You are on page 1of 19

MiniReview

Glucose-sensing and -signalling mechanisms in yeast


Filip Rolland
1
, Joris Winderickx, Johan M. Thevelein

Laboratorium voor Moleculaire Celbiologie, Institute of Botany and Microbiology, Katholieke Universiteit Leuven, Kasteelpark Arenberg 31,
B-3001 Leuven-Heverlee, Flanders, Belgium
Received 20 September 2001; received in revised form 14 January 2002; accepted 28 January 2002
First published online 12 March 2002
Abstract
Glucose has dramatic effects on the regulation of carbon metabolism and on many other properties of yeast cells. Several sensing and
signalling pathways are involved. For many years attention has focussed on the main glucose-repression pathway which is responsible for
the downregulation of respiration, gluconeogenesis and the transport and catabolic capacity of alternative sugars during growth on
glucose. The hexokinase 2- dependent glucose-sensing mechanism of this pathway is not well understood but the downstream part of the
pathway has been elucidated in great detail. Two putative glucose sensors, the Snf3 and Rgt2 non-transporting glucose carrier homologs,
control the expression of many functional glucose carriers. Recently, several new components of this glucose-induction pathway have been
identified. The Ras-cAMP pathway controls a wide variety of cellular properties in correlation with cellular proliferation. Glucose is a
potent activator of cAMP synthesis. In this case glucose sensing is carried out by two systems, a G-protein-coupled receptor system and a
still elusive glucose-phosphorylation-dependent system. The understanding of glucose sensing and signalling in yeast has made dramatic
advances in recent years and has become a strong paradigm for the elucidation of nutrient-sensing mechanisms in other eukaryotic
organisms. 2002 Federation of European Microbiological Societies. Published by Elsevier Science B.V. All rights reserved.
Keywords: Glucose sensing; Signal transduction; G-protein-coupled receptor; cAMP; Saccharomyces cerevisiae
1. Introduction
In the free-living micro-organisms constantly changing
environment nutrient availability is the major factor con-
trolling growth and development. For yeasts, like for
many other micro-organisms, glucose is the preferred car-
bon and energy source. It is therefore not surprising that
glucose is an important primary messenger molecule, sig-
nalling optimal growth conditions to the cellular machin-
ery. Accordingly, glucose also aects many of the yeasts
commercially important traits such as growth rate, fer-
mentation capacity and stress resistance. Together with
its genetic amenability as a unicellular eukaryote, this
has stimulated the thorough characterization of a variety
of glucose-signalling pathways in Saccharomyces cerevisi-
ae. Whereas downstream components and their function-
ing have often been claried in great detail, elucidation of
the initial glucose-sensing and -activation mechanisms has
proven to be more dicult. This is largely due to the
sugars apostrophe dual function as a nutrient and signal-
ling molecule, and the intertwining of the molecular basis
of the two functions. Recently, however, substantial prog-
ress has been made with the identication of several pro-
teins with an apparently specic function in glucose sens-
ing. In higher multicellular organisms similar mechanisms
might be involved in the vital control of glucose homeo-
stasis.
2. Stationary phase, respiration and fermentation
Unicellular free-living organisms like yeasts in general
have adapted very well to constantly changing environ-
mental conditions. More specically, they have developed
mechanisms to respond to extreme variations in nutrient
availability by modulating their growth and metabolism.
The most dramatic eect in micro-organisms is observed
upon nutrient starvation. Micro-organisms are able to sur-
vive long periods of starvation by drastically decreasing
their metabolic activity upon growth and cell cycle arrest,
1567-1356 / 02 / $22.00 2002 Federation of European Microbiological Societies. Published by Elsevier Science B.V. All rights reserved.
PII: S 1 5 6 7 - 1 3 5 6 ( 0 2 ) 0 0 0 4 6 - 6
* Corresponding author. Tel. : +32 (16) 321507; +32 (16) 321500
(secr.) ; Fax: +32 (16) 321979.
E-mail address: johan.thevelein@bio.kuleuven.ac.be (J.M. Thevelein).
1
Present address: Department of Genetics, Harvard Medical School
and Department of Molecular Biology, Massachusetts General Hospital,
Boston, MA 02114, USA.
FEMSYR 1457 5-6-02
FEMS Yeast Research 2 (2002) 183^201
www.fems-microbiology.org
combined with a range of physiological and often also
morphological changes. These stationary-phase cells are
also characterized by their high tolerance to heat and oth-
er stress conditions and to cell wall-degrading enzymes. A
wide variety of genes involved in stress resistance is in-
duced and the reserve carbohydrate glycogen as well as
the stress protectant and reserve carbohydrate trehalose
accumulate to high levels. Although yeasts are basically
unicellular fungi, nutrient limitation can also cause a dras-
tic morphogenetic switch in diploid cells, resulting in pseu-
dohyphal growth. This morphology resembles that of the
lamentous fungi and reminds of the yeasts derivation
from multicellular ancestors. Such lamentous growth oc-
curs for instance when fermentable sugars are available
but nitrogen is lacking, presumably enabling the yeast to
actively search for a nitrogen source.
Yeast cells are not only able to detect the mere presence
or absence of nutrients, depending on the carbon source
available, they display totally dierent metabolic modes.
Glucose-sensitive yeasts like S. cerevisiae and Schizosac-
charomyces pombe prefer fermentation over respiration
even under aerobic conditions. In these yeasts, synthesis
of key enzymes of respiratory sugar dissimilation is re-
pressed by the ample presence of rapidly-fermentable sug-
ars, such as glucose or fructose. Although, per mole of
sugar, alcoholic fermentation yields fewer ATP equivalents
than respiration, it can proceed at much higher rates. This
enables these yeasts to compete eectively for survival,
especially because the ethanol produced during fermenta-
tion inhibits growth of competing micro-organisms. This
ethanol can subsequently aerobically be used as a non-
fermentable carbon source resulting in a complete use of
all available carbon. In the presence of oxygen, cells are
able to respire and generate ATP from non-fermentable
carbon sources by mitochondrial oxidative phosphoryla-
tion. Cells that use non-fermentable carbon sources grow
much slower than fermenting cells. In addition, they dis-
play several features which are similar to those of station-
ary-phase cells, such as high expression levels of genes
involved in stress resistance and accumulation of reserve
carbohydrates.
The addition of glucose to cells growing on non-fer-
mentable carbon sources or to stationary-phase cells trig-
gers a wide variety of regulatory processes directed to-
wards the exclusive and optimal utilization of the
preferred carbon source. Glycolysis is activated and glu-
cose is almost completely converted into ethanol and car-
bon dioxide. While glucose inux and the ow through
glycolysis are stimulated, gluconeogenesis is inhibited. In
addition, there is a drastic increase in growth rate which is
preceded by a characteristic upshift in ribosomal RNA
and protein synthesis. Genes encoding enzymes involved
in the uptake and metabolization of alternative carbon
sources and gene products involved in stress resistance
are repressed. Reserve carbohydrates are mobilized.
Yeast cells use both positive and negative control mech-
anisms to regulate enzyme levels and activities in order to
accomplish this drastic metabolic switch. Enzyme levels
are regulated at the stage of gene transcription (repression
and induction), mRNA stability, translation and protein
stability, while enzyme activities are regulated post-tran-
scriptionally by allosteric and covalent activation and in-
hibition. Most of these processes are aected either di-
rectly or indirectly by specic glucose sensing and signal
transduction pathways.
3. Glucose-signalling pathways
The major downregulating eect of glucose takes place
at the transcriptional level. One class of genes repressed by
glucose encodes proteins involved in respiration (Krebs
cycle and electron transport chain proteins), gluconeogen-
esis and the glyoxylate cycle. Another important class en-
codes proteins that are specically involved in the uptake
and metabolization steps of alternative carbon sources,
such as the GAL, SUC and MAL genes and genes in-
volved in utilization of ethanol, lactate and glycerol.
Also, high-anity glucose transport is repressed by high
levels of glucose. Several families of genes involved in the
use of other carbon sources are under control of family-
specic inducers enabling a co-ordinated regulation of
their expression. In the presence of glucose, the family-
specic inducers as well as the individual genes are subject
to repression. Finally, also a large group of STRE (stress
response element)-controlled genes encoding proteins pri-
marily involved in the yeasts response to various stresses
are repressed by glucose.
3.1. Glucose repression by the main glucose-repression
pathway
Not all glucose-repressible genes are repressed in the
same way but isolation and characterization of repression
and derepression mutants has identied a general glucose-
repression machinery involved in the regulation of expres-
sion of a large number of glucose-repressed genes. As il-
lustrated in Fig. 1A, its central components are the Mig1
transcriptional repressor complex, the Snf1-protein kinase
complex and protein phosphatase 1.
Mig1 is a DNA-binding zinc-nger protein that recruits
the general co-repressor proteins Ssn6 and Tup1 to exert
repression of diverse gene families and their family-specic
transcriptional inducer genes [1]. Essential for the function
of Mig1 in glucose repression is its glucose-regulated sub-
cellular localization. In the presence of high levels of glu-
cose, Mig1 rapidly moves into the nucleus, where it binds
to the promoters of glucose-repressible genes. When the
cells are deprived of glucose, Mig1 is rapidly transported
back to the cytoplasm [2]. In addition to Mig1 other
DNA-binding proteins (such as its homolog Mig2) are
involved in glucose repression.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 184
Transcription of the glucose-repressible genes in dere-
pressing conditions is dependent on the Snf1-protein ki-
nase complex. In the absence of glucose Snf1 probably
phosphorylates and thereby causes translocation of Mig1
to the cytoplasm [2^4]. The Snf1 Ser/Thr kinase is associ-
ated with an activating subunit and three scaolding pro-
teins in high-molecular-mass complexes. The activating
subunit Snf4 (Cat3) is required for Snf1 activity [5,6],
while the Sip1, Sip2 and Gal83 proteins maintain associa-
tion of Snf4 with the Snf1 kinase [7] and confer specicity
to the kinase complex [8], possibly through regulation of
its subcellular localization [9].
Snf1 kinase activity is inhibited by glucose and stimu-
lated when glucose is limiting [6,10]. Activation of the ki-
nase in response to glucose limitation is apparently accom-
panied by a conformational change of the kinase complex
[11] (Fig. 1B). According to the model derived from the
observed alterations in protein interactions within the
complex, the Snf1 regulatory domain auto-inhibits the cat-
alytic domain in glucose-grown cells. In the absence of
glucose, however, the Snf4-activating subunit binds to
the Snf1 regulatory domain, counteracting the auto-inhib-
itory interaction and thereby enabling Mig1 phosphoryla-
tion and its translocation to the cytoplasm [5]. The glucose
signal apparently regulates (inhibits) the Snf1^Snf4 inter-
action, thereby stimulating auto-inhibition of the kinase.
The kinase is then unable to inhibit Mig1-mediated repres-
sion [11]. Snf1 has also been shown to regulate activity as
well as (Mig1-dependent) expression of the two zinc-clus-
ter-activator proteins Cat8 and Sip4 which are involved in
the induction of gluconeogenic genes through carbon
source-responsive promoter elements [8,12^14].
Snf1 kinase activity itself also appears to be regulated
by phosphorylation and dephosphorylation. Several ex-
periments suggest the existence of a protein kinase that
activates Snf1 by phosphorylating a conserved Thr kinase
phosphorylation site in the activation loop [6,10,15,16].
Protein phosphatase 1 (Glc7) has been shown to act an-
tagonistically to Snf1 in glucose repression. This phospha-
tase is involved in the control of a variety of processes and
its glucose-repression-specic regulatory subunit Reg1/
Hex2 targets its activity to the activated Snf1 kinase do-
main, presumably dephosphorylating Snf1 or another
component of the complex and facilitating the return to
the auto-inhibited state [16^18]. Hence, although the glu-
cose signal most likely inhibits the initial phosphorylation
of Snf1, it may also activate Reg1-Glc7 phosphatase 1
function.
3.2. Glucose induction
Yeast cells growing on glucose obtain their energy
mainly through fermentation. Since fermentation is a rel-
atively inecient way of generating energy, a high glyco-
lytic ux is essential. Yeast cells are able to increase their
glycolytic capacity by the induction of a large number of
glycolytic genes. In addition, glucose-uptake capacity is
increased through the induction of several glucose-trans-
porter-encoding HXT genes. Separate signal transduction
pathways and mechanisms seem to be involved.
In the presence of rapidly fermentable sugars yeast gly-
colysis is fully activated. Glucose causes a fast increase
and transient overshoot in glycolytic intermediates and
mutant studies have shown that increased levels of dier-
Fig. 1. The main glucose-repression pathway. A: Simplied schematic
representation of mediators and targets of the main catabolite-repression
pathway. Repression is exerted by the complex Mig1/Ssn6/Tup1 on dif-
ferent gene families including family-specic transcriptional activators
such as Gal4 (galactose utilization), MalR (maltose utilization), Hap4
(respiratory genes) and Cat8 (gluconeogenic genes). The Snf1 kinase as-
sociated with one of the regulatory subunits Sip1, Sip2 or Gal83 and
the activating subunit Snf4 has a negative eect on the activity of the
repression complex. During growth on glucose, Snf1 activity is inhibited
by dierent upstream regulators which include the hexose kinases and
the Glc7 phosphatase. The Snf1 kinase complex is also required for acti-
vation of Sip4 which is required in concert with Cat8 for the derepres-
sion of the gluconeogenic genes. B: Glucose-induced conformational
change of the Snf1-protein kinase complex.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 185
ent metabolites trigger the induction of glycolytic genes
[19]. Dierent glycolytic intermediates seem to act as sig-
nalling molecules or metabolic messengers to adapt gly-
colytic activity to the presence of varying amounts of sug-
ars in a very complex but highly controlled and ecient
way. How these metabolic signals are transmitted is still
unclear but in several genes sequence elements have been
dened that are responsible for sugar-induced expression
and dierent DNA-binding factors have been identied
that are required for high-level expression of glycolytic
genes. The Gcr1 protein, for instance, seems to be of cen-
tral importance for the coordinated regulation of glyco-
lytic gene expression. It is a trans-acting positive regulator
of transcription that binds to the CTTCC motif which is
conserved in most glycolytic genes [20,21]. The glycolytic
pathway is also subject to extensive post-translational al-
losteric and covalent regulation. An increase in the glu-
cose-6-P level, for example, also triggers rapid activation
of 6-phosphofructo-2-kinase (PF2K) which catalyzes syn-
thesis of fructose-2,6-bisP, one of the allosteric regulators
in glycolysis [19]. In addition, rapid inactivation of gluco-
neogenesis is required for an ecient start-up of glycolysis.
S. cerevisiae contains a whole series of hexose transport-
ers homologues (Hxt1-17, Gal2, Snf3 and Rgt2), all dis-
playing dierent substrate anities and expression pat-
terns [22^24]. Depending on the amount of glucose
present in the medium, specic transporters are expressed.
The mechanisms involved in the expression of the appro-
priate transporters and their post-translational modica-
tion have recently become more clear. High-anity trans-
porters like Hxt6 and Hxt7 are highly expressed on non-
fermentable carbon-sources and repressed by high levels of
glucose, whereas transporters with low anity, such as
Hxt1 and Hxt3, are induced by the presence of a high
concentration of glucose. The transporters with intermedi-
ate anity for glucose like Hxt2 and Hxt4, on the other
hand, are induced by low levels of glucose and repressed
by high levels of glucose. As shown in Fig. 2, both the
intermediate and the low-anity transporters are re-
pressed by Rgt1 in the absence of glucose. Rgt1 is a
zinc-nger-containing DNA-binding protein that, like
Mig1, recruits the Ssn6 repressor to the promoters of spe-
cic genes [25]. Low amounts of glucose inhibit Rgt1-re-
pressor function, resulting in derepression of HXT expres-
sion. This inhibition requires the presence of the Grr1
protein [26]. This protein is part of a multiprotein SCF
complex containing the Skp1, Cdc53 and Cdc34 proteins
and the F-box Grr1 protein [27^29]. SCF complexes direct
protein ubiquitination and dier in their F-box-containing
component which is thought to recruit specic substrates
to the complex. Subsequent ubiquitination then marks
the substrate for degradation [30]. Glucose derepression
apparently involves ubiquitin-mediated proteolysis but it
is not clear whether the SCF complex directly modies
Rgt1. The HXT2 and HXT4 genes which encode trans-
porters with intermediate anity for glucose are repressed
by high glucose levels. This repression is mediated by the
Mig1 main glucose-repression pathway [26]. Also, repres-
sion of the high-anity transporter HXT6 is, at least in
part, mediated by the main glucose-repression pathway
[24]. However, in contrast to other glucose-repressed
genes, maintenance of HXT6 repression is strictly depen-
dent on Snf3 [31]. Expression of HXT1, encoding a low-
anity transporter is further induced by high glucose lev-
els. Besides the Grr1-Rgt1-dependent pathway, this also
involves another mechanism, that shares some compo-
nents with the main glucose-repression pathway [26].
This induction is independent of Rgt1 and apparently re-
quires a yet unidentied transcriptional activator or, alter-
natively, an additional Ssn6-dependent repression mecha-
nism that is inactivated by high levels of glucose. Full
induction of HXT1 expression at high glucose concentra-
tions, however, does require Rgt1. Rgt1 apparently can be
converted into an activator of HXT1 expression under
these conditions. Interestingly, Grr1 is required for both
low-glucose-induced inactivation and high-glucose-in-
duced conversion of Rgt1 [25]. In addition to glucose-con-
centration-dependent induction and repression, glucose
transport is also subject to extensive post-translational
regulation [24].
3.3. The Ras-cAMP pathway
A major glucose-signalling pathway involved in post-
translational regulation by phosphorylation is the Ras-
cAMP pathway (Fig. 3A). Synthesis of cAMP from ATP
is catalyzed by the enzyme adenylate cyclase and cAMP
activates cAMP-dependent protein kinase A (PKA) by
binding to its regulatory subunits (encoded by BCY1),
thereby releasing and activating the catalytic protein ki-
nase subunits (encoded by TPK1, TPK2 and TPK3). In
derepressed yeast cells (growing on a non-fermentable car-
bon source or in stationary phase) rapidly-fermentable
sugars, and especially glucose, trigger a rapid, transient
increase in the cAMP level, initiating a PKA phosphory-
lation cascade. Also, intracellular acidication is able to
trigger a pronounced increase in the cAMP level. Like in
higher eukaryotes, yeast adenylate cyclase activity is con-
trolled by G-proteins. Remarkably, in S. cerevisiae the
two small G-proteins Ras1 and Ras2 are essential for ade-
nylate cyclase activity. They therefore have been thought
for many years to act as functional equivalents of the
mammalian heterotrimeric GK-proteins of adenylate cy-
clase. Recently, however, a G-protein-coupled receptor
(GPCR) system has been identied that specically con-
trols glucose-induced activation of cAMP synthesis.
In S. cerevisiae cAMP signalling plays a central role in
the control of metabolism, stress resistance and prolifera-
tion. Translational control of Cln3 synthesis by PKA has
been proposed as a link between nutrient availability and
cell cycle control [32,33]. Indeed, several phenotypic prop-
erties controlled by PKA are indicative of high PKA ac-
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 186
tivity during fast growth on glucose and low activity dur-
ing growth on non-fermentable carbon sources and in sta-
tionary phase [34,35]. However, there is no clear correla-
tion with basal cAMP levels. Moreover, glucose-induced
activation of adenylate cyclase is repressed by glucose and
therefore considered not to be operative during growth on
glucose. This appears to conne the physiological role of
this pathway to the short period of transition from the
derepressed state to the repressed state by means of a
cAMP-triggered protein phosphorylation cascade. Most
of the downstream targets of PKA identied are enzymes
involved in intermediary metabolism and carbon metabo-
lism in particular, consistent with a role for cAMP signal-
ling in stimulation of fermentation. In addition to activa-
tion of enzymes involved in energy metabolism, glucose-
induced activation of protein synthesis through PKA-de-
pendent induction of ribosomal protein genes stimulates
growth and proliferation [36]. In their natural environ-
ment, yeast cells experience long periods of nutrient star-
vation, alternating with very short periods of nutrient
abundance. Under such conditions, fast recovery from sta-
tionary phase and initiation of fermentation clearly oer a
selective advantage [35].
Initiation of fermentation also coincides with a loss of
stress resistance. Two multicopy suppressors of the snf1
defect, Msn2 and Msn4, appear to mediate glucose repres-
sion of stress resistance by the cAMP-PKA pathway.
These zinc-nger proteins act as positive transcription fac-
tors in the general stress-response pathway by binding to
STREs in the promoters of stress-regulated genes [37,38].
Nuclear localization of Msn2 and Msn4 is regulated an-
tagonistically by stress conditions and PKA activity [39].
Consistently, a large number of STRE-controlled genes
which are dependent on Msn2 and Msn4 for induction
upon sugar depletion was found to be repressed by
cAMP [40,41]. Moreover, PKA activity was shown to be
dispensable in a strain lacking Msn2 and Msn4, indicating
that Msn2/4-dependent gene expression actually accounts
for many of the pleiotropic eects of PKA. PKA appar-
ently regulates processes such as glycogen accumulation
and stress response as well as growth by suppression of
Msn2/4-gene expression [42]. Interestingly, also the rapa-
mycin-sensitive TOR-signalling pathway was shown to in-
hibit expression of carbon-source-regulated genes by se-
questration of Msn2 and Msn4 in the cytoplasm [43].
The central role of the cAMP-PKA pathway in the control
of stress resistance is supported by the isolation of mu-
tants decient in fermentation-induced loss of stress resis-
tance (l). The l1 mutant carries a point mutation in the
CYR1/CDC35 gene [44], encoding adenylate cyclase, con-
sistent with the previous isolation of stress-resistant ade-
nylate cyclase mutants [45]. Interestingly, the l1 mutant
still displays wild-type growth and fermentation rates, as
opposed to other mutants with reduced activity of the
cAMP pathway. The l2 mutation was identied in the
gene encoding the GPCR Gpr1, which is specically in-
volved in glucose activation of cAMP synthesis [46]. In-
terestingly, a positive correlation was reported between
activity of the PKA pathway and longevity [47].
The observation that cAMP synthesis is apparently only
Fig. 2. Regulation of HXT transporter gene expression in response to glucose. In the absence of glucose, Rgt1-represses transcription of HXT1-4. Low
amounts of glucose inhibit the Rgt1-repressing activity, a process triggered by Snf3 via Grr1-mediated ubiquitination. At high concentrations of glucose,
Rgt2 triggers HXT1 expression. This involves Grr1-dependent conversion of Rgt1 into a transcriptional activator and another mechanism in which sev-
eral components of the main glucose-repression pathway are involved. The Snf3- and Rgt2-mediated derepression of the HXT genes also involves se-
questering at the plasma membrane of the transcriptional repressors Mth1 and Std1. At high glucose concentrations HXT2, HXT4, HXT6 and SNF3
are repressed by Mig1 via the main glucose-repression pathway. In addition, Snf3 is involved in a second pathway leading to the high-glucose-induced
repression of HXT6.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 187
activated by rapidly-fermentable sugars and not by other
nutrients, and the glucose-repressible character of the ac-
tivation mechanism, seem to argue against a role for
cAMP-signalling in growth control by nutrients. Bcy1 v
mutants with attenuated catalytic subunits have indeed
been shown to respond appropriately to nutritional stress
conditions, even in the absence of adenylate cyclase [48].
This indicates that cAMP-independent mechanisms exist
for regulation of these responses. Interestingly, in the pres-
ence of glucose, other essential nutrients (such as N, S or P
Fig. 3. Control of PKA activity in yeast. A: Activation of the cAMP pathway occurs when glucose is added to cells growing on non-fermentable car-
bon sources or to stationary phase cells. Glucose is detected via a dual sensing process: an intracellular glucose-sensing process involving the hexose ki-
nases following transport of the glucose, and the extracellular glucose detection system involving the Gpr1^Gpa2 GPCR system. How the glucose signal
is transmitted to adenylate cyclase is still unknown but a possible involvement of the Ras proteins and their regulators Cdc25 and the Ira proteins can-
not be excluded. B: The FGM pathway integrates the availability of dierent nutrients including the fermentable carbon source. It supports mainte-
nance of high PKA activity during growth on glucose via a cAMP-independent signalling cascade that involves the Sch9-protein kinase. In contrast to
the cAMP pathway the intra- and extracellular glucose-sensing process is apparently able to sustain activation of the pathway separately. Detection of
other nutrients seems to be triggered by specic transporters such as Gap1 for amino acids and Mep2 for ammonium.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 188
sources) are able to trigger similar eects on the PKA
targets when re-added to cells starved for such a nutrient.
These eects are independent of activation of cAMP syn-
thesis but are still dependent on the free catalytic Tpk
subunits (Fig. 3B). Since these eects require the presence
of both a fermentable carbon source and a complete
growth medium for sustained activation, the signalling
pathway involved has been called the fermentable-
growth-medium-induced (FGM) pathway [34,49]. The
FGM pathway controls PKA targets during growth on
glucose through the Sch9-protein kinase [50].
Often dierent mechanisms and signal transduction
pathways collaborate to control enzyme levels and activ-
ities. The extreme glucose sensitivity of the gluconeogenic
enzymes for example is mediated by glucose-induced allo-
steric inhibition, covalent modication and protein degra-
dation [51^53] as well as transcriptional repression and
accelerated mRNA degradation [54^56]. The combination
of these mechanisms ensures the rapid decrease in gluco-
neogenic enzyme levels when yeast cells switch to glyco-
lytic metabolism. Also, in the case of enzymes and perme-
ases involved in the metabolism of alternative carbon
sources, such as maltose and galactose, repression of
gene expression is preceded by rapid glucose-induced in-
activation and degradation [57].
4. Glucose-sensing mechanisms
The dramatic eects of glucose on growth and metabo-
lism clearly support a hormone-like function for this sugar
in yeast cells. However, since it is also taken up and me-
tabolized as a nutrient, glucose can be detected by the cells
in many more ways than is the case for classical primary
messenger molecules [58]. Although cells could use the
activity of a component of the existing metabolic machin-
ery or the level of one or more glucose catabolites to
detect its presence (and metabolization), the lack of spec-
icity of such a system could have stimulated the develop-
ment of more specic sensors as illustrated in Fig. 4. Re-
ceptors could have evolved from existing glucose-binding
proteins such as transporters or kinases (with or without
maintenance of the catalytic activity) or members of more
classical receptor families could have been recruited and
modied (or used originally) to gather specic information
on the nutritional status in the environment.
There is now much evidence that yeast uses a whole
range of such sensing mechanisms to ne-tune growth
and metabolic activity to the amount and quality of the
sugars available. For genes encoding glycolytic enzymes
and requiring glucose for full expression, induction by
glucose was shown to depend on the accumulation of in-
termediary metabolites. For some genes, an increase in the
level of hexose-6-phosphates is required while for others
induction is triggered by glycolytic three-carbon metabo-
lites [19,59,60]. Also, for glucose sensing and signalling in
pancreatic L-cells a more extensive metabolization of the
sugar is required since it appears that the actual trigger for
insulin release is the ATP produced in glycolysis and res-
piration. An increase in the ATP:ADP ratio inhibits ATP-
sensitive K

-channels. Membrane depolarization then ac-


tivates voltage-gated Ca
2
-channels, triggering a rise in
intracellular Ca
2
which stimulates fusion of insulin stor-
age vesicles with the plasma membrane. Many glucose-
Fig. 4. Possible mechanisms for glucose sensing. Glucose can be detected by specic glucose receptors in the plasma membrane (a), by an active glucose
transporter (b) or transporter homologs that developed into a glucose sensor (c). When the glucose-sensing mechanism is dependent on metabolism the
sensor can be a hexose kinase homolog that developed into a regulatory protein with weak or no catalytic activity (d) or an active glucose-phosphory-
lating enzyme in which the catalytic and regulatory functions are closely related (e). Finally, the glucose signal can be a metabolic messenger (f), either
glucose-6-phosphate or a downstream metabolite.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 189
induced eects studied in yeast, however, require only
partial metabolization of the sugar. This makes it possible
to distinguish clearly between the regulatory function of
glucose and its nutrient function: most glucose-induced
signal transduction pathways apparently require no me-
tabolization beyond the sugar kinase step for their activa-
tion. This also points at a central role for transport and/or
phosphorylation in the sensing process. The use of non- or
partially-metabolizable sugar analogues is a very useful
tool to determine to which extent the sugar has to be
metabolized to trigger a response.
Interestingly, sugar transport and phosphorylation
themselves are subject to complex regulation by glucose
through dierent signal transduction mechanisms. Re-
cently, research on the regulation of expression of glucose
transporter genes has contributed signicantly to our
understanding of the glucose-sensing process. Two trans-
porter homologs, Rgt2 and Snf3, have been proposed to
function as sensors or receptors of extracellular glucose for
induction of HXT expression. Research on the glucose-
sensing mechanisms involved in catabolite repression, on
the other hand, has focussed from the very beginning on
the involvement of the sugar kinases and more specically
on a predominant role for hexokinase 2 (Hxk2) as an
intracellular sensor. Finally, work in our laboratory has
revealed the involvement of a GPCR system in concert
with a glucose phosphorylation-dependent mechanism
for glucose-induced activation of cAMP-synthesis.
4.1. The Rgt2 and Snf3 glucose sensors
Rgt2 and Snf3 are two unusual members of the hexose
transporter family. They have only limited sequence sim-
ilarities to the other hexose transporter homologs and pos-
sess long C-terminal cytoplasmic tails. The SNF3 gene was
originally identied in a screen for mutants decient in the
utilization of the trisaccharide ranose [61,62], based on
the inability to derepress the invertase-encoding SUC2
gene. Snf3 (sucrose non-fermenting) mutants in addition
are unable to grow fermentatively on low concentrations
of glucose or fructose and kinetic analysis showed that the
SNF3 gene is required for high-anity glucose transport
[63]. Sequence homology with mammalian glucose trans-
porters [64] supported the idea of a function as high-an-
ity glucose transporter. The RGT2 gene was cloned as a
dominant mutant allele (RGT2-1) that bypasses the re-
quirement of Snf3 for growth on low concentrations of
glucose by restoring high-anity transport [65,66]. More
recent results indicate that Snf3 and Rgt2 do not directly
support catabolic sugar transport [31,67] but rather act as
extracellular glucose sensors, involved in the regulation of
expression of catabolic hexose transporter genes (Fig. 2).
Snf3 was found to be required for induction of transcrip-
tion of the HXT2, HXT3 and HXT4 genes by low levels of
glucose, suggesting that snf3 mutants are defective in high-
anity transport because of decient expression of the
high-anity transporter-encoding genes [26]. Rgt2 is re-
quired for maximal induction of HXT1 expression by
high concentrations of glucose [66]. Interestingly, a dom-
inant mutation in RGT2 was identied that causes consti-
tutive glucose-independent expression of the HXT1 gene
[66]. The mutation results in the substitution of arginine
231 into lysine. This residue is located at the start of the
fth cytoplasmic loop of the protein in a highly conserved
region within the transporter superfamily. When intro-
duced in SNF3 (Arg229Lys), the mutation causes similar
eects, resulting in constitutive expression of HXT2. These
results suggest that Rgt2 senses high extracellular glucose
concentrations, while Snf3 senses low glucose concentra-
tions. The fact that RGT2 is expressed constitutively at a
low level while SNF3 is glucose-repressed is also consistent
with a role as respectively low- and high-anity glucose
sensors, although the observation that the dominant mu-
tant RGT2-1 allele restores high-anity glucose uptake in
a snf3 mutant strain suggests some overlap in the target
genes. To date, little is known about the actual Snf3 and
Rgt2 glucose-sensing mechanism and the nature of the
signal they transmit.
The molecular structure of Rgt2 and Snf3 is distinct
from that of most other glucose-transporter proteins, es-
pecially in the long carboxy-terminal extension that is be-
lieved to be located in the cytoplasm [64]. Deletion anal-
ysis of Snf3 showed that this carboxy-terminal extension is
indeed required for Snf3-dependent expression of the high-
anity transporter genes. The Snf3 and Rgt2 C-terminal
tails are relatively dissimilar except for a sequence motif
that occurs twice in the Snf3 tail and once in the tail of
Rgt2. This motif is essential for their signalling function
[68^70]. Although the dominant mutations suggest the in-
volvement of the conserved arginine residue in the fth
cytoplasmic loop in the glucose-sensing process, the car-
boxy-terminal tails indeed appear to be the actual signal-
ling domains. Transplantation of the Snf3-tail onto the
Hxt1 and Hxt2 glucose transporters was suggested to con-
vert these transporters into functional glucose sensors able
to generate the signal for glucose-induced HXT gene ex-
pression [68]. Moreover, even when expressed as a soluble
protein, the cytoplasmic Snf3 tail was still found to signal
[70,71]. This obviously has important implications for the
way in which the sensors function.
Two proteins, Std1 and Mth1, have been shown to in-
teract with the tails of the glucose sensors and genetic
analysis suggests that they are involved in transduction
of the glucose signal to regulate invertase and hexose
transporter gene expression [72^74]. STD1 (MSN3) was
originally isolated as a multicopy suppressor of the snf
(sucrose non-fermenting) phenotype of an snf4 mutant
by a partial relief of SUC2 repression [75,76]. Its MTH1
homolog is allelic to the genes HTR1, DGT1 and BPC1,
for which previously dominant mutant alleles have been
isolated [73,74,77]. These dominant mutations were shown
to cause severely impaired glucose uptake [78^80]. MTH1
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 190
deletion suppresses the ranose growth defect of an snf3
mutant as well as the glucose fermentation defect of snf3
rgt2 double mutants through increased and unregulated
expression of the HXT2, HXT3 and HXT4 hexose trans-
porter genes. Deletion of STD1 cannot suppress the fer-
mentation defect but specically increases HXT expression
in the presence of low glucose concentrations. Std1 and
Mth1 apparently act through distinct pathways and, like
Snf3 and Rgt2, respond to dierent levels of glucose. Std1
was shown to act upstream of the Snf1 kinase, both for
derepression of SUC2 and high-anity transporter gene
expression and repression of HXT1 expression, whereas
Mth1 mediates repression via an Snf1-independent path-
way [72]. The mutant forms of MTH1 (HTR1, DGT1 and
BPC1) apparently block transduction of the Snf3- and
Rgt2-mediated glucose signals upstream of the Rgt1 re-
pressor [73,74]. Studies with green uorescent protein fu-
sions indicated that Std1 is localized in the cell periphery
and the cell nucleus, supporting the idea that it may trans-
duce signals from the plasma membrane to the nucleus
[72]. The HXT expression data and the fact that Snf3
overexpression blocks the ability of Std1 to induce
SUC2 expression suggest that the glucose sensors and
the Std1 and Mth1 proteins act antagonistically, with the
sensors being required for HXT induction and the Std1
and Mth1 proteins being required for their repression
[72]. Possibly, Snf3 and Rgt2 inhibit the negative (repres-
sing) eects of Mth1 and Std1 by sequestering them at the
plasma membrane [73].
The observation that Rgt2 and Snf3 alone do not sus-
tain transport of glucose to enable growth, (not even when
overexpressed) and isolation of the dominant RGT2-1 and
SNF3-1 mutations in the fth cytoplasmic loop, have led
to the hypothesis that Snf3 and Rgt2 function as classical
signal receptors, in which binding of the extracellular li-
gand, in this case glucose, causes a conformational change
in a cytoplasmic domain [66]. Consistently, the hxt1-7v
strain, which is decient in glucose uptake was reported
to exhibit normal glucose induction of HXT1 and HXT2
(as measured by fusions of their promoter to LacZ) [68].
However, Hxk2 was shown to be partially required for full
induction of HXT expression [26]. Phosphorylation of the
sugar might be important for wild-type Rgt2- and Snf3-
mediated signalling and the small amounts of glucose that
are still taken up and phosphorylated in an hxt1-7v strain
may be sucient to enable signalling.
The use of transporter-like proteins as nutrient sensors
may be a more common strategy in eukaryotic cells. The
conserved sequence motif in the C-termini of Rgt2 and
Snf3 is also present in the C-terminal extension of Rag4
from Kluyveromyces lactis. Rag4 was shown to control
expression of the Rag1 glucose transporter [81] and may
function both as a high- and low-anity glucose sensor
[82]. In Neurospora crassa, the transporter homolog and
glucose sensor Rco3 contains a C-terminal extension sim-
ilar to that of Snf3 and Rgt2 [83]. Also, the yeast Ssy1
protein, which is homologous to amino acid permeases
and contains an N-terminal cytoplasmic tail, was shown
to act as an amino acid sensor controlling amino acid
permease gene expression. In Arabidopsis thaliana, evi-
dence exists for specic Hxk-independent sucrose sensing
and transporter homologs have been proposed to act as
sucrose sensors [84,85]. On the other hand, functional nu-
trient transporters might also play a role in nutrient sens-
ing. Evidence for a role of the ammonium transporter
Mep2 in nitrogen sensing for control of pseudohyphal
growth in yeast has been reported [86]. Also, for Gap1,
evidence for a role in amino acid sensing for control of the
PKA- and FGM-pathways targets has been obtained re-
cently (Donaton, M.C.V., Holsbeeks, I., Lagatie, O.,
Crauwels, M., Winderickx, J. and Thevelein, J.M., unpub-
lished results) (see Fig. 3B). In higher eukaryotes nutrient
transporters with a sensing function might also be present.
Recently, a regulatory function for mammalian Glut1 in
glucose-induced activation of ERK (MAPK)-signalling
has been suggested [87].
4.2. Glucose repression: the Hxk glucose sensor
A second mechanism controlling expression of sugar
transporters is the main glucose-repression pathway. In
the presence of glucose, high-anity glucose transport is
repressed together with a broad range of other genes in-
volved in the utilization of alternative carbon sources. In
addition, also Hxk1 and glucokinase (Glk1) are repressed
by glucose. Both high-anity transporters and sugar ki-
nases appear to be involved at least to some extent in
triggering their own repression, since activation of the
glucose-repression mechanism requires uptake and subse-
quent phosphorylation of the sugar.
In a variety of conditions the level of glucose repression
was found to correlate well with the level of glucose-trans-
port activity [78^80] and it has often been speculated that
specic glucose transporters or pairs of them could play a
role in triggering glucose-induced regulatory responses
[34,78,88,89]. The requirement of a signicant amount of
glucose ( s20 mM) to fully trigger these signalling path-
ways was thought to indicate the specic involvement of
low-anity transport. Experiments with yeast strains ex-
pressing individual transporters showed that triggering of
glucose repression is not dependent on a specic hexose
transporter protein but rather correlates with the glucose
uptake activity of the cells and with glycolytic ux [90,91].
HXK2 was identied as one of the rst genes involved in
glucose repression [92^94] but whether its requirement for
signalling simply reects the need for glucose phosphory-
lation or involves a separate regulatory function for the
Hxk2 protein has been and still is a matter of debate.
Early experiments suggested that of the three sugar ki-
nases Hxk2 played a unique role in glucose repression
and it was proposed that this kinase was a bifunctional
enzyme with catalytic and regulatory domains for glucose
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 191
repression [95,96]. However, this could not be conrmed
in later experiments; in a large number of mutant Hxk
strains a good correlation between glucose repression
and residual phosphorylating capacity of the mutated
Hxk was observed [97]. A similar correlation was observed
with a series of Hxk1^Hxk2 hybrid constructs [98]. Also,
when Hxk1 was removed in addition to Hxk2, glucose
repression further diminished. Stable overexpression
showed that Hxk1 was also capable of mediating glucose
repression at least to a certain extent [98]. Interestingly, no
further metabolization beyond the sugar phosphorylation
step appears to be necessary for triggering glucose repres-
sion since phosphoglucoisomerase mutants with only 1%
residual isomerase activity still showed normal glucose re-
pression [98]. In addition, 2-deoxyglucose, which is trans-
ported and phosphorylated but not further metabolized,
also triggers repression [99]. This glucose analogue was
used to isolate glucose-repression mutants which, as op-
posed to wild-type cells, are able to grow on ranose in its
presence [92]. Overexpression of GLK1 did not restore
glucose repression in a Hxk mutant [98] indicating that
glucose phosphorylation by itself is not sucient to trigger
glucose repression. These results supported the idea of a
specic function of the Hxk proteins in the activation
mechanism of glucose repression. More recently, new
data on the dierential requirement of the sugar kinases
in short- and long-term glucose and fructose repression
and the complex transcriptional regulation of the kinases
themselves has put the predominant role of Hxk2 in a new
light. Catabolite repression was shown to involve two dis-
tinct mechanisms: an initial rapid response is mediated
through any kinase able to phosphorylate the sugar, in-
cluding Glk1, while long-term repression specically re-
quires Hxk2 for repression by glucose and either Hxk1
or Hxk2 for repression by fructose [100,101]. Both
HXK1 and GLK1 are repressed upon addition of glucose
or fructose but fructose repression of HXK1 is only tran-
sient. This is consistent with the preference of Hxk1 for
fructose as a substrate and its requirement for long-term
fructose repression [101]. Apparently, activation of catab-
olite repression is controlled by a complex interregulatory
network, involving all three sugar kinases and the mecha-
nisms and pathways controlling their expression. In this
way not only the main glucose-repression pathway itself
but also cAMP signalling indirectly aects catabolite re-
pression [101]. Consistently, rapid repression of the gluco-
neogenic genes FBP1 and PCK1 by very low levels of
glucose was shown to be triggered in the presence of any
one of the three kinases, whereas in the presence of high
glucose levels repression was mediated specically by the
Hxk2-dependent Mig1-repression mechanism [55]. It was
proposed that HXK2 gene expression could act as a sensor
for the glucose concentration in the medium [102]. Also,
more recently, novel alleles of Hxk2 have been isolated
that have distinct eects on catalytic activity and catabo-
lite repression of SUC2 [46,103,104] and long- and short-
term phases of catabolite repression could be dissected
[103]. The lack of correlation between in vitro catalytic
activity of Hxk, in vivo sugar phosphate accumulation
and the establishment of catabolite repression again sug-
gested that the production of sugar phosphate is not the
only role of Hxk in repression but that also a regulatory
signalling site of the protein may be required (Fig. 5A).
For galactokinase a clear distinction between the catalytic
function and the regulatory function in induction of GAL
gene expression was made [105]. A similar situation might
apply to Hxk2. Structure^function analysis of Hxk2 more
specically suggests that the establishment of catabolite
repression is dependent on the onset of the phosphoryl
transfer reaction on Hxk and is probably related to the
stable formation of a transition intermediate and concom-
itant conformational changes within the enzyme [46]. Also,
in plants, Hxk is proposed to be a glucose sensor and
extensive mutant analysis seems to uncouple regulatory
and catalytic activity (Moore and Sheen; pers. comm).
Although the core components of the main glucose-re-
pression pathway and the important role of Hxk2 as a
putative glucose sensor have been identied, it still re-
mains to be elucidated what the actual glucose signal is
that triggers glucose repression. Since the rate of glucose
transport and phosphorylation correlate well with the level
of glucose repression, glucose-6-P or other initial glyco-
lytic metabolites have often been proposed to be the trig-
gering molecules. Interestingly, also ATP, the second sub-
strate for the sugar kinases during sugar phosphorylation,
has recently been implicated in the triggering reaction (re-
viewed by [106]). ATP and ADP, respectively, are the sub-
strate and product of the phosphorylation reaction. There-
fore the AMP/ATP ratio could in principle act as some
sort of sensor for sugar phosphorylation and metabolic
activity (Fig. 5B). One model proposes a signalling role
for these nucleotides in triggering glucose repression based
on the fact that the three components of the Snf1 kinase
(Snf1, Snf4 and the Sip proteins) are similar to the sub-
units of the functionally related mammalian AMP-acti-
vated protein kinase (AMPK) [6,107]. Mammalian
AMPK is involved in the cellular response to a variety
of stresses, like heat shock and nutrient starvation. Inacti-
vation of a number of biosynthetic enzymes under these
conditions ensures better conservation of cellular ATP
[10,108]. Likewise, since it is responsible for triggering
derepression, Snf1 is involved indirectly in the generation
of ATP by enabling the cells to metabolize alternative
carbon sources in the absence of fermentable amounts of
glucose. Although it has been shown that Snf1 is not di-
rectly activated by AMP [6,107], a good correlation be-
tween Snf1 activity and the AMP/ATP ratio was reported
[10]. In glucose-growing cells, ATP generation by glycol-
ysis depletes AMP. When the glucose is exhausted, the
AMP level is repleted, resulting in a high AMP/ATP ratio
which could then activate Snf1 and relieve repression.
Thus, in this model the triggering signal for repression is
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 192
generated by the metabolism of glucose, consistent with
the predominant role of Hxk2 in both glucose phosphor-
ylation during fermentative growth and glucose repression.
However, although an increase in the AMP/ATP ratio was
observed when repressed cells are shifted to low-glucose
medium [10], AMP and ATP levels during growth on
glycerol and glucose appear to be very similar [109]. In
addition, this model does not t with the indications for
a separate regulatory function for Hxk. Snf-related protein
kinase (SnRK) signalling is also conserved in plants. Plant
Snf1 homologs have been shown to complement yeast snf1
mutants and are proposed to act as global regulators of
carbon metabolism in plants [110]. As in yeast, plant
SnRKs are not directly activated by AMP although
AMP seems to inhibit their dephosphorylation [111]. In-
terestingly, glucose-6-P was reported to negatively regulate
a plant SnRK [112].
Yeast Hxk2 has recently been shown to have a role in
regulating the phosphorylation status of the regulatory
subunit of protein phosphatase 1 Reg1/Hex2. Reg1 is
phosphorylated in response to glucose limitation in an
Snf1-dependent way and dephosphorylated by Glc7
when glucose is present. Phosphorylation of Reg1 by
Snf1 appears to stimulate both Glc7 activity in promoting
closure of the Snf1 complex and release of Reg1-Glc7
from the kinase complex. Hxk2 either stimulates binding
and/or phosphorylation of Reg1 or inhibits dephosphory-
lation of Reg1 by Glc7 [18].
Other recent data suggest that the Hxk2 protein might
have an even more direct role in signalling to the repres-
sion machinery. It was found that Hxk2 resides partly in
the cell nucleus [113] and that this nuclear localization,
which is dependent on a specic internal nuclear localiza-
tion sequence, is necessary for glucose-repression signal-
ling [114]. Furthermore, the Hxk2 protein was shown to
participate in regulatory DNA^protein complexes with cis-
acting regulatory elements of the SUC2 promoter [114].
Hxk2 therefore might be involved in transducing the glu-
cose signal by interacting directly with transcriptional fac-
tors controlling the expression of glucose-repressed genes.
Phosphorylation at Ser-15, which also shifts the dimeric^
monomeric equilibrium, does not seem to aect nuclear
targetting [114]. Phosphorylation and the concomitant in-
crease in glucose anity of monomeric Hxk could provide
a mechanism to optimize glucose utilization at low con-
centrations [115], but although protein phosphatase 1 is
involved in dephosphorylation of the Hxk2 monomer
[113,116] seemingly contradictory results were obtained
as to whether this phosphorylation/dephosphorylation is
involved in signalling [97,113,114].
4.3. cAMP signalling: a dual sensing system
Experiments with hexose kinase and other glycolysis
mutants showed that transport and phosphorylation but
no further metabolization of the sugar is required to acti-
vate cAMP synthesis by glucose [117]. However, glucose-
6-P does not appear to be the trigger of the activation
reaction: the increase in the level of glucose-6-P after ad-
dition of dierent glucose concentrations did not show a
good correlation with the increase in the cAMP level.
From the increase in the cAMP level after addition of
dierent extracellular glucose concentrations an apparent
K
a
for the activation mechanism of about 25 mM was
deduced, tting with the K
m
of what was believed to be
the low-anity glucose transporter system but diering by
at least one order of magnitude from the K
m
values of the
three hexose kinases. Together, these results suggested that
Fig. 5. Role of Hxk in the main glucose-repression pathway. A: Model in which a regulatory signaling function is associated with Hxk. Although the
regulatory function can be closely associated with the catalytic activity, neither the substrates glucose and ATP nor the products glucose-6-phosphate
and ADP act as metabolic messengers. B: Model based on metabolic messenger function of nucleotides. Glycolysis changes the ADP/ATP and AMP/
ATP ratios. Changes in the nucleotide levels may act as a sensor of metabolic activity and exert a signalling function in triggering glucose repression.
This model is based on the similarity between mammalian AMPK and the dierent components of the Snf1 kinase complex.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 193
the primary triggering reaction was situated at the level of
transport and phosphorylation, possibly even transport-
associated phosphorylation. Glucose-induced cAMP-sig-
nalling is indeed dependent on transport of the sugar
but not on any specic glucose transporter. Also, the
transporter homologs and putative glucose sensors Rgt2
and Snf3 are not directly involved in this glucose-sensing
process [58]. The role of sugar transport is apparently
limited to the provision of a sucient amount of substrate.
Although glucose-6-P does not seem to be the metabolic
messenger for activation, in Hxk mutants a clear correla-
tion was always observed between catalytic activity and
the triggering of cAMP signalling [46,103]. Apparently,
the role of yeast Hxk in sugar-induced activation of
cAMP signalling is closely connected to the catalytic func-
tion of the enzyme. How glucose phosphorylation is
coupled to the control of cAMP synthesis is still unclear.
Basal activity of the cAMP pathway is essential for via-
bility and this makes it dicult to study the activation
mechanism.
It was proposed that the Ras proteins are not only
essential for maintaining a basal level of cAMP by sustain-
ing basal adenylate cyclase activity, but in addition are
signal transmitters in the pathway leading from glucose
to adenylate cyclase [118]. Subsequently also Cdc25, the
Ras-GEF, was shown to be involved in glucose-induced
activation of cAMP synthesis [119^121]. Glucose therefore
appeared to be a direct or indirect stimulator of Cdc25.
Recently, the possible involvement of the Ras proteins in
glucose signalling was investigated more directly [122]. In-
tracellular acidication, another stimulator of in vivo
cAMP synthesis, but not glucose, caused an increase in
the GTP/GDP ratio on the Ras proteins. Stimulation of
cAMP synthesis by glucose was shown to be dependent on
another G-protein, Gpa2. The GPA2 gene was originally
cloned as a yeast homolog of mammalian heterotrimeric
Ga-proteins and was already implicated in cAMP signal-
ling. However, although overexpression of the gene clearly
aected cAMP levels, no eect was observed in a gpa2v
strain on glucose-induced cAMP signalling [123,124]. This
was later shown to be due to interference with the eect of
intracellular acidication caused by the addition of glu-
cose. The increase in cAMP observed after addition of
100 mM glucose shortly after pre-addition of 5 mM glu-
cose was entirely dependent on the presence of Gpa2 [122].
Gpa2 does not seem to play an important role in the
control of the basal cAMP level. Moreover, although de-
letion of GPA2 confers to some extent the typical pheno-
type associated with a reduced level of cAMP, the function
of Gpa2 appears to be limited mainly to the stimulation of
cAMP synthesis during the transition from respirative
growth on a non-fermentable carbon source to fermenta-
tive growth on glucose [122].
Using the two-hybrid screen and Gpa2 as bait, a frag-
ment of a putative GPCR, Gpr1, was isolated [46,125,
126]. Surprisingly, Plc1 (phospholipase C) appears to be
required for this interaction [127]. The GPCR Gpr1, like
Gpa2, was shown to be specically required for glucose
activation of the cAMP pathway during the transition to
growth on glucose and a gpr1v mutant could be rescued
by the constitutively activated GPA2
val132
allele [46]. Ap-
parently, Gpr1 and Gpa2 constitute a glucose-sensing
GPCR system for activation of the cAMP pathway (Fig.
3). This not only brings the yeast adenylate cyclase system
back in line with the mammalian system of adenylate cy-
clase control, it also appears to be the rst example of a
GPCR system activated by a nutrient in eukaryotic cells.
S. pombe contains a similar glucose-sensing GPCR system
for activation of cAMP synthesis (consisting of the GK-
protein gpa2 and the putative glucose receptor git3) and
also Candida albicans contains a Gpr1 homolog with ex-
tensive similarity to its S. cerevisiae counterpart, suggest-
ing the existence of a new GPCR family involved in glu-
cose sensing [128].
Consistent with its requirement for glucose-induced
cAMP accumulation, GPR1 was also isolated as a mutant
allele (l2) in a screen for mutants decient in fermenta-
tion-induced loss (l) of heat resistance [46]. In a similar
screen, the RGS2 gene was isolated as a multi-copy sup-
pressor of glucose-induced loss of heat resistance [129].
RGS2 encodes a protein with a typical conserved RGS
(regulator of heterotrimeric G-protein signalling) domain
and was indeed shown to negatively regulate glucose acti-
vation of the cAMP pathway through direct inhibition of
Gpa2. Consistent with its homology to other RGS pro-
teins, Rgs2 acts as a stimulator of the GTPase activity of
Gpa2. It remains to be shown, however, that Gpa2 indeed
acts as the signal transducer from glucose to adenylate
cyclase. The fact that deletion of GPA2 is lethal in the
absence of Ras2 is consistent with a role for Gpa2 as
stimulator of adenylate cyclase [130].
A mutation in the catalytic domain of adenylate cyclase
(cyr1
met1876
) has been identied that specically aects glu-
cose- and acidication-induced cAMP signalling and not
the basal cAMP level [131]. This lcr1 (lack of cAMP re-
sponse) mutation not only abolishes the cAMP signal but
also the transient increase in the basal cAMP level ob-
served during the lag phase of growth on glucose [132].
In addition, it appears to counteract the overactivating
eect of both the RAS2
val19
- and GPA2
val132
- dominant
alleles, supporting the theory that Gpa2 indeed acts up-
stream of adenylate cyclase. The observation that elimina-
tion of glucose activation of cAMP-synthesis by the lcr1
mutation only results in a delay in glucose-induced
changes in PKA targets associated with the adaptation
to growth on glucose, and does not aect the typical var-
iations of PKA-controlled phenotypic properties during
diauxic growth, supports the idea of an alternative path-
way responsible for glucose signalling during growth.
Gpa2 was also found to be required for pseudohyphal
growth [130,133]. Pseudohyphal dierentiation is induced
in diploid cells in response to nitrogen starvation in the
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 194
presence of a fermentable carbon source and is mediated
both by the pheromone-responsive MAPK cascade and
the cAMP pathway. Consistent with the fact that the con-
stitutively active GPA2
val132
allele stimulates lamentation,
even on nitrogen-rich media, it was proposed that Gpa2 is
an element of the nitrogen-sensing machinery that regu-
lates pseudohyphal dierentiation by modulating cAMP
levels [133]. However, genetic and physiological studies
on pseudohyphal growth recently conrmed that the
Gpr1^Gpa2 GPCR system is activated by glucose. Be-
cause of the fact that GPR1 expression is induced by nitro-
gen starvation, it is proposed that the receptor acts as a
dual sensor for both abundant carbon and nitrogen star-
vation [134]. Obviously, the demonstration that Gpr1 itself
binds glucose and acts as a real glucose receptor is an
important issue.
As mentioned before, elucidation of the exact mecha-
nisms of glucose sensing is often complicated because of
the requirement for partial metabolism of the glucose.
This is also the case for the glucose-induced activation
mechanism of cAMP synthesis and the involvement of
the Gpr1^Gpa2 GPCR system. In spite of this, an actual
glucose-sensing function for Gpr1 has recently become
more apparent with the demonstration that Gpr1 is essen-
tial for the sensing of extracellular glucose. The glucose-
induced cAMP signal is not only dependent on the GPCR
system but also on transport and phosphorylation of the
sugar (Fig. 6A). We showed that it is possible to uncouple
the GPCR-dependent sensing process from the glucose
phosphorylation. For this purpose a method was estab-
lished allowing independent investigation of the two re-
quirements based on the observation that the absence of
the glucose-induced cAMP signal can be restored in the
Hxt null strain by pre-addition of a low concentration
(0.025% or 0.7 mM) of maltose (Fig. 6B). This concentra-
tion of maltose does not aect the cAMP level by itself but
apparently fullls the glucose phosphorylation require-
ment for activation of the cAMP pathway by glucose,
which in the Hxt null strain cannot be transported into
the cell. Using this set-up it was shown that the GPCR
Gpr1 or at least the glucose-sensing mechanism that is
dependent on it, specically responds to extracellular glu-
cose (and also sucrose, but not fructose or other sugars)
with low apparent anity. This is consistent with the fact
that yeast cells switch metabolism to the fermentative
mode only at glucose concentrations of at least 20 mM.
Interestingly, the presence of the constitutively active
GPA2
val132
allele increases the fructose-induced cAMP sig-
nal to the same intensity as the glucose signal in trans-
porter wild-type cells and enables concentrations as low as
5 mM glucose to fully activate the pathway. This is con-
sistent with the fact that in such a strain activation of the
pathway is only dependent on phosphorylation of the sug-
ar, since the GPCR system is constitutively activated. In
conclusion, the two essential requirements for glucose-in-
duced activation of cAMP synthesis can be fullled sepa-
rately. It remains unclear at what point the two require-
ments are integrated. Apparently, glucose phosphorylation
is required in some way to make adenylate cyclase respon-
sive to activation by the GPCR system. Since no increase
in the GTP/GDP ratio of Ras is observed after addition of
glucose, it seems unlikely that the hexose kinase-dependent
sensing system acts through Cdc25-Ras2. The kinases
might also act directly on adenylate cyclase, possibly re-
leasing inhibition of catalytic activity by the N-terminal
regulatory domain.
Fig. 6. Fulllment of the glucose phosphorylation requirement for cAMP signalling. A: The relationship between the intracellular glucose phosphoryla-
tion process and the Gpr1/Gpa2 dependent extracellular glucose detection system in a wild-type strain. B: In a strain without functional glucose trans-
porters (Hxt), the glucose phosphorylation requirement for cAMP signalling can be fullled by addition of a low level of maltose which is transported
and hydrolyzed by a specic system consisting of the maltose transporter (MalT) and maltase (MalS).
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 195
The demonstration that the Gpr1^Gpa2 GPCR system
is responsible for glucose control of the cAMP pathway
has brought up again the question as to what is the actual
function of Ras-dependent control of adenylate cyclase in
yeast [35]. Ras mutants exist which seem to be specically
aected in signal transduction. Strains carrying the
ras2
ser318
allele as their sole RAS gene, for example, dis-
play normal steady-state levels of cAMP, while the glu-
cose-induced cAMP signal is totally absent [135]. Also,
several mutants decient in post-translational modication
of Ras are specically decient in cAMP signalling [136].
Many results indicating a role for the Cdc25-Ras system in
glucose-induced cAMP signalling could possibly be ex-
plained by their requirement for localization of adenylate
cyclase to the plasma membrane. Consistent with such a
role for Cdc25 is the recent evidence of direct binding of
Cdc25 to adenylate cyclase through an SH3 domain. This
binding might promote an ecient assembly of the ade-
nylate cyclase complex [137]. Proper membrane localiza-
tion of the adenylate cyclase complex might be essential
for optimal interaction with and activation by the Gpr1^
Gpa2 system. The main function of the Ras proteins
might therefore be to control basal adenylate cyclase ac-
tivity [35]. S. cerevisiae cells indeed have a very high ca-
pacity to synthesize cAMP and a strict control of the
cAMP level is clearly essential, especially under less-favor-
able conditions that require slow growth and high stress
resistance. The association with Ras might increase the
responsiveness of adenylate cyclase to stimulation by the
GPCR system when it is activated by a high level of glu-
cose in the medium.
FGM signalling still occurs in hxk1vhxk2vglk1 and
gpr1v or gpa2v strains, possibly pointing to a totally dif-
ferent glucose-sensing system for FGM signalling com-
pared to cAMP signalling. However, recent results indi-
cate that the presence of one of the two glucose-sensing
systems might be sucient for FGM signalling while they
are both required for glucose activation of cAMP signal-
ling (Donaton, M., Winderickx, J. and Thevelein, J.M.,
unpublished results).
4.4. Allosteric regulation
Not all glucose-induced regulatory eects require a sig-
nal transduction mechanism. Allosteric activation and in-
hibition is exerted by metabolic intermediates of glucose
catabolism. Allosteric regulation has been studied rst
with respect to the control of glycolysis, which was the
rst metabolic pathway to be discovered and elucidated.
The main allosteric regulators of glycolysis appeared to be
fructose-2,6-bisP and fructose-1,6-bisP, controlling two of
its irreversible steps, catalyzed respectively by phospho-
fructokinase (PFK) and pyruvate kinase (PYK). Fruc-
tose-2,6-bisP not only activates PFK, but in addition in-
hibits fructose-1,6-bis-phosphatase, which catalyzes the
reverse reaction in gluconeogenesis. The product of the
PFK reaction, fructose-1,6-bis-phosphate, in turn allosteri-
cally activates PYK more downstream in glycolysis [138].
However, enhanced expression of both PFK1 and PYK1
does not change glycolytic ux signicantly [139] and mu-
tant studies of PF2Ks did not reveal an essential role for
fructose-2,6-bis-P in the regulation of carbon uxes in
yeast cells [140]. Apparently, these enzymes do not cata-
lyze rate-limiting steps in glycolysis and allosteric eects
appear to control metabolite homeostasis rather then met-
abolic uxes. Metabolic control analysis indeed pointed to
sugar uptake as the major ux-controlling step in glycol-
ysis [141]. The control coecient of glucose transport was
calculated to be signicantly higher then that of PFK. The
high level of control by transport over growth and glyco-
lytic ux has also been conrmed in an hxt null strain
expressing a single transporter [91]. More recently, the
trehalose-6-phosphate synthase subunit of the trehalose
synthase complex was found to control in some way the
entry of glucose into glycolysis [88]. While in mammalian
cells glucose-6-P is the most important allosteric inhibitor
of the hexose kinases, in yeast cells this function appears
to be exerted by trehalose-6-phosphate [142]. In addition,
there is evidence for the possible involvement of the Tps1
protein itself in controlling glycolytic ux [143,144]. Prop-
er control of glucose inux into glycolysis is required for a
wide range of glucose-signalling eects in yeast, as was
demonstrated with the tps1v mutant which is unable to
synthesize trehalose-6-phosphate and therefore shows a
severe deregulation of glycolysis after addition of glucose
[145]. This indicates that the research on glucose-sensing
mechanisms cannot be seen as separate from that on glu-
cose metabolism, and that for every mutant aected in
glucose signalling, investigation of possible interference
with glucose metabolism is of paramount importance.
5. Conclusions and perspectives
The preference of S. cerevisiae for glucose as a carbon
and energy source is reected by the variety of glucose-
sensing and -signalling mechanisms ensuring its optimal
use. Nutrient-sensing and -signalling mechanisms must
have evolved early in evolution and might be at the origin
of the sophisticated hormone- and growth factor-induced
signal transduction pathways best known from research
on mammalian cells. The glucose-sensing mechanisms in
yeast are therefore an excellent model system for studying
signal transduction in general. Glucose is also the prime
carbon and energy source in higher multicellular organ-
isms and it is becoming clear that glucose-sensing and -
signalling in these organisms is of vital importance for
maintenance of sugar homeostasis [58]. In mammals glu-
cose serves as the blood sugar and maintenance of the
glucose concentration within narrow limits is controlled
by a complex interplay of several endocrine and neural
glucostatic systems that direct its uptake and release
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 196
[146]. In addition, glucose also plays a more direct role in
transcriptional regulation [147]. In plants, sugars like su-
crose, glucose and fructose are the main products of pho-
tosynthesis and the primary carbon source for respiration.
Sugar-sensing and -signalling aects many aspects of
growth, metabolism and development throughout the
whole plant life cycle [148,149]. It is therefore likely that
higher eukaryotes are equally well supplied with glucose-
sensing and -signalling mechanisms. One striking example
of an apparently conserved signalling mechanism in eu-
karyotes is involved in the control of life span. Yeast lon-
gevity was shown to be regulated by PKA (adenylate cy-
clase) and Sch9. Longevity is often associated with
increased resistance to (oxidative) stress and the stress-re-
sistance transcription factors Msn2 and Msn4 were indeed
shown to be required for life-span extension in Sch9 and
adenylate cyclase mutants [150]. Another report empha-
sized the requirement of NAD (and its regulation of the
silencing protein Sir2) for life-span extension by caloric
restriction and the involvement of the cAMP-PKA path-
way in this process independently of stress resistance [47].
Interestingly, deletion of Gpr1 or Gpa2 had similar eects
on longevity as caloric restriction (growth on low glucose),
conrming the role of this GPCR system in the sensing of
high glucose concentrations. Sch9 shows most similarity to
Akt/PKB. This protein kinase is involved in a signalling
pathway controlled by an insulin receptor-like protein and
regulating carbon metabolism, stress resistance and lon-
gevity in Caenorhabditis elegans [151]. Since also human
Akt/PKB is involved in insulin signalling, translocation of
glucose transporters, apoptosis and cellular proliferation,
an ancient (glucose) signalling mechanism that coordin-
ately regulates metabolism, stress resistance and longevity
(enabling survival over long periods of starvation) may
have been conserved in all eukaryotic organisms [150].
Interestingly, also yeast Snf1 in addition to cellular energy
utilization was reported to control aging, and conserved
homologs in other organisms might have similar eects.
Since glucose signalling appears to be fundamental to cel-
lular and organismal function and therefore widespread
(and in some cases conserved), it is likely that similar
specic sensing mechanisms as in yeast are also present
in higher eukaryotes. An abundance of results seems to
point at a central role for Hxk in eukaryotic glucose sens-
ing. Although structure^function analysis and mutagenesis
have enabled separation of catalytic and regulatory activ-
ity to some extent, more detailed analysis will be required
to elucidate the actual Hxk- sensing and -signalling mech-
anism. Subcellular localization might be an important fac-
tor in Hxk regulatory function. In addition, more specic
sensors might be involved in higher eukaryotic glucose
sensing. As mentioned above, glucose transporter-like pro-
teins might have tissue- or cell-specic regulatory func-
tions in mammals and plants. Finally, classical receptor
families may be involved, as shown by the example of
yeast Gpr1. The elucidation of the yeast glucose-sensing
GPCR system obviously tempts to speculate that amongst
the hundreds of eukaryotic orphan receptors a subfamily
of nutrient sensors is waiting to be discovered.
References
[1] Treitel, M.A. and Carlson, M. (1995) Repression by SSN6-TUP1 is
directed by MIG1, a repressor activator protein. Proc. Natl. Acad.
Sci. USA 92, 3132^3136.
[2] Devit, M.J., Waddle, J.A. and Johnston, M. (1997) Regulated nuclear
translocation of the Mig1 glucose repressor. Mol. Biol. Cell. 8, 1603^
1618.
[3] Ostling, J. and Ronne, H. (1998) Negative control of the Mig1p re-
pressor by Snf1p-dependent phosphorylation in the absence of glu-
cose. Eur. J. Biochem. 252, 162^168.
[4] Treitel, M.A., Kuchin, S. and Carlson, M. (1998) Snf1 protein kinase
regulates phosphorylation of the Mig1 repressor in Saccharomyces
cerevisiae. Mol. Cell. Biol. 18, 6373^6380.
[5] Celenza, J.L. and Carlson, M. (1989) Mutational analysis of the
Saccharomyces cerevisiae SNF1 protein kinase and evidence for func-
tional interaction with SNF4 protein. Mol. Cell. Biol. 9, 5034^
5044.
[6] Woods, A., Munday, M.R., Scott, J., Yang, X., Carlson, M. and
Carling, D. (1994) Yeast SNF1 is functionally related to mammalian
AMP-activated protein kinase and regulates acetyl-CoA carboxylase
in vivo. J. Biol. Chem. 269, 19509^19515.
[7] Jiang, R. and Carlson, M. (1997) The Snf1 protein kinase and its
activating subunit, Snf4, interact with distinct domains of the Sip1/
Sip2/Gal83 component in the kinase complex. Mol. Cell. Biol. 17,
2099^2106.
[8] Vincent, O. and Carlson, M. (1999) Gal83 mediates the interaction of
the Snf1 kinase complex with the transcription activator Sip4. EMBO
J. 18, 6672^6681.
[9] Vincent, O., Townley, R., Kuchin, S. and Carlson, M. (2001) Sub-
cellular localization of the Snf1 kinase is regulated by specic beta
subunits and a novel glucose signaling mechanism. Genes Dev. 15,
1104^1114.
[10] Wilson, W.A., Hawley, S.A. and Hardie, D.G. (1996) Glucose repres-
sion/derepression in budding yeast: SNF1 protein kinase is activated
by phosphorylation under derepressing conditions, and this correlates
with a high AMP:ATP ratio. Curr. Biol. 6, 1426^1434.
[11] Jiang, R. and Carlson, M. (1996) Glucose regulates protein interac-
tions within the yeast SNF1 protein kinase complex. Genes Dev. 10,
3105^3115.
[12] Vincent, O. and Carlson, M. (1998) Sip4, a Snf1 kinase dependent
transcriptional activator binds to the carbon source-responsive ele-
ments of gluconeogenic genes. EMBO J. 17, 7002^7008.
[13] Hiesinger, M., Roth, S., Meissner, E. and Schuller, H.J. (2001) Con-
tribution of Cat8 and Sip4 to the transcriptional activation of yeast
gluconeogenic genes by carbon-source responsive elements. Curr.
Genet. 39, 68^76.
[14] Rahner, A., Scholer, A., Martens, E., Gollwitzer, B. and Schuller,
H.J. (1996) Dual inuence of the yeast Cat1p (Snf1p) protein kinase
on carbon source-dependent transcriptional activation of gluconeo-
genic genes by the regulatory gene CAT8. Nucleic Acids Res. 24,
2331^2337.
[15] Estruch, F., Treitel, M.A., Yang, X.L. and Carlson, M. (1992)
N-terminal mutations modulate yeast SNF1 protein kinase function.
Genetics 132, 639^650.
[16] Ludin, K., Jiang, R. and Carlson, M. (1998) Glucose-regulated inter-
action of a regulatory subunit of protein phosphatase 1 with the Snf1
protein kinase in Saccharomyces cerevisiae. Proc. Natl. Acad. Sci.
USA 95, 6245^6250.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 197
[17] Sanz, P., Ludin, K. and Carlson, M. (2000) Sip5 interacts with both
the Reg1/Glc7 protein phosphatase and the Snf1 protein kinase of
Saccharomyces cerevisiae. Genetics 154, 99^107.
[18] Sanz, P., Alms, G.R., Haystead, T.A. and Carlson, M. (2000) Reg-
ulatory interactions between the Reg1-Glc7 protein phosphatase and
the Snf1 protein kinase. Mol. Cell. Biol. 20, 1321^1328.
[19] Boles, E., Heinisch, J. and Zimmermann, F.K. (1993) Dierent sig-
nals control the activation of glycolysis in the yeast Saccharomyces
cerevisiae. Yeast 9, 761^770.
[20] Uemura, H., Koshio, M., Inoue, Y., Lopez, M.C. and Baker, H.V.
(1997) The role of Gcr1p in the transcriptional activation of glyco-
lytic genes in yeast Saccharomyces cerevisiae. Genetics 147, 521^
532.
[21] Clifton, D. and Fraenkel, D.G. (1981) The gcr (glycolysis regulation)
mutation of Saccharomyces cerevisiae. J. Biol. Chem. 256, 13074^
13078.
[22] Bisson, L.F., Coons, D.M., Kruckeberg, A.L. and Lewis, D.A. (1993)
Yeast sugar transporters. Crit. Rev. Biochem. Mol. Biol. 28, 259^308.
[23] Kruckeberg, A.L. (1996) The hexose transporter family of Saccharo-
myces cerevisiae. Arch. Microbiol. 166, 283^292.
[24] Boles, E. and Hollenberg, C.P. (1997) The molecular genetics of
hexose transport in yeasts. FEMS Microbiol. Rev. 21, 85^111.
[25] O

zcan, S., Leong, T. and Johnston, M. (1996) Rgt1p of Saccharo-


myces cerevisiae, a key regulator of glucose-induced genes, is both an
activator and a repressor of transcription. Mol. Cell. Biol. 16, 6419^
6426.
[26] O

zcan, S. and Johnston, M. (1995) Three dierent regulatory mech-


anisms enable yeast hexose transporter (HXT) genes to be induced by
dierent levels of glucose. Mol. Cell. Biol. 15, 1564^1572.
[27] Li, F.N. and Johnston, M. (1997) Grr1 of Saccharomyces cerevisiae is
connected to the ubiquitin proteolysis machinery through Skp1: cou-
pling glucose sensing to gene expression and the cell cycle. EMBO J.
16, 5629^5638.
[28] Skowyra, D., Craig, K.L., Tyers, M., Elledge, S.J. and Harper, J.W.
(1997) F-box proteins are receptors that recruit phosphorylated sub-
strates to the SCF ubiquitin-ligase complex. Cell 91, 209^219.
[29] Kishi, T., Seno, T. and Yamao, F. (1998) Grr1 functions in the
ubiquitin pathway in Saccharomyces cerevisiae through association
with Skp1. Mol. Gen. Genet. 257, 143^148.
[30] Patton, E.E., Willems, A.R. and Tyers, M. (1998) Combinatorial
control in ubiquitin-dependent proteolysis: dont Skp the F-box hy-
pothesis. Trends Genet. 14, 236^243.
[31] Liang, H. and Gaber, R.F. (1996) A novel signal transduction path-
way in Saccharomyces cerevisiae dened by Snf3-regulated expression
of HXT6. Mol. Biol. Cell. 7, 1953^1966.
[32] Hall, D.D., Markwardt, D.D., Parviz, F. and Heideman, W. (1998)
Regulation of the Cln3-Cdc28 kinase by cAMP in Saccharomyces
cerevisiae. EMBO J. 17, 4370^4378.
[33] Mendenhall, M.D. and Hodge, A.E. (1998) Regulation of Cdc28
cyclin-dependent protein kinase activity during the cell cycle of the
yeast Saccharomyces cerevisiae. Microbiol. Mol. Biol. Rev. 62, 1191^
1243.
[34] Thevelein, J.M. (1994) Signal transduction in yeast. Yeast 10, 1753^
1790.
[35] Thevelein, J.M. and de Winde, J.H. (1999) Novel sensing mechanisms
and targets for the cAMP-protein kinase A pathway in the yeast
Saccharomyces cerevisiae. Mol. Microbiol. 32, 1002^1012.
[36] Mager, W.H. and Planta, R.J. (1991) Coordinate expression of ribo-
somal protein genes in yeast as a function of cellular growth rate.
Mol. Cell. Biochem. 104, 181^187.
[37] Martinez-Pastor, M.T., Marchler, G., Schuller, C., Marchlerbauer,
A., Ruis, H. and Estruch, F. (1996) The Saccharomyces cerevisiae
zinc nger proteins Msn2p and Msn4p are required for transcrip-
tional induction through the stress-response element (STRE).
EMBO J. 15, 2227^2235.
[38] Schmitt, A.P. and Mcentee, K. (1996) Msn2p, a zinc nger DNA-
binding protein, is the transcriptional activator of the multistress
response in Saccharomyces cerevisiae. Proc. Natl. Acad. Sci. USA
93, 5777^5782.
[39] Gorner, W., Durchschlag, E., Martinez-Pastor, M.T., Estruch, F.,
Ammerer, G., Hamilton, B., Ruis, H. and Schu ller, C. (1998) Nuclear
localization of the C
2
H
2
zinc nger protein Msn2p is regulated by
stress and protein kinase A activity. Genes Dev. 10, 1516^1531.
[40] Boy-Marcotte, E., Perrot, M., Bussereau, F., Boucherie, H. and Jac-
quet, M. (1998) Msn2p and Msn4p control a large number of genes
induced at the diauxic transition which are repressed by cyclic AMP
in Saccharomyces cerevisiae. J. Bacteriol. 180, 1044^1052.
[41] Tadi, D., Hasan, R.N., Bussereau, F., Boy-Marcotte, E. and Jacquet,
M. (1999) Selection of genes repressed by cAMP that are induced by
nutritional limitation in Saccharomyces cerevisiae. Yeast 15, 1733^
1745.
[42] Smith, A., Ward, M.P. and Garrett, S. (1998) Yeast PKA represses
Msn2p/Msn4p-dependent gene expression to regulate growth, stress
response and glycogen accumulation. EMBO J. 17, 3556^3564.
[43] Beck, T. and Hall, M.N. (1999) The TOR signalling pathway controls
nuclear localization of nutrient-regulated transcription factors. Na-
ture 402, 689^692.
[44] Van Dijck, P., Gorwa, M.-F., Lemaire, K., Teunissen, A., Versele,
M., Colombo, S., Dumortier, F., Ma, P., Tanghe, A., Loiez, A. and
Thevelein, J.M. (2000) Characterization of a new set of mutants
decient in fermentation-induced loss of stress resistance for use in
frozen dough applications. Int. J. Food Microbiol. 55, 187^192.
[45] Iida, H. (1988) Multistress resistance of Saccharomyces cerevisiae
is generated by insertion of retrotransposon Ty into the 5P coding
region of the adenylate cyclase gene. Mol. Cell. Biol. 8, 5555^
5560.
[46] Kraakman, L., Lemaire, K., Ma, P., Teunissen, A.W.R.H., Donaton,
M.C.V., Van Dijck, P., Winderickx, J., de Winde, J.H. and Thevelein,
J.M. (1999) A Saccharomyces cerevisiae G-protein coupled receptor,
Gpr1, is specically required for glucose activation of the cAMP
pathway during the transition to growth on glucose. Mol. Microbiol.
32, 1002^1012.
[47] Lin, S.J., Defossez, P.A. and Guarente, L. (2000) Requirement of
NAD and SIR2 for life-span extension by caloric restriction in Sac-
charomyces cerevisiae. Science 289, 2126^2128.
[48] Cameron, S., Levin, L., Zoller, M. and Wigler, M. (1988) cAMP-
independent control of sporulation, glycogen metabolism and heat
shock resistance in S. cerevisiae. Cell 53, 555^566.
[49] Thevelein, J.M. (1991) Fermentable sugars and intracellular acidi-
cation as specic activators of the Ras-adenylate cyclase signalling
pathway in yeast: the relationship to nutrient-induced cell cycle con-
trol. Mol. Microbiol. 5, 1301^1307.
[50] Crauwels, M., Donaton, M.C.V., Pernambuco, M.B., Winderickx, J.,
de Winde, J.H. and Thevelein, J.M. (1997) The Sch9 protein kinase in
the yeast Saccharomyces cerevisiae controls cAPK activity and is
required for nitrogen activation of the fermentable-growth-medium-
induced (FGM) pathway. Microbiology 143, 2627^2637.
[51] Funayama, S., Gancedo, J.M. and Gancedo, C. (1980) Turnover of
yeast fructose-bisphosphatase in dierent metabolic conditions. Eur.
J. Biochem. 109, 61^66.
[52] Mazo n, M.J., Gancedo, J.M. and Gancedo, C. (1982) Inactivation of
yeast fructose-1,6-bisphosphatase. In vivo phosphorylation of the en-
zyme. J. Biol. Chem. 257, 1128^1130.
[53] Mazo n, M.J., Gancedo, J.M. and Gancedo, C. (1982) Phosphoryla-
tion and inactivation of yeast fructose-bisphosphatase in vivo by
glucose and by proton ionophores. A possible role for cAMP. Eur.
J. Biochem. 127, 605^608.
[54] Mercado, J.J., Smith, R., Sagliocco, F.A., Brown, A.J.P. and Gance-
do, J.M. (1994) The levels of yeast gluconeogenic mRNAs respond to
environmental factors. Eur. J. Biochem. 224, 473^481.
[55] Yin, Z.K., Smith, R.J. and Brown, A.J.P. (1996) Multiple signalling
pathways trigger the exquisite sensitivity of yeast gluconeogenic
mRNAs to glucose. Mol. Microbiol. 20, 751^764.
[56] Yin, Z., Hatton, L. and Brown, A.J. (2000) Dierential post-tran-
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 198
scriptional regulation of yeast mRNAs in response to high and low
glucose concentrations. Mol. Microbiol. 35, 553^565.
[57] Jiang, H., Medintz, I. and Michels, C.A. (1997) Two glucose sensing/
signaling pathways stimulate glucose-induced inactivation of maltose
permease in Saccharomyces. Mol. Biol. Cell. 8, 1293^1304.
[58] Rolland, F., Wanke, V., Cauwenberg, L., Ma, P., Boles, E., Vanoni,
M., de Winde, J.H., Thevelein, J.M. and Winderickx, J. (2001) The
role of hexose transport and phosphorylation in cAMP signalling in
the yeast Saccharomyces cerevisiae. FEMS Yeast Res. 1, 33^45.
[59] Boles, E. and Zimmermann, F.K. (1993) Induction of pyruvate de-
carboxylase in glycolysis mutants of Saccharomyces cerevisiae corre-
lates with the concentrations of 3-carbon glycolytic metabolites.
Arch. Microbiol. 160, 324^328.
[60] Muller, S., Boles, E., May, M. and Zimmermann, F.K. (1995) Dier-
ent internal metabolites trigger the induction of glycolytic gene ex-
pression in Saccharomyces cerevisiae. J. Bacteriol. 177, 4517^4519.
[61] Neigeborn, L. and Carlson, M. (1984) Genes aecting the regulation
of SUC2 gene expression by glucose repression in Saccharomyces
cerevisiae. Genetics 108, 845^858.
[62] Neigeborn, L., Schwartzberg, P., Reid, R. and Carlson, M. (1986)
Null mutations in the SNF3 gene of Saccharomyces cerevisiae cause a
dierent phenotype than do previously isolated missense mutations.
Mol. Cell. Biol. 6, 3569^3574.
[63] Bisson, L.F., Neigeborn, L., Carlson, M. and Fraenkel, D.G. (1987)
The SNF3 gene is required for high-anity glucose transport in Sac-
charomyces cerevisiae. J. Bacteriol. 169, 1656^1662.
[64] Celenza, J.L., Marshall-Carlson, L. and Carlson, M. (1988) The yeast
SNF3 gene encodes a glucose transporter homologous to the mam-
malian protein. Proc. Natl. Acad. Sci. USA 85, 2130^2134.
[65] Marshall-Carlson, L., Neigeborn, L., Coons, D., Bisson, L. and Carl-
son, M. (1991) Dominant and recessive suppressors that restore glu-
cose transport in a yeast snf3 mutant. Genetics 128, 505^512.
[66] O

zcan, S., Dover, J., Rosenwald, A.G., Wo l, S. and Johnston, M.


(1996) Two glucose transporters in S. cerevisiae are glucose sensors
that generate a signal for induction of gene expression. Proc. Natl.
Acad. Sci. USA 93, 12428^12432.
[67] Reifenberger, E., Freidel, K. and Ciriacy, M. (1995) Identication of
novel HXT genes in Saccharomyces cerevisiae reveals the impact of
individual hexose transporters on glycolytic ux. Mol. Microbiol. 16,
157^167.
[68] O

zcan, S., Dover, J. and Johnston, M. (1998) Glucose sensing and


signaling by two glucose receptors in the yeast Saccharomyces cere-
visiae. EMBO J. 17, 2566^2573.
[69] Marshall-Carlson, L., Celenza, J.L., Laurent, B.C. and Carlson, M.
(1990) Mutational analysis of the SNF3 glucose transporter of Sac-
charomyces cerevisiae. Mol. Cell. Biol. 10, 1105^1115.
[70] Vagnoli, P., Coons, D.M. and Bisson, L.F. (1998) The C-terminal
domain of Snf3p mediates glucose-responsive signal transduction in
Saccharomyces cerevisiae. FEMS Microbiol. Lett. 160, 31^36.
[71] Coons, D.M., Vagnoli, P. and Bisson, L.F. (1997) The C-terminal
domain of Snf3p is sucient to complement the growth defect of snf3
null mutations in Saccharomyces cerevisiae: SNF3 functions in glu-
cose recognition. Yeast 13, 9^20.
[72] Schmidt, M.C., McCartney, R.R., Zhang, X., Tillman, T.S., Solimeo,
H., Wol, S., Almonte, C. and Watkins, S.C. (1999) Std1 and Mth1
proteins interact with the glucose sensors to control glucose-regulated
gene expression in Saccharomyces cerevisiae. Mol. Cell. Biol. 19,
4561^4571.
[73] Lafuente, M.J., Gancedo, C., Jauniaux, J.-C. and Gancedo, J.M.
(2000) Mth1 receives the signal given by the glucose sensors Snf3
and Rgt2 in Saccharomyces cerevisiae. Mol. Microbiol. 35, 161^
172.
[74] Schulte, F., Wieczorke, R., Hollenberg, C.P. and Boles, E. (2000) The
HTR1 gene is a dominant negative mutant allele of MTH1 and
blocks Snf3- and Rgt2-dependent glucose signaling in yeast. J. Bac-
teriol. 182, 540^542.
[75] Hubbard, E.J.A., Jiang, R. and Carlson, M. (1994) Dosage-depen-
dent modulation of glucose repression by msn3 (Std1) in Saccharo-
myces cerevisiae. Mol. Cell. Biol. 14, 1972^1978.
[76] Tillman, T.S., Ganster, R.W., Jiang, R., Carlson, M. and Schmidt,
M.C. (1995) STD1 (MSN3) interacts directly with the TATA-binding
protein and modulates transcription of the SUC2 gene of Saccharo-
myces cerevisiae. Nucleic Acids Res. 23, 3174^3180.
[76] Schulte, F. and Ciriacy, M. (1995) HTR1/MTH1 encodes a repressor
for HXT genes. Yeast 11, S239.
[78] O

zcan, S., Freidel, K., Leuker, A. and Ciriacy, M. (1993) Glucose


uptake and catabolite repression in dominant HTR1 mutants of Sac-
charomyces cerevisiae. J. Bacteriol. 175, 5520^5528.
[79] Gamo, F.J., Lafuente, M.J. and Gancedo, C. (1994) The mutation
DGT1-1 decreases glucose transport and alleviates carbon catabolite
repression in Saccharomyces cerevisiae. J. Bacteriol. 176, 7423^7429.
[80] Blazquez, M.A., Gamo, F.J. and Gancedo, C. (1995) A mutation
aecting carbon catabolite repression suppresses growth defects in
pyruvate carboxylase mutants from Saccharomyces cerevisiae. FEBS
Lett. 377, 197^200.
[81] Chen, X.J., Wesolowski-Louvel, M. and Fukuhara, H. (1992) Glu-
cose transport in the yeast Kluyveromyces lactis. II. Transcriptional
regulation of the glucose transporter gene RAG1. Mol. Gen. Genet.
233, 97^105.
[82] Betina, S., Gorini, P., Ferrero, I. and Wesolowski-Louvel, M. (2001)
RAG4 gene encodes a glucose sensor in Kluyveromyces lactis. Genet-
ics 158, 541^548.
[83] Madi, L., McBride, S.A., Bailey, L.A. and Ebbole, D.J. (1997) rco-3,
a gene involved in glucose transport and conidiation in Neurospora
crassa. Genetics 146, 499^508.
[84] Lalonde, S., Boles, E., Hellman, H., Barker, L., Patrick, J.W.,
Frommer, W.B. and Ward, J.M. (1999) The dual function of sugar
carriers: transport and sugar sensing. Plant Cell. 11, 707^726.
[85] Barker, L., Kuhn, C., Weise, A., Schulz, A., Gebhardt, C., Hirner,
B., Hellmann, H., Schulze, W., Ward, J.M. and Frommer, W.B.
(2000) SUT2, a putative sucrose sensor in sieve elements. Plant
Cell. 12, 1153^1164.
[86] Lorenz, M.C. and Heitman, J. (1998) The MEP2 ammonium perme-
ase regulates pseudohyphal dierentiation in Saccharomyces cerevisi-
ae. EMBO J. 17, 1236^1247.
[87] Bandyopadhyay, G., Sajan, M.P., Kanoh, Y., Standaert, M.L.,
Burke, T.R.J., Quon, M.J., Reed, B.C., Dikic, I., Noel, L.E., New-
gard, C.B. and Farese, R. (2000) Glucose activates mitogen-activated
protein kinase (extracellular signal-regulated kinase) through proline-
rich tyrosine kinase-2 and the Glut1 glucose transporter. J. Biol.
Chem. 275, 40817^40826.
[88] Thevelein, J.M. and Hohmann, S. (1995) Trehalose synthase: Guard
to the gate of glycolysis in yeast? Trends Biochem. Sci. 20, 3^10.
[89] Walsh, M.C., Scholte, M., Valkier, J., Smits, H.P. and van Dam, K.
(1996) Glucose sensing and signalling properties in Saccharomyces
cerevisiae require the presence of at least two members of the glucose
transporter family. J. Bacteriol. 178, 2593^2597.
[90] Reifenberger, E., Boles, E. and Ciriacy, M. (1997) Kinetic character-
ization of individual hexose transporters of Saccharomyces cerevisiae
and their relation to the triggering mechanisms of glucose repression.
Eur. J. Biochem. 245, 324^333.
[91] Ye, L., Kruckeberg, A.L., Berden, J.A. and van Dam, K. (1999)
Growth and glucose repression are controlled by glucose transport
in Saccharomyces cerevisiae cells containing only one glucose trans-
porter. J. Bacteriol. 181, 4673^4675.
[92] Zimmermann, F.K. and Scheel, I. (1977) Mutants of Saccharomyces
cerevisiae resistant to carbon catabolite repression. Mol. Gen. Genet.
154, 75^82.
[93] Entian, K.D. and Zimmermann, F.K. (1980) Glycolytic enzymes and
intermediates in carbon catabolite repression mutants of Saccharomy-
ces cerevisiae. Mol. Gen. Genet. 177, 345^350.
[94] Michels, C.A. and Romanowski, A. (1980) Pleiotropic glucose repres-
sion-resistant mutation in Saccharomyces carlsbergensis. J. Bacteriol.
143, 674^679.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 199
[95] Entian, K.D. and Frohlich, K.U. (1984) Saccharomyces cerevisiae
mutants provide evidence of hexokinase PII as a bifunctional en-
zyme with catalytic and regulatory domains for triggering carbon
catabolite repression. J. Bacteriol. 158, 29^35.
[96] Entian, K.D., Kopetzki, E., FrEhlich, K.U. and Mecke, D. (1984)
Cloning of hexokinase isoenzyme PI from Saccharomyces cerevisiae.
PI transformants conrm the unique role of hexokinase isoenzyme
PII for glucose repression in yeasts. Mol. Gen. Genet. 198, 50^54.
[97] Ma, H., Bloom, L.M., Walsh, C.T. and Botstein, D. (1989) The
residual enzymatic phosphorylation activity of hexokinase II mu-
tants is correlated with glucose repression in Saccharomyces cerevi-
siae. Mol. Cell. Biol. 9, 5643^5649.
[98] Rose, M., Albig, W. and Entian, K.D. (1991) Glucose repression in
Saccharomyces cerevisiae is directly associated with hexose phos-
phorylation by hexokinase-PI and hexokinase-PII. Eur. J. Biochem.
199, 511^518.
[99] Witt, I., Kronau, R. and Holzer, H. (1966) Repression von Alko-
holdehydrogenase, Malatdehydrogenase, Isocitratlyase und Malat-
synthase in Hefe durch Glucose. Biochim. Biophys. Acta. 118,
522^537.
[100] Sanz, P., Nieto, A. and Prieto, J.A. (1996) Glucose repression may
involve processes with dierent sugar kinase requirements. J. Bac-
teriol. 178, 4721^4723.
[101] de Winde, J.H., Crauwels, M., Hohmann, S., Thevelein, J.M. and
Winderickx, J. (1996) Dierential requirement of the yeast sugar
kinases for sugar sensing in establishing the catabolite-repressed
state. Eur. J. Biochem. 241, 633^643.
[102] Herrero, P., Ramirez, M., Martinezcampa, C. and Moreno, F.
(1996) Identication and characterisation of two transcriptional re-
pressor elements within the coding sequence of the Saccharomyces
cerevisiae HXK2 gene. Nucleic Acids Res. 24, 1822^1828.
[103] Hohmann, S., Winderickx, J., de Winde, J.H., Valckx, D., Cob-
baert, P., Luyten, K., de Meirsman, C., Ramos, J. and Thevelein,
J.M. (1999) Novel alleles of yeast hexokinase PII with distinct eects
on catalytic activity and catabolite repression of SUC2. Microbiol-
ogy 145, 703^714.
[104] Mayordomo, I. and Sanz, P. (2001) Hexokinase PII: structural anal-
ysis and glucose signalling in the yeast Saccharomyces cerevisiae.
Yeast 18, 923^930.
[105] Vollenbroich, V., Meyer, J., Engels, R., Cardinali, G., Menezes,
R.A. and Hollenberg, C.P. (1999) Galactose induction in yeast in-
volves association of Gal80p with Gal1p or Gal3p. Mol. Gen. Gen-
et. 261, 495^507.
[106] Johnston, M. (1999) Feasting, fasting and fermenting ^ glucose
sensing in yeast and other cells. Trends Genet. 15, 29^33.
[107] Stapleton, D., Gao, G.A., Michell, B.J., Widmer, J., Mitchelhill, K.,
Teh, T., House, C.M., Witters, L.A. and Kemp, B.E. (1994) Mam-
malian 5P-AMP-activated protein kinase non-catalytic subunits are
homologs of proteins that interact with yeast Snf1 protein kinase.
J. Biol. Chem. 269, 29343^29346.
[108] Hardie, D.G. and Carling, D. (1997) The AMP-activated protein
kinase-fuel gauge of the mammalian cell ? Eur. J. Biochem. 246,
259^273.
[109] Ban uelos, M., Gancedo, C. and Gancedo, J.M. (1977) Activation by
phosphate of yeast phosphofructokinase. J. Biol. Chem. 252, 6394^
6398.
[110] Halford, N.G. and Hardie, D.G. (1998) SNF1-related protein ki-
nases: global regulators of carbon metabolism in plants? Plant Mol.
Biol. 37, 735^748.
[111] Sugden, C., Crawford, R.M., Halford, N.G. and Hardie, D.G.
(1999) Regulation of spinach SNF1-related (SnRK1) kinases by
protein kinases and phosphatases is associated with phosphorylation
of the T loop and is regulated by 5P-AMP. Plant J. 19, 433^439.
[112] Toroser, D., Plaut, Z. and Huber, S.C. (2000) Regulation of a plant
SNF1-related protein kinase by glucose-6-phosphate. Plant Physiol.
123, 403^412.
[113] Randez-Gil, F., Herrero, P., Sanz, P., Prieto, J.A. and Moreno, F.
(1998) Hexokinase PII has a double cytosolic-nuclear localisation in
Saccharomyces cerevisiae. FEBS Lett. 425, 475^478.
[114] Herrero, P., Martinez Campa, C. and Moreno, F. (1998) The hexo-
kinase 2 protein participates in regulatory DNA^protein complexes
necessary for glucose repression of the SUC2 gene in Saccharomyces
cerevisiae. FEBS Lett. 434, 71^76.
[115] Behlke, J., Heidrich, K., Naumann, M., Muller, E.C., Otto, A.,
Reuter, R. and Kriegel, T. (1998) Hexokinase 2 from Saccharomyces
cerevisiae: Regulation of oligomeric structure by in vivo phosphor-
ylation at serine-14. Biochemistry 37, 11989^11995.
[116] Alms, G.R., Sanz, P., Carlson, M. and Haystead, T.A.J. (1999)
Reg1p targets protein phosphatase 1 to dephosphorylate hexokinase
II in Saccharomyces cerevisiae: characterizing the eects of a phos-
phatase subunit on the yeast proteome. EMBO J. 18, 4157^4168.
[117] Beullens, M., Mbonyi, K., Geerts, L., Gladines, D., Detremerie, K.,
Jans, A.W.H. and Thevelein, J.M. (1988) Studies on the mechanism
of the glucose-induced cAMP signal in glycolysis and glucose repres-
sion mutants of the yeast Saccharomyces cerevisiae. Eur. J. Biochem.
172, 227^231.
[118] Mbonyi, K., Beullens, M., Detremerie, K., Geerts, L. and Thevelein,
J.M. (1988) Requirement of one functional RAS gene and inability
of an oncogenic ras-variant to mediate the glucose-induced cAMP
signal in the yeast Saccharomyces cerevisiae. Mol. Cell. Biol. 8,
3051^3057.
[119] Munder, T. and Ku ntzel, H. (1989) Glucose-induced cAMP signal-
ing in Saccharomyces cerevisiae is mediated by the CDC25 protein.
FEBS Lett. 242, 341^345.
[120] Van Aelst, L., Boy-Marcotte, E., Camonis, J.H., Thevelein, J.M.
and Jacquet, M. (1990) The C-terminal part of the CDC25 gene
product plays a key role in signal transduction in the glucose-in-
duced modulation of cAMP level in Saccharomyces cerevisiae. Eur.
J. Biochem. 193, 675^680.
[121] Van Aelst, L., Jans, A.W.H. and Thevelein, J.M. (1991) Involve-
ment of the CDC25 gene product in the signal transmission pathway
of the glucose-induced RAS-mediated cAMP signal in the yeast
Saccharomyces cerevisiae. J. Gen. Microbiol. 137, 341^349.
[122] Colombo, S., Ma, P., Cauwenberg, L., Winderickx, J., Crauwels,
M., Teunissen, A., Nauwelaers, D., de Winde, J.H., Gorwa, M.-
F., Colavizza, D. and Thevelein, J.M. (1998) Involvement of distinct
G-proteins, Gpa2 and Ras, in glucose- and intracellular acidica-
tion-induced cAMP signalling in the yeast Saccharomyces cerevisiae.
EMBO J. 17, 3326^3341.
[123] Nakafuku, M., Obara, T., Kaibuchi, K., Miyajima, I., Miyajima,
A., Itoh, H., Nakamura, S., Arai, K., Matsumoto, K. and Kaziro,
Y. (1988) Isolation of a second yeast Saccharomyces cerevisiae gene
(GPA2) coding for guanine nucleotide-binding regulatory protein:
studies on its structure and possible functions. Proc. Natl. Acad. Sci.
USA 85, 1374^1378.
[124] Papasavvas, S., Arkinstall, S., Reid, J. and Payton, M. (1992) Yeast
alpha-mating factor receptor and G-protein-linked adenylyl cyclase
inhibition requires RAS2 and GPA2 activities. Biochem. Biophys.
Res. Commun. 184, 1378^1385.
[125] Yun, C.W., Tamaki, H., Nakayama, R., Yamamoto, K. and Ku-
magai, H. (1997) G-protein coupled receptor from yeast Saccharo-
myces cerevisiae. Biochem. Biophys. Res. Commun. 240, 287^292.
[126] Xue, Y., Batlle, M. and Hirsch, J.P. (1998) GPR1 encodes a putative
G protein-coupled receptor that associates with the Gpa2p G sub-
unit and functions in a Ras-independent pathway. EMBO J. 17,
1996^2007.
[127] Ansari, K., Martin, S., Farkasovsky, M., Ehbrecht, I.M. and Kunt-
zel, H. (1999) Phospholipase C binds to the receptor-like GPR1
protein and controls pseudohyphal dierentiation in Saccharomyces
cerevisiae. J. Biol. Chem. 274, 30052^30058.
[128] Versele, M., Lemaire, K. and Thevelein, J.M. (2001) Sex and sugar
in yeast: two distinct GPCR systems. EMBO Rep. 2, 574^579.
[129] Versele, M., de Winde, J.H. and Thevelein, J.M. (1999) A novel
regulator of G-protein signalling in yeast, Rgs2, downregulates glu-
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 200
cose-activation of the cAMP pathway through direct inhibition of
Gpa2. EMBO J. 18, 5577^5591.
[130] Kubler, E., Mosch, H.U., Rupp, S. and Lisanti, M.P. (1997) Gpa2p,
a G-protein alpha-subunit, regulates growth and pseudohyphal de-
velopment in Saccharomyces cerevisiae via a cAMP-dependent
mechanism. J. Biol. Chem. 272, 20321^20323.
[131] Vanhalewyn, M., Dumortier, F., Debast, G., Colombo, S., Ma, P.,
Winderickx, J., Van Dijck, P. and Thevelein, J.M. (1999) A muta-
tion in Saccharomyces cerevisiae adenylate cyclase, Cyr1
K1876M
, spe-
cically aects glucose- and acidication-induced cAMP signalling
and not the basal cAMP level. Mol. Microbiol. 33, 363^376.
[132] Ma, P., Goncalves, T., Maretzek, A., Loureiro-Dias, M.C. and
Thevelein, J.M. (1997) The lag phase rather than the exponential-
growth phase on glucose is associated with a higher cAMP level in
wild-type and cAPK-attenuated strains of the yeast Saccharomyces
cerevisiae. Microbiology 143, 3451^3459.
[133] Lorenz, M.C. and Heitman, J. (1997) Yeast pseudohyphal growth is
regulated by GPA2, a G protein K homolog. EMBO J. 16, 7008^
7018.
[134] Lorenz, M.C., Pan, X., Harashima, T., Cardenas, M.E., Xue, Y.,
Hirsch, J.P. and Heitman, J. (2000) The G protein-coupled receptor
Gpr1 is a nutrient sensor that regulates pseudohyphal dierentiation
in Saccharomyces cerevisiae. Genetics 154, 609^622.
[135] Jiang, Y., Davis, C. and Broach, J.R. (1998) Ecient transition to
growth on fermentable carbon sources in Saccharomyces cerevisiae
requires signaling through the Ras pathway. EMBO J. 17, 6942^
6951.
[136] Bhattacharya, S., Chen, L., Broach, J.R. and Powers, S. (1995) Ras
membrane targeting is essential for glucose signaling but not for
viability in yeast. Proc. Natl. Acad. Sci. USA 92, 2984^2988.
[137] Mintzer, K.A. and Field, J. (1999) The SH3 domain of the S cere-
visiae Cdc25p binds adenylyl cyclase and facilitates Ras regulation
of cAMP signalling. Cell. Signal. 11, 127^135.
[138] Boles, E., Zimmermann, F.K. and Thevelein, J.M. (1997) In: Yeast
Sugar Metabolism: Biochemistry, Genetics, Biotechnology and Ap-
plications (Zimmermann, F.K. and Entian, K.-D., Eds.), pp. 379^
407. Technomic Publishing Co., Lancaster, PA.
[139] Schaa, I., Heinisch, J. and Zimmermann, F.K. (1989) Overproduc-
tion of glycolytic enzymes in yeast. Yeast 5, 285^290.
[140] Muller, S., Zimmermann, F.K. and Boles, E. (1997) Mutant studies
of phosphofructo-2-kinases do not reveal an essential role of fruc-
tose-2,6-bisphosphate in the regulation of carbon uxes in yeast
cells. Microbiology 143, 3055^3061.
[141] Galazzo, J.L. and Bailey, J.E. (1990) Fermentation pathway kinetics
and metabolic ux control in suspended and immobilized Saccha-
romyces cerevisiae. Enz. Microb. Technol. 12, 162^172.
[142] Blazquez, M.A., Lagunas, R., Gancedo, C. and Gancedo, J.M.
(1993) Trehalose-6-phosphate, a new regulator of yeast glycolysis
that inhibits hexokinases. FEBS Lett. 329, 51^54.
[143] Bonini, B.M., Van Vaeck, C., Larsson, C., Gustafsson, L., Ma, P.,
Winderickx, J., Van Dijck, P. and Thevelein, J.M. (2000) Expression
of Escherichia coli otsA in a Saccharomyces cerevisiae tps1 mutant
restores trehalose-6-phosphate levels and partly restores growth and
fermentation with glucose and control of glucose inux into glycol-
ysis. Biochem. J. 350, 261^268.
[144] Van Vaeck, C., Wera, S., Van Dijck, P. and Thevelein, J.M. (2001)
Analysis and modication of trehalose-6-phosphate levels in the
yeast Saccharomyces cerevisiae using Bacillus subtilis phosphotreha-
lase. Biochem. J. 353, 157^162.
[145] Van Aelst, L., Hohmann, S., Bulaya, B., de Koning, W., Sierkstra,
L., Neves, M.J., Luyten, K., Alijo, R., Ramos, J., Coccetti, P.,
Martegani, E., de Magalhaes-Rocha, N.M., Brandao, R.L., Van
Dijck, P., Vanhalewyn, M., Durnez, P., Jans, A.W.H. and Theve-
lein, J.M. (1993) Molecular cloning of a gene involved in glucose
sensing in the yeast Saccharomyces cerevisiae. Mol. Microbiol. 8,
927^943.
[146] Matschinsky, F.M., Glaser, B. and Magnusson, M.A. (1998) Pan-
creatic beta-cell glucokinase: Closing the gap between theoretical
concepts and experimental realities. Diabetes 47, 307^315.
[147] Vaulont, S., Vasseur-Cognet, M. and Kahn, A. (2000) Glucose reg-
ulation of gene transcription. J. Biol. Chem. 275, 31555^31558.
[148] Sheen, J., Zhou, L. and Jang, J.-C. (1999) Sugars as signaling mol-
ecules. Curr. Opin. Plant Biol. 2, 410^418.
[149] Smeekens, S. (1998) Sugar regulation of gene expression in plants.
Curr. Opin. Plant Biol. 1, 230^234.
[150] Fabrizio, P., Pozza, F., Pletcher, S.D., Gendron, C.M. and Longo,
V.D. (2001) Regulation of longevity and stress resistance by Sch9 in
yeast. Science 292, 288^290.
[151] Guarente, L. and Kenyon, C. (2000) Genetic pathways that regulate
ageing in model organisms. Nature 408, 255^262.
FEMSYR 1457 5-6-02
F. Rolland et al. / FEMS Yeast Research 2 (2002) 183^201 201

You might also like