediate the           Chemischen Industrie.
acetylation of core histones of Myc responsive promoters. A
more fundamental question that has not been addressed yet
is whether Myc has the ability to open silent promoters, i.e.          References
whether Myc can function as the initial chromatin remodel-
                                                                       Aalfs, J.D., Kingston, R.E., 2000. What does ‘chromatin remodeling’
ing activity that is required to make promoters permissive                mean? Trends Biochem. Sci. 25, 548–555.
for transcription factors. Or is Myc a factor that can only            Adams, J.M., Cory, S., 1992. Oncogene co-operation in leukaemogenesis.
function when promoters are preactivated? From the deci-                  Cancer Surv. 15, 119–141.
B. Lüscher / Gene 277 (2001) 1–14                                                             11

Adams, P.D., 2001. Regulation of the retinoblastoma tumor suppressor             Blake, R.A., Broome, M.A., Liu, X., Wu, J., Gishizky, M., Sun, L., Court-
    protein by cyclin/cdks. Biochim. Biophys. Acta 1471, M123–M133.                  neidge, S.A., 2000. SU6656, a selective src family kinase inhibitor,
Albert, T., Urlbauer, B., Kohlhuber, F., Hammersen, B., Eick, D., 1994.              used to probe growth factor signaling. Mol. Cell Biol. 20, 9018–9027.
    Ongoing mutations in the N-terminal domain of c-Myc affect transacti-        Blomberg, I., Hoffmann, I., 1999. Ectopic expression of Cdc25A acceler-
    vation in Burkitt’s lymphoma cell lines. Oncogene 9, 759–763.                    ates the G(1)/S transition and leads to premature activation of cyclin E-
Alland, L., Muhle, R., Hou Jr., H., Potes, J., Chin, L., Schreiber-Agus, N.,         and cyclin A-dependent kinases. Mol. Cell Biol. 19, 6183–6194.
    DePinho, R.A., 1997. Role for N-CoR and histone deacetylase in Sin3-         Bouchard, C., Thieke, K., Maier, A., Saffrich, R., Hanley-Hyde, J.,
    mediated transcriptional repression. Nature 387, 49–55.                          Ansorge, W., Reed, S., Sicinski, P., Bartek, J., Eilers, M., 1999. Direct
Amati, B., 2001. Integrating Myc and TGF-beta signalling in cell-cycle               induction of cyclin D2 by Myc contributes to cell cycle progression and
    control. Nat. Cell Biol. 3, E112–E113.                                           sequestration of p27. EMBO J. 18, 5321–5333.
Amati, B., Dalton, S., Brooks, M.W., Littlewood, T.D., Evan, G.I., Land,         Bouchard, C., Dittrich, O., Kiermaier, A., Dohmann, K., Menkel, A., Eilers,
    H., 1992. Transcriptional activation by the human c-Myc oncoprotein in           M., Lüscher, B., 2001. Regulation of cyclin D2 gene expression by the
    yeast requires interaction with Max. Nature 359, 423–426.                        Myc/Max/Mad network: Myc-dependent TRRAP recruitment and
Amati, B., Brooks, M.W., Levy, N., Littlewood, T.D., Evan, G.I., Land, H.,           histone acetylation at the cyclin D2 promoter. Genes Dev. 15, 2042–
    1993a. Oncogenic activity of the c-Myc protein requires dimerization             2047.
    with Max. Cell 72, 233–245.                                                  Boyd, K.E., Farnham, P.J., 1999. Identification of target genes of oncogenic
Amati, B., Littlewood, T.D., Evan, G.I., Land, H., 1993b. The c-Myc                  transcription factors. Proc. Soc. Exp. Biol. Med. 222, 9–28.
    protein induces cell cycle progression and apoptosis through dimeriza-       Cairo, S., Merla, G., Urbinati, F., Ballabio, A., Reymond, A., 2001.
    tion with Max. EMBO J. 12, 5083–5087.                                            WBSCR14, a gene mapping to the Williams–Beuren syndrome deleted
Amati, B., Frank, S.R., Donjerkovic, D., Taubert, S., 2001. Function of the          region, is a new member of the Mlx transcription factor network. Hum.
    c-Myc oncoprotein in chromatin remodeling and transcription.                     Mol. Genet. 10, 617–627.
    Biochim. Biophys. Acta 1471, M135–M145.                                      Charron, J., Malynn, B.A., Fisher, P., Stewart, V., Jeannotte, L., Goff, S.P.,
Antonson, P., Pray, M.G., Jacobsson, A., Xanthopoulos, K.G., 1995. Myc               Robertson, E.J., Alt, F.W., 1992. Embryonic lethality in mice homo-
    inhibits CCAAT/enhancer-binding protein alpha-gene expression in                 zygous for a targeted disruption of the N-myc gene. Genes Dev. 6,
    HIB-1B hibernoma cells through interactions with the core promoter               2248–2257.
    region. Eur. J. Biochem. 232, 397–403.                                       Chen, L., Smith, L., Accavitti-Loper, M.A., Omura, S., Bingham Smith, J.,
Axelson, H., Henriksson, M., Wang, Y., Magnusson, K.P., Klein, G., 1995.             2000. Ubiquitylation and destruction of endogenous c-mycS by the
    The aminoterminal phosphorylation sites are frequenyly mutated in                proteasome: are myc boxes dispensable? Arch. Biochem. Biophys.
    Burkitt Lymphomas but not in mouse plasmacytomas. Eur. J. Cancer                 374, 306–312.
    31, 2099–2104.                                                               Cheng, S.W.G., Davies, K.P., Yung, E., Beltran, R.J., Yu, J., Kalpana, G.V.,
Ayer, D.E., Eisenman, R.N., 1993. A switch from Myc:Max to Mad:Max                   1999. c-Myc interacts with INI1/hSNF5 and requires the SWI/SNF
    heterocomplexes accompanies monocyte/macrophage differentiation.                 complex for activation function. Nat. Genet. Nat. Genet. 22, 102–105.
    Genes Dev. 7, 2110–2119.                                                     Cheung, P., Allis, C.D., Sassone-Corsi, P., 2000. Signaling to chromatin
Ayer, D.E., Kretzner, L., Eisenman, R.N., 1993. Mad: a heterodimeric                 through histone modifications. Cell 103, 263–271.
    partner for Max that antagonizes Myc transcriptional activity. Cell          Chiariello, M., Marinissen, M.J., Gutkind, J.S., 2001. Regulation of c-myc
    72, 211–222.                                                                     expression by PDGF through Rho GTPases. Nat. Cell Biol. 3, 580–586.
Bahram, F., von der Lehr, N., Cetinkaya, C., Larsson, L.G., 2000. c-Myc          Claassen, G.F., Hann, S.R., 1999. Myc-mediated transformation: the
    hot spot mutations in lymphomas result in inefficient ubiquitination and         repression connection. Oncogene 18, 2925–2933.
    decreased. Blood 95, 2104–2110.                                              Claassen, G.F., Hann, S.R., 2000. A role for transcriptional repression of
Barone, M.V., Courtneidge, S.A., 1995. Myc but not Fos rescue of PDGF                p21CIP1 by c-Myc in overcoming transforming growth factor beta -
    signalling block caused by kinase- inactive Src. Nature 378, 509–512.            induced cell-cycle arrest. Proc. Natl. Acad. Sci. USA 97, 9498–9503.
Bartek, J., Bartkova, J., Lukas, J., 1997. The retinoblastoma protein path-      Coller, H.A., Grandori, C., Tamayo, P., Colbert, T., Lander, E.S., Eisen-
    way in cell cycle control and cancer. Exp. Cell Res. 237, 1–6.                   man, R.N., Golub, T.R., 2000. Expression analysis with oligonucleotide
Baudino, T.A., Cleveland, J.L., 2001. The Max network gone mad. Mol.                 microarrays reveals that MYC regulates genes involved in growth, cell
    Cell Biol. 21, 691–702.                                                          cycle, signaling, and adhesion. Proc. Natl. Acad. Sci. USA 97, 3260–
Berns, A., 1991. Tumorigenesis in transgenic mice: identification and char-          3265.
    acterization of synergizing oncogenes. J. Cell Biochem. 47, 130–135.         Dang, C.V., 1999. c-Myc target genes involved in cell growth, apoptosis,
Bhatia, K., Huppi, K., Spangler, G., Siwarski, D., Iyer, R., Magrath, I.,            and metabolism. Mol. Cell. Biol. 19, 1–11.
    1993. Point mutations in the c-Myc transactivation domain are common         Davis, A.C., Wims, M., Spotts, G.D., Hann, S.R., Bradley, A., 1993. A null
    in Burkitt’s lymphomas and mouse plasmacytomas. Nat. Genet. 5, 56–               c-myc mutation causes lethality before 10.5 days of gestation in homo-
    61.                                                                              zygotes and reduced fertility in heterozygous female mice. Genes Dev.
Bhatia, K., Spangler, G., Gaidano, G., Hamdy, N., Dalla-Favera, R.,                  7, 671–682.
    Magrath, I., 1994. Mutations in the coding region of c-myc occur             Dey, A., She, H., Kim, L., Boruch, A., Guris, D.L., Carlberg, K., Sebti,
    frequently in acquired immunodeficiency syndrome-associated                      S.M., Woodley, D.T., Imamoto, A., Li, W., 2000. Colony-stimulating
    lymphomas. Blood 84, 883–888.                                                    factor-1 receptor utilizes multiple signaling pathways to induce cyclin
Billin, A.N., Eilers, A.L., Queva, C., Ayer, D.E., 1999. Mlx, a novel Max-           D2 expression. Mol. Biol. Cell 11, 3835–3848.
    like BHLHZip protein that interacts with the Max network of transcrip-       Eilers, M., Schirm, S., Bishop, J.M., 1991. The Myc protein activates
    tion factors. J. Biol. Chem. 274, 36344–36350.                                   transcription of the a-prothymosin gene. EMBO J. 10, 133–141.
Billin, A.N., Eilers, A.L., Coulter, K.L., Logan, J.S., Ayer, D.E., 2000.        Ekholm, S.V., Reed, S.I., 2000. Regulation of G(1) cyclin-dependent
    MondoA, a novel basic helix-loop-helix-leucine zipper transcriptional            kinases in the mammalian cell cycle. Curr. Opin. Cell Biol. 12, 676–
    activator that constitutes a positive branch of a max-like network. Mol.         684.
    Cell Biol. 20, 8845–8854.                                                    Evan, G.I., Wyllie, A.H., Gilbert, C.S., Littlewood, T.D., Land, H., Brooks,
Blackwood, E.M., Eisenman, R.N., 1991. Max: a helix-loop-helix zipper                M., Waters, C.M., Penn, L.Z., Hancock, D.C., 1992. Induction of apop-
    protein that forms a sequence-specific DNA-binding complex with                  tosis in fibroblasts by c-myc protein. Cell 69, 119–128.
    Myc. Science 251, 1211–1217.                                                 Facchini, L.M., Penn, L.Z., 1998. The molecular role of Myc in growth and
Blackwood, E.M., Kretzner, L., Eisenman, R.N., 1992. Myc and Max func-               transformation: recent discoveries lead to new insights. FASEB J. 12,
    tion as a nucleoprotein complex. Curr. Opin. Genet. Dev. 2, 227–235.             633–651.
You can also read