You are on page 1of 30

MICROBIOLOGY AND MOLECULAR BIOLOGY REVIEWS, Mar. 2006, p. 253–282 Vol. 70, No.

1
1092-2172/06/$08.00⫹0 doi:10.1128/MMBR.70.1.253–282.2006
Copyright © 2006, American Society for Microbiology. All Rights Reserved.

Glucose Signaling in Saccharomyces cerevisiae


George M. Santangelo*
Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi 39406-5018

INTRODUCTION .......................................................................................................................................................253
NATURE OF THE SIGNAL......................................................................................................................................254
Signaling without Extracellular Sensing, Transport, or Phosphorylation of Glucose..................................254
Redundant but Unified Glucose Signaling Pathways ........................................................................................255
Early Steps in Signal Transduction .....................................................................................................................256
The Ras/cAMP/PKA pathway............................................................................................................................256
The Gpr1/Gpa2 pathway ....................................................................................................................................258

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


Activation of Ras and Gpa2 by glucose ...........................................................................................................258
(i) Extracellular sensing ................................................................................................................................258
(ii) Intracellular sensing................................................................................................................................258
Extracellular sensing by Snf3/Rgt2 ..................................................................................................................259
SIGNAL RECEPTION: GLUCOSE REPRESSION ..............................................................................................259
Involvement of the Hexokinase Hxk2 in the Glucose Response ......................................................................259
Snf1-Mediated Signal Transduction in Glucose Repression and Derepression ............................................260
Dueling Snf1 kinase and Glc7 phosphatase mediate glucose regulation....................................................260
Transmission of the Ras/cAMP-dependent glucose signal to Reg1/Glc7 and Snf1/Snf4..........................263
DNA-bound repressors controlled by Snf1......................................................................................................263
DNA-bound activators controlled by Snf1.......................................................................................................265
SIGNAL RECEPTION: GLUCOSE INDUCTION.................................................................................................265
Snf3/Rgt2 Signaling of the Rgt1 Repressor Complex ........................................................................................265
The Cell Cycle Connection ....................................................................................................................................267
Transcriptional Induction of Glycolytic and Ribosomal Protein Genes .........................................................268
The Repressor/Activator Theme in Glucose Regulation....................................................................................269
Reverse Recruitment: Rap1/Gcr1 Activation at the Nuclear Periphery..........................................................269
GENE REGULATION VIA REVERSE RECRUITMENT.....................................................................................272
A New Model for Eukaryotic Transcriptional Activation and Repression .....................................................272
Glucose May Signal a Highly Networked and Structurally Stable Perinuclear Machine............................273
ACKNOWLEDGMENTS ...........................................................................................................................................274
REFERENCES ............................................................................................................................................................274

INTRODUCTION repression and glucose induction). The response to glucose


also involves direct modification of metabolic pathways (termed
Jacques Monod’s seminal discovery of the diauxic growth of
glucose or catabolite inactivation) and other processes such as
bacterial cultures (243) demonstrated that microbes are par-
mRNA turnover (7, 76, 103, 121, 153, 154, 156, 161, 166,
ticularly adept at targeting and responding to glucose, the most
171–173, 232, 294, 307, 329, 334, 335, 337, 391). Although the
abundant monosaccharide in nature. A wealth of subsequent
latter phenomena are outside the scope of this review, they
work has shown that virtually all cells possess a sophisticated
appear to be mediated by the same two central signal trans-
genetic program that responds to this rich source of carbon
duction pathways, Ras/cyclic AMP (cAMP)/protein kinase A
and energy. Study of the glucose response in eukaryotes has
(PKA) and Snf3/Rgt2/Yck, which are described below in
been greatly aided by the development of the malleable bud-
detail.
ding yeast Saccharomyces cerevisiae as a model system (see,
The recent application of yeast genomics and proteomics is
e.g., references 361 and 417). S. cerevisiae shares with complex
a first step towards comprehensive characterization of the glu-
multicellular eukaryotes many of the signal transduction com-
cose response. For example, we now know that within 20 min
ponents that detect glucose, transduce the corresponding sig-
of glucose addition, ⬃20% of the roughly 6,200 genes in S.
nals to the interior of the cell, and make the needed adjust-
ments to cellular metabolism and gene expression profiles. cerevisiae exhibit a greater-than-threefold change in expression
This review will focus on the cytoplasmic regulators that (either induction or repression), and ⬃40% show at least a
transmit the glucose signal from the plasma membrane into the twofold change (408). Glucose repression largely affects genes
yeast nucleus, as well as the nuclear apparatus that mediates encoding enzymes involved in respiration or alternative carbon
the global transcriptomic response to the sugar (both glucose source metabolism, and glucose induction stimulates transcrip-
tion of glycolytic genes and ribosomal protein (RP) genes (see
below). Genomic and proteomic analyses are particularly use-
* Mailing address: Department of Biological Sciences, University of
ful when supplemented with high-throughput genetic tech-
Southern Mississippi, Hattiesburg, MS 39406-5018. Phone: (601) 266- niques such as synthetic genetic array analysis, which allows the
6080. Fax: (601) 266-5068. E-mail: George.Santangelo@mfgn.usm.edu. identification of synthetic phenotypes by simultaneous screen-

253
254 SANTANGELO MICROBIOL. MOL. BIOL. REV.

ing of the entire collection of mutants in which each of the


⬃4,700 nonessential yeast genes has been deleted (373).
The continued pursuit of such large data sets, in combina-
tion with the now-facile elimination, overexpression, or point
mutation of relevant yeast regulators, should eventually permit
the delineation of a global wiring diagram that mirrors its
biological template when virtually challenged with a wide
range of concentrations of the sugar. For such a computational
model to be valid, it must at a minimum be informed by (i) an
accurate molecular map of cytoplasmic and nuclear substruc-
ture, (ii) a detailed diagram of the physical and functional
connections that link the relevant signal transduction compo-
nents to each other and to adjacent networks, and (iii) the
temporal progression of the response following the appearance

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


of glucose. Here I review our progress toward that ambitious
goal and attempt to identify the most important issues that
require further clarification.

NATURE OF THE SIGNAL


Two recent surprises uncovered by global analyses of Saccha-
romyces cerevisiae are the extent to which the glucose induction
and repression mechanisms appear to overlap and the fact that
yeast cells can be tricked into generating the full-blown response
(both induction and repression) in the complete absence of glu-
cose. These and other recent developments place G-protein sig-
naling, in particular the Ras/cAMP/PKA pathway, front and cen-
ter among the cellular components that participate in early events
in the glucose response.

Signaling without Extracellular Sensing, Transport,


or Phosphorylation of Glucose
A recent microarray experiment from the Broach laboratory
has clearly demonstrated the universality of induction/repres-
sion overlap in glucose signal transduction. A unified transcrip-
FIG. 1. Wild-type versus Ras2 mimicry of the glucose response.
tome-wide response (both upshifted and downshifted gene ex- Each curve represents the expression profile of genes exhibiting a
pression) was observed in yeast cells by stimulating early steps twofold or greater change, both within 20 min of glucose addition and
in the glucose signaling pathway; a clever experimental scheme within 20 min of the appearance of activated Ras2. The average values
allowed the generation and detection of this comprehensive for 250 such induced genes (upper graph) and repressed genes (lower
glucose response without addition of the sugar (408). The yeast graph) are shown (see supplemental data associated with reference
208); error bars represent standard errors of the means. Squares,
transcriptome was analyzed after heterologous induction of expression profile of wild-type cells following glucose addition; filled
activated alleles of Ras2 or Gpa2, signal transmitters that act in triangles, expression profile of cells containing Gal-regulated Ras2*
parallel early in the cAMP-dependent protein kinase (PKA) (activated Ras) following addition of galactose; open triangles, expres-
pathway (see below). Astonishingly, virtually all of the changes sion profile of cells containing Gal-regulated Ras2* (activated Ras) in
a tpkw (weak PKA) background following addition of galactose. It is
in transcript levels after addition of glucose (both up-regula- important to note that the ⬎100-fold induction and repression ob-
tion and down-regulation) were also observed in the absence of served for a small subset of glucose regulated genes (e.g., HXT genes;
glucose following expression of the activated form of Ras2. Of see reference 272) is not seen within the 60-minute time frame of this
the genes that showed at least a threefold change in expression data set.
within an hour following glucose addition, almost all (92%)
changed at least twofold in the same direction after Ras acti-
vation (Fig. 1; Table 1). The result was similar with activated dent) appear to be fully capable of transducing the glucose
Gpa2, though the intensity of the transcriptional response was signal. Little is known about the PKA-independent mecha-
reduced relative to that seen with activated Ras. Those authors nism, although it presumably targets a similar set of down-
also demonstrated that all of the changes in transcript levels stream factors in the absence of normal PKA catalytic activity.
resulting from Ras2 and Gpa2 activation were mediated by The observed transcriptome-wide up-regulation and down-
PKA (408) (Fig. 1). Importantly, since much of this transcrip- regulation of gene expression in response to activated Ras was
tional reprogramming can also be accomplished in the absence particularly surprising, since it had long been assumed (despite
of signaling through cAMP, redundant overlapping pathways early hints to the contrary [see, e.g., reference 233]) that the
(Ras2/Gpa2/PKA dependent and Ras2/Gpa2/PKA indepen- primary glucose repression pathways in yeast are distinct from
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 255

TABLE 1. Genes that respond similarly to both glucose addition and Ras2 activation a
Induced genes Repressed genes

Protein Energy reserve Protein Stress


Transcription Transportc
biosynthesisb metabolismd turnover response

RPS9A RPA43 NUP1 BCY1 PEP4 HSP30


RPS22B RPA49 NUP100 SDC25 RPN2 SSE2
RPS27A RPB5 NUP133 GAL83 RPN3 SSA3
RPL7B RPB9 NUP170 PHO85 RPN5 SSA4
RPL14B RPC40 MEX67 TPS1 RPN6 SSD1
RPL17B RPC82 RNA1 CIT3 RPN7 STO1
TIF11 RPO26 GSP1 GPD1 RPT1 CDC55
TIF3 RPO31 PSE1 MDH3 RPT3 UBC5
TIF34 RRP43 SXM1 PGM2 RPT4 PRE1
TEF4 RRP45 ALR21 HXT10 RPT5 PRE3
CDC60 RRP5 FET3 MRPL23 DOA1 GTS1
ARG8 ARG80 FET4 MRPL32 DOA4 CRS5

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


ARO7 BAS1 FKS1 PET112 APG13 CTA1
ARO8 DBP2 HNM1 PET117 APG14 GRX2
ASP1 DBP3 HXT2 PET191 APC9 TRX2
HIS1 GCR2 HXT4 PET54 HRD1 MDR1
HIS3 IFH1 LYP1 COQ1 HRD3 PAU1
ILS1 HMALPHA1 NPL3 COQ3 PEX14 PAU2
ILV1 IMP4 PHO84 COX10 MPD2 PAU3
LYS4 LEU3 ZRT1 COX11 SBA1 PPZ2
a
See supplemental data associated with reference 408; a grand total of 758 genes exhibited a twofold or greater change in expression both within 20 min of glucose
addition and within 20 min of the appearance of activated Ras2. Gene categories are based on Munich Information Center for Protein Sequences functional
classification; a representative list from each category is shown. A total of 329 and 429 genes were in the induced and repressed categories, respectively.
b
Includes genes encoding translational components (including ribosomal proteins), rRNA-processing factors, and amino acid biosynthetic enzymes.
c
Includes genes encoding nucleocytoplasmic transport factors and plasma membrane transporters.
d
Includes genes encoding factors involved in carbon storage and respiration (including mitochondrial factors).

glucose induction and are also essentially Ras independent (44, The Snf1 serine-threonine kinase represents another exam-
45, 118, 121, 123, 178, 315, 335, 365). Further, stimulation of ple of downstream regulatory overlap; Snf1 activates genes
the glucose response in the absence of either transport or that are repressed in glucose-containing media but are dere-
phosphorylation of the sugar (408) is particularly significant. pressed and needed for growth in the presence of nonferment-
Transport of glucose and its conversion to glucose-6-phosphate able carbon sources (46). Recent transcriptomic and proteomic
by hexokinases had been explored as a potentially essential analyses of snf1 cells revealed a previously unnoticed upshift in
feature of the response (121, 314, 316) and now appears to at expression of genes (e.g., ADH1) involved in glucose catabo-
most participate in redundant signaling that operates through lism (hereafter the term “glycolytic genes” will be used for this
or in parallel with the cAMP-PKA pathway. Whatever extra- group) (432). Thus, in addition to its well-established role in
cellular or intracellular glucose sensing is needed to activate derepressing glucose-repressed genes, Snf1 also somehow re-
Ras or Gpa2 is therefore sufficient for both glucose induction presses transcription of glucose-induced genes (including HXT1)
and repression. This interpretation has the added advantage of (372). Snf1 repression of glycolytic gene expression has been
bringing our assessment of glucose-stimulated signal transduc- further substantiated by proteomic analysis of cells lacking Snf4,
tion in yeast cells closer to what is known about the analogous the ␥ subunit of the Snf1 kinase complex (140).
response in bacterial and mammalian cells (for reviews, see A final example of downstream regulatory overlap is the
references 35, 129, and 322). phosphoprotein Gcr1, which was first identified as an activator
of glycolytic genes (13, 63, 64, 152). Transcriptomics and other
analyses have confirmed that expression of numerous glucose-
Redundant but Unified Glucose Signaling Pathways
repressed genes is derepressed in the absence of Gcr1 at high
The first hint of overlap between glucose induction and glucose concentrations, suggesting a new role for this regulator
repression resulted from analysis of regulators that are “down- in repression of transcription (327, 383; K. Barbara and G. M.
stream” in the signaling cascade, i.e., factors that for the most Santangelo, unpublished data).
part function within the yeast nucleus. For example, the Rgt1 The roles of Rgt1, Hxk2, Snf1/Snf4, and Gcr1, as well as
repressor blocks transcription of HXT1 through HXT4 (4 of numerous other repressor/activator polypeptides that partici-
the 18 glucose transporter genes in S. cerevisiae) (see refer- pate in the glucose response, are presented in greater detail in
ences 118 and 272 for reviews). At high glucose concentrations, later sections of this review. In addition to these specific reg-
although its contact with the HXT1 promoter can no longer be ulators, at least eight components of the general transcription
detected, Rgt1 is converted into an activator through hyper- machinery that interact with the carboxy-terminal domain of
phosphorylation (142, 186, 250). The hexokinase Hxk2, long RNA polymerase II (Gal11, Sin4, Rgr1, Rox3, Srb8, Ssn2,
known to participate in glucose repression, is also required for Ssn3, and Ssn8) have been found to play both positive and
glucose induction of HXT1 (231, 271). negative regulatory roles; the function of many of these factors
256 SANTANGELO MICROBIOL. MOL. BIOL. REV.

is known to be influenced by glucose signaling (43, 195). Thus, failure of ras2⌬ cells to grow on nonfermentable carbon
the overlapping positive and negative circuitry in the glucose sources (363), suggesting that the latter defect is due to the fact
response may extend from the plasma membrane through to that Ras1 is more weakly expressed than Ras2 (249, 361),
the ultimate choice of induction or shutoff of transcription initi- rather than a specific function that maps to the divergent and
ation of each glucose-regulated gene by RNA polymerase II. slightly extended C terminus of Ras2.
The induction/repression duality of signal recipients such as Important effector molecules modulate Ras activity during
Rgt1, Hxk2, Snf1/Snf4, Gcr1, and other transcription factors the glucose response; posttranslational modification and mem-
clearly requires further characterization of the precise mecha- brane localization of Ras are two important features of these
nisms involved. Nonetheless, together with the transcriptomic regulatory inputs. For example, Ras farnesylation contributes
analysis of activated Ras, it suggests the existence of a more to nucleotide exchange by the GEF Cdc25 (34, 73, 134);
unified and interdigitated response to glucose than had previ- CDC25 is an essential gene and was originally identified by a
ously been appreciated. Although it now seems certain that screen for mutations that cause G1 arrest (139). A second yeast
transmission of the glucose signal is redundant (408), those RasGEF that can substitute for Cdc25 when overexpressed is
signaling pathways may converge on only a relatively small encoded by the dispensable SDC25 gene (31) (note that this

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


group of transcription factors in the yeast nucleus (see below). gene was originally named SCD25 [79]).
Coordinated remodeling of the nuclear regulatory apparatus in Ras stimulation by the RasGEFs in turn stimulates produc-
response to glucose might therefore involve a parsimonious tion of cAMP by the essential product of the adenylate cyclase
series of alterations in key regulators. Although for the sake of gene (CYR1) (229). Inactivation of Ras, resulting in lower
clarity and historical perspective the discussion of nuclear reg- cAMP levels, is facilitated by the GAPs Ira1 and Ira2 (358,
ulators is divided into separate sections of this review, in reality 360) (Fig. 2). Presumably due to their competing effects on Ras
the mechanisms of “glucose repression” and “glucose induc- activation, IRA1 deletion rescues the lethality caused by dele-
tion” may be no more segregated than the two faces of a coin. tion of the RasGEF CDC25 (359). Low-affinity and high-affin-
ity cAMP phosphodiesterases in S. cerevisiae (Pde1 and Pde2,
respectively) also antagonize the Ras signal through enzymatic
Early Steps in Signal Transduction removal of cAMP; in the presence of exogenous cAMP, dele-
Given the sufficiency of Ras or Gpa2 activation to produce tion of PDE2 restores growth to ras1⌬ ras2⌬ strains (266, 420).
the transcriptome-wide spectrum of changes in response to The Ras C terminus also undergoes functionally significant
glucose, it is important first to review what is known about posttranslational modification. Yeast Ras2 is normally pal-
these factors, their respective signaling pathways, and how they mitoylated via a thioester linkage at cysteine 318 and farnesy-
are activated immediately following appearance of the sugar. lated via a thioether linkage at cysteine 319. Blocking both
The Ras/cAMP/PKA pathway. Ras proteins are monomeric modifications with the double point mutation C318S C319S
GTPases that function as switches; these so-called G proteins results in a nonfunctional molecule (92). Farnesylation is re-
are inactive in the GDP-bound state and active when GTP is quired for palmitoylation and all subsequent modifications,
bound. The switch from the active to the inactive form involves which include removal of the three C-terminal residues by
hydrolysis of bound GTP by the intrinsic GTPase and is stim- proteolytic cleavage and methyl esterification of the revealed
ulated by GTPase-activating proteins (GAPs). The reverse (in- C-terminal cysteine 319 (19).
active-to-active) switch requires replacement of bound GDP The palmitoyl moiety anchors Ras to the cytoplasmic face of
with GTP, which is normally accomplished with the aid of the yeast plasma membrane (199); blocking addition of palmi-
guanine nucleotide exchange factors (GEFs). Activating mu- tate via the single point mutation C318S causes mislocalization
tations in Ras polypeptides (e.g., human rasVal12 or yeast of the mutated Ras2 protein to the cytoplasm (176). Ras pal-
Ras2Val19) increase GTP association in a GEF-independent mitoylation is reversible and therefore may be a regulatory
fashion and are responsible for many human cancers (78, 419). step. This is of particular interest because mutation of pal-
Ras mutations are found in approximately 30% of human mitoylation sites abrogates both the glucose response in yeast
tumors; the frequency of oncogenic forms of Ras is as high as cells (see below) and transformation of mammalian cells by
50% or 90% in colorectal or pancreatic tumors, respectively oncogenic forms of Ras (418).
(345). Although it remains dimly understood, it seems likely The appearance of glucose thus generates one or more sig-
that there is an important connection between these recent nals that converge on membrane-bound Ras and its modifiers
data and a very old observation that altered glucose metabo- (in particular the RasGEFs and RasGAPs). This glucose sig-
lism is a hallmark of cancerous cells in solid tumors (71, 179). naling results in a rapid but transient spike in the level of
The polypeptides encoded by the two RAS genes in S. intracellular cAMP, which increases 5- to 50-fold within 1 to 2
cerevisiae, Ras1 (309 residues) and Ras2 (322 residues), are min of glucose addition and returns to near-basal levels within
⬎70% identical overall and approximately 90% identical over 20 min. When cells express the nonpalmitoylated cytoplasmic
the N-terminal 180 residues (291). Growth on glucose is unaf- Ras2C318S form as their only Ras protein, they fail to exhibit
fected by the absence of either Ras1 or Ras2, whereas loss of the glucose-induced cAMP spike and are defective in transi-
both causes arrest in the G1 phase of the cell cycle (362, 367). tioning to rapid growth upon addition of glucose (160). Thus,
These early findings were suggestive of a connection to nutri- glucose-regulated cAMP production appears to require attach-
ent sensing, since nutrient-deprived cells also arrest in G1 ment of Ras to the yeast plasma membrane. Interestingly,
phase. Overexpression of RAS1 was also found to suppress the membrane localization of yeast Ras2 was recently shown to
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 257

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


FIG. 2. Cytoplasmic events in PKA signaling. Glucose regulation relies upon the intracellular signaling molecule (“second messenger”) cAMP.
GTP-bound G proteins (Ras and Gpa2) bind independently to adenylate cyclase (Cyr1) and stimulate its production of cAMP. The monomeric
Ras G proteins are anchored in the plasma membrane via a posttranslationally added palmitoyl moiety (red squiggle). RasGEFs (Cdc25 and Sdc25)
and RasGAPs (Ira1 and Ira2) are also present in the Ras/Cyr1 complex and regulate adenylate cyclase by controlling the Ras switch (see text).
The seven-transmembrane polypeptide Gpr1 acts upstream of Gpa2 in glucose signaling and is a member of the G protein-coupled receptor
(GPCR) superfamily. Gpa2 was identified as a result of its similarity to the mammalian G␣ subunits of heterotrimeric G proteins, and the putative
G␤ subunits Gpb1 and Gpb2 were found via their interaction with Gpa2; a corresponding ␥ subunit has yet to be identified (question mark). A
Gpa2GEF also awaits identification; Rgs2 is a known Gpa2GAP. Phosphodiesterases (Pde1 and Pde2) antagonize glucose signaling via enzymatic
inactivation of cAMP (conversion to AMP). The PKA tetramer is the regulatory target of cAMP. While bound to the kinase subunits (TPK), the
regulatory Bcy1 subunits keep PKA in an inactive state; cAMP activates the catalytic subunits by binding to Bcy1 subunits and promoting
dissociation of the complex (dashed arrow).

occur despite disruption of the classical secretory pathway that restores growth to ras1⌬ ras2⌬ strains (369). PKA is a het-
normally mediates translocation from the endoplasmic reticu- erotetramer consisting of two catalytic (Tpk) subunits and two
lum to the plasma membrane (92). regulatory (Bcy1) subunits (Fig. 2); addition of regulatory sub-
Additional signal transduction components appear to con- units to catalytic subunits in vitro converts protein kinase ac-
tribute to glucose signaling by contacting adenylate cyclase tivity into a completely cAMP-dependent form (151, 177, 370).
(Cyr1); such Cyr1 protein-protein interactions identified thus Interestingly, the subcellular localization of PKA catalytic
far include those with (i) an N-terminal SH3 domain in the and regulatory subunits appears to respond to glucose signal-
RasGEF Cdc25 (237), (ii) the farnesylated Ras2 C terminus ing; Bcy1 is present in both the cytoplasm and nucleus in the
(73, 74, 184, 199, 338, 355), (iii) the C terminus of the putative absence of glucose but is exclusively nuclear in its presence
cochaperonin Sgt1 (95), (iv) the N terminus of the ancillary (129, 130). Cross talk from the TOR (target of rapamycin)
factor Srv2 (CAP) via coiled-coil formation (267), and (v) the pathway, another major set of nutrient-responsive signaling
RasGAP Ira1, which mediates peripheral association of ade- components, may help to maintain PKA activity in part by
nylate cyclase with the yeast plasma membrane (241). regulating nuclear translocation of Tpk1 (330). Although it has
It seems likely that the sole purpose of the glucose-induced yet to be demonstrated that the observed translocation of PKA
spike in cAMP levels is to block the inhibitory effect of the subunits between compartments plays an important role in
BCY1-encoded regulatory subunit on the catalytic subunits of glucose signaling in yeast cells, targeting of PKA regulatory
PKA, encoded redundantly by TPK1, TPK2, and TPK3; over- substrates would appear to undergo a qualitative and/or quan-
expression of any of these three genes, or deletion of BCY1, titative shift in response to the appearance or disappearance of
258 SANTANGELO MICROBIOL. MOL. BIOL. REV.

the sugar. Further progress in this area is complicated by the growth rates of gpr1⌬ ras2⌬, gpa2⌬ ras2⌬, and gpa2⌬ gpr1⌬
fact that signaling via each of the three different catalytic sub- ras2⌬ strains are essentially identical, and introduction of ei-
units of PKA (Tpk1, Tpk2, and Tpk3) results in a distinct ther multicopy or activated GPA2 suppresses the defect of the
pattern of downstream gene expression readouts (309). gpr1⌬ ras2⌬ strain (428). Deletion of GPR1 diminishes (al-
Although active PKA is thought to phosphorylate proteins though does not eliminate) genome-wide transcriptional in-
involved in transcription, energy metabolism, and cell cycle duction and repression in response to glucose (408). The sim-
progression (129), further study is required to identify the plest explanation for these data is that Gpr1 is the only
exact targets and sequence of protein translocation and phos- receptor coupled to Gpa2 and that it too contributes to acti-
phorylation events that transmit the glucose regulatory signal vation of the cAMP pathway in response to glucose (190).
to the yeast nucleus. Significantly, mutations impairing specific Although Gpa2 was originally thought to function upstream
components of the RNA polymerase II transcriptional machin- of the Sch9 kinase (368, 428), it was subsequently found that
ery are synthetically lethal with the hyperactive Ras2Val19 allele SCH9 deletion is synthetically lethal with gpr1⌬, gpa2⌬, or
(157), and PKA can phosphorylate a component of the Srb ras2⌬, suggesting that Sch9 acts instead in the pseudohyphal
complex (Ssn2) both in vitro and in vivo (55). The latter may be differentiation pathway that is parallel to both Gpr1/Gpa2 and

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


one of but a few Ras/cAMP/PKA-mediated modifications in Ras (217). Sch9 may nevertheless participate somehow in glu-
nuclear factors needed to accomplish the broad sweep of al- cose signaling, since (i) it is the yeast ortholog of a component
terations to the yeast transcriptome upon glucose addition or of the mammalian insulin response pathway (Aft/protein ki-
depletion. nase B), (ii) it shares similarity with PKA catalytic subunits,
The Ras/cAMP/PKA pathway has also been implicated in (iii) its overexpression compensates for the loss of Ras func-
aging (212, 216), thermotolerance (439), bud site selection, tion, and (iv) PKA activity is elevated in a sch9⌬ background
actin repolarization (333), glycogen accumulation (342), stress (72, 109).
resistance (39, 407), and sporulation (39, 41); it may also reg- Activation of Ras and Gpa2 by glucose. Since artificial acti-
ulate pseudohyphal differentiation (growth as chain-like mul- vation of Ras and Gpa2 is sufficient to generate the glucose
ticellular filaments) in response to nutrient limitation. Each of response, and it seems unlikely that glucose interacts directly
these interesting topics (see reference 411 for a recent review), with the G proteins themselves, how do Ras and Gpa2 become
as well as the involvement in these processes of other nutrient- activated in wild-type yeast cells exposed to the sugar? It is
stimulated kinases (e.g., Snf1 and TOR [45, 75, 196, 197, 401, convenient to separate potential answers to this question into
441]), GTPases (e.g., Gpa2 [277]), and transcription factors two categories: recognition of glucose by receptors on the outside
(e.g., Msn2/Msn4 [30, 96, 112]), is outside the scope of this surface of the cell versus direct signaling of Gpa2 and Ras (or
review. their regulators) by intracellular glucose or its metabolites.
The Gpr1/Gpa2 pathway. Heterotrimeric G proteins are sig- (i) Extracellular sensing. There are 18 known hexose trans-
naling molecules composed of ␣, ␤, and ␥ subunits. They porter genes in S. cerevisiae (HXT1 to HXT17 and GAL2).
represent a second class of factors that, like monomeric Ras Deletion of seven of these genes results in the failure to trans-
polypeptides, are capable of binding guanine nucleotides (Fig. port or grow on glucose (308, 413). In this hxt1 to hxt7 null
2). The G␣ subunit Gpa2 and its negatively acting regulator background, constitutive expression of the GAL2-encoded ga-
(GAP) Rgs2 have been shown to participate in glucose signal- lactose permease restores glucose-induced cAMP synthesis
ing in S. cerevisiae (190, 394). Gpa2 was originally identified by (313). This suggests that the yeast hexose transporters do not
screening a yeast genomic library with cDNA probes encoding participate in glucose signaling but are required only for
mammalian G␣ subunits (252), and the putative ␤ subunits transport.
Gpb1 and Gpb2 were identified as interaction partners of The seven-transmembrane receptor Gpr1 may detect extra-
Gpa2 (135); no corresponding G␥ subunits involved in glucose cellular glucose and thereby activate Gpa2, but it is not re-
signaling have yet been identified (but see reference 135). quired to signal Ras; Gpr1 or Gpa2 deletion does not prevent
Whereas deletion of either GPA2 or RAS2 causes little or no the glucose-induced increase in Ras2-GTP levels (66). In con-
impairment of growth on glucose media, deletion of both trast, the Ras2-GTP increase is absent in a glucose phosphor-
causes a severe synthetic growth defect (428). Removal of the ylation-deficient (hxk1 hxk2 glk1) strain (66). Thus, Ras ap-
major cAMP phosphodiesterase in the cell (Pde2) restores pears to be activated by intracellular phosphorylated glucose
normal growth to gpa2⌬ ras2⌬ strains, suggesting that an in- but not by extracellular glucose. A clear demonstration of
crease in cAMP levels bypasses the loss of both Gpa2 and Ras2 extracellular sensing by Gpr1 will require direct evidence, for
(428). Most importantly, as mentioned above, the transcrip- example, as suggested by recent mutagenic analysis (205), that
tomic response to glucose is successfully mimicked in the ab- it interacts with glucose.
sence of the sugar by turning on expression of either activated (ii) Intracellular sensing. If extracellular signaling does not
Gpa2 or activated Ras2; the fact that this signaling is PKA participate in Ras activation (66, 313), it will be particularly
dependent confirms their redundant participation in the important to understand how intracellular glucose triggers the
cAMP-dependent branch of glucose signaling (408). glucose response. The longstanding idea that glucose phos-
A two-hybrid protein interaction screen with GPA2 as the phorylation (in particular by the glucose-induced hexokinase
bait led to the identification of the GPR1 gene, which encodes Hxk2; see below) and/or subsequent glycolytic reactions are
a member of the G protein-coupled seven-transmembrane re- required to alter yeast gene expression in response to glucose
ceptor superfamily (Fig. 2). Gpr1 is located on the yeast cell has been controversial (see references 20, 121, 244, and 314 for
surface (428). Genetic analysis strongly suggests that Gpa2 acts reviews of this topic). Since repression occurs even when the
downstream from Gpr1 in the same signaling pathway: the low metabolic steps immediately following glucose phosphoryla-
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 259

tion are reduced to less than 2% of wild-type levels, it seems tively new concept. Further study of receptor-mediated signal-
likely that nothing beyond the production of glucose-6-phos- ing in yeast may prove critical in understanding similar nutrient
phate is required (317). Further, since cells can be tricked into sensors in mammalian cells (179). How PKA, once unleashed,
the full-blown glucose response by expressing activated Ras or remodels the architecture of the yeast nucleus to produce the
Gpa2, glucose phosphorylation must generate the signal ultimate transcriptomic consequences of glucose induction and
through the G proteins and/or through a redundant and as- repression will be a central consideration of the remainder of
yet-obscure alternative pathway. A recent interesting sugges- this review.
tion is that glucose phosphorylation increases Ras2-GTP levels Most of the ⬃1,000 genes in S. cerevisiae that are transcrip-
by inhibiting the Ira (RasGAP) proteins (66). tionally downshifted during growth on glucose can be sorted
Although progress has been made, much more work is into one of three groups: gluconeogenic genes, Krebs cycle/
needed to establish the precise mechanism(s) through which respiratory genes, and genes involved in alternate carbon source
Ras, Gpa2, and/or their regulatory GEFs and GAPs is signaled metabolism. This section will review known mechanisms through
by glucose. Intracellular acidification (316, 365) and energy which the glucose signal is received in the yeast nucleus and
charge (the AMP/ATP ratio) (134, 244) are additional poten- modulates expression of glucose-repressed target genes.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


tial signaling mechanisms that require further testing. The
events that immediately follow activation of either G protein Involvement of the Hexokinase Hxk2
are much clearer (Fig. 2). Activated Ras or Gpa2 can each in the Glucose Response
generate a spike in cAMP production by adenylate cyclase;
cAMP in turn releases PKA catalytic (Tpk) subunits from The first, irreversible step in the intracellular metabolism of
inhibition by its regulatory (Bcy1) subunits. glucose is C6 phosphorylation. Three enzymes (encoded by
Extracellular sensing by Snf3/Rgt2. The putative glucose HXK1, HXK2, and GLK1) can catalyze this reaction; of these,
sensors Snf3 and Rgt2 are 60% identical to each other and, like only HXK2 is highly expressed in the presence of glucose (147).
hexose transporters found in bacteria, plants, and mammals HXK2 lesions (both point and null mutations) were long ago
(228, 259, 332), contain 12 predicted transmembrane-spanning shown to cause failures in glucose repression (99, 100, 104,
domains (272). Snf3 and Rgt2 do not appear to act as glucose 222). After much controversy, it is now deemed unlikely that
transporters but instead signal the presence of the sugar (118, Hxk2 catalytic activity plays an important role in the intracel-
178, 182, 247, 273, 314). Since the transcriptional response of lular glucose signaling pathway (244). The surviving version
Snf3/Rgt2 target genes (e.g., HXT genes encoding the glucose of this model postulates that, following the onset of the
transporters) is generated in the absence of glucose by expres- phosphoryl transfer reaction, a stable transition intermediate
sion of hyperactive Ras or Gpa2, the Snf3 and Rgt2 sensors alters Hxk2 conformation and mediates its regulatory function
appear either to act upstream of Ras/cAMP/PKA or to play an in altering target gene expression (20, 191). However, this
ancillary or redundant role in the initial detection of glucose. model fails to explain the fact that sugars metabolized in an
This view is consistent with the possibility that glucose sensing Hxk2-independent fashion can cause genome-wide transcrip-
by Snf3/Rgt2 is required for maintenance of glucose induction tional repression comparable to that observed after the addi-
or repression for at least a subset of the transcriptome (182, tion of glucose to wild-type cells or upon expression of acti-
272). Further, it seems clear that the Snf3/Rgt2 regulatory vated Ras or Gpa2 (17, 214, 312, 397, 408).
pathway helps to ensure that changes in gene expression are Interestingly, approximately 14% of Hxk2 is nuclear in glu-
commensurate with the glucose concentration in the environ- cose-grown cells (146). This discovery suggests an alternative
ment, a mechanism that is likely to be particularly important in explanation for the regulatory involvement of this hexokinase
nature. in the glucose response (244, 304). Hxk2 has been detected in
As mentioned above for Gpr1, a clear demonstration of the nucleus by enzymatic assay, immunoblotting, and fluores-
extracellular sensing by Snf3 and Rgt2 will require direct evi- cent visualization of a green fluorescent protein (GFP)-tagged
dence that they interact with glucose. Intriguing hints along chimera, confirming controversial reports of nuclear hexoki-
these lines have already been uncovered. The V404I mutation nase that first appeared in the early 1960s (234, 340, 390). The
in Rgt2 appears to abolish glucose signaling; this residue is negatively acting DNA-bound glucose regulator Mig1 (see be-
orthologous to one in Hxt1 (F371) that is necessary for glucose low) appears to mediate nuclear localization of Hxk2; Mig1
transport (247). V404 in Rgt2 may therefore be required for and Hxk2 have been shown to interact via two-hybrid, immu-
glucose sensing because it helps to form the glucose binding noprecipitation, and glutathione S-transferase pull-down as-
domain in the receptor. Similarly, dominant mutations in both says (2, 245). An interaction between Hxk2 and the Mediator
Snf3 (R229K) and Rgt2 (R231K) that cause constitutive sig- component Med8 has also been described, and the latter ap-
naling, even in the absence of glucose, map to the putative pears to bind directly to both positively and negatively acting
glucose binding pocket (272, 273). Progress has also been glucose regulatory elements in DNA (57, 80, 246). A previously
made with respect to the elaboration of a mechanism for sub- reported example of a metabolic enzyme that doubles as a
sequent signaling by Snf3 and Rgt2 across the plasma mem- transcriptional regulator is the galactokinase Gal1, which both
brane (see below). phosphorylates galactose and appears to activate the transcrip-
tion factor Gal4 by binding to the Gal4 inhibitor Gal80 (436).
Despite these intriguing hints, contradictory results prevent
SIGNAL RECEPTION: GLUCOSE REPRESSION
an unambiguous assignment of the Hxk2 function in glucose
Direct sensing of extracellular glucose by yeast cells, rather regulation to the nuclear form of the protein. For example,
than intracellular sensing of its metabolic products, is a rela- removal of a small N-terminal segment of Hxk2 (residues 7
260 SANTANGELO MICROBIOL. MOL. BIOL. REV.

through 16) in a putative nuclear localization sequence (Lys8- sence of SNF4 (53, 108, 204). Since Snf4 also coimmunopre-
Pro-Gln-Ala-Arg12) has been reported to abolish both Hxk2 cipitates with Snf1 and is required for maximal Snf1 protein
nuclear localization and glucose repression of SUC2, HXK1, kinase activity (in cells grown on a nonfermentable carbon
and GLK1 without impairing hexokinase catalytic activity (146, source) (52), it is a physically associated activator of the Snf1
311). The exact opposite result (intact glucose signaling and kinase (108, 113). Like Glc7 (and perhaps Reg1; see below),
low catalytic activity), however, was obtained by others upon Snf1 appears to have a regulatory role in glycogen metabolism
deletion of an overlapping and slightly larger Hxk2 segment that is somehow influenced by PKA signaling (see reference
(residues 1 through 15) (231). Likewise, the phosphorylatable 119 for a review). While this apparent connection between the
Ser14 residue in Hxk2 (193) is reported either to mediate (305) signal transduction pathways governing glucose catabolism and
or not to mediate (146, 231) glucose repression. Further anal- glycogen storage is intriguing, it remains poorly defined and
ysis is needed to resolve these discrepancies. Most importantly, will not be considered further here.
proof of a regulatory role for nuclear Hxk2 awaits an unam- Identification of a role in glucose repression for Glc7, the
biguous demonstration that its role in the glucose response yeast homolog of mammalian type I protein phosphatase (PP1)
relies upon the relatively small fraction of the protein that (111), began with a selection for mutants defective in glucose

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


resides in the nucleus. repression of a target gene (SUC2, encoding invertase) (260).
All of the recovered mutations were recessive; two of the three
complementation groups found in this screen proved to be
Snf1-Mediated Signal Transduction in Glucose
allelic with HXK2 and REG1, respectively. The third group
Repression and Derepression
identified a new locus (designated cid1, for constitutive inver-
The uncertainty surrounding the role of Hxk2 subcellular tase derepression). Complementation of one of these muta-
localization and phosphorylation state in the glucose response tions (cid1-226, now designated glc7-T152K) (378) led to the
is particularly important to resolve because it represents one of isolation of GLC7, a locus that was also identified indepen-
several potential connections between Ras/cAMP/PKA-depen- dently in a screen for glycogen-deficient mutants. Interestingly,
dent events that occur early in the signal transduction cascade in addition to SNF1 (GLC2), other glc lesions were allelic with
and downstream modifications that have a direct impact on IRA1 (GLC1), IRA2 (GLC4), and RAS2 (GLC5) (42), consis-
target gene expression. Importantly, increasing the extracellu- tent with numerous studies connecting Ras and Gpa2 signaling
lar glucose concentration, or hyperactivation of PKA via bcy1 with accumulation of the storage carbohydrates trehalose and
deletion, results in reduced Hxk2 phosphorylation, whereas glycogen (40, 41, 67, 125, 190, 284, 330, 363). Further investi-
attenuation of PKA activity increases 32P labeling of Hxk2 gation is needed to pursue this implicit regulatory linkage
(398). These data are consistent with a model in which Hxk2 between glucose catabolic pathways and carbohydrate storage.
dephosphorylation, which converts it from a monomer to a Complicating matters further, in addition to glucose repres-
dimer (305), somehow transduces the glucose signal to down- sion and carbohydrate accumulation, GLC7 phosphatase is
stream effectors, eventually resulting in repression (and per- needed for a variety of other cellular functions in S. cerevisiae
haps also induction) of glucose-regulated genes. The nuclear (14, 350, 437), including normal progression through the G2/M
form of Hxk2 might accomplish this via its direct interaction phase of the cell cycle (23, 24, 150, 159, 223, 288), actin orga-
with Med8 and/or Mig1 (see above). Alternatively or addition- nization (6, 54), translation (412), and sporulation (12, 42, 302,
ally, such an Hxk2 signal might be received by two additional 357, 378, 380). Recent work has further expanded this list of
phosphoproteins that are well established as downstream com- functions assigned to yeast PP1; it is also required for the
ponents in the glucose signal transduction pathway: the Reg1- glucose-induced vacuolar degradation of fructose-1,6-bisphos-
targeting subunit of the Glc7 phosphatase and the Snf1 kinase phatase (76) and for normal Mex67-mediated export of mRNA
(325). from the nucleus to the cytoplasm (127; see also references
Dueling Snf1 kinase and Glc7 phosphatase mediate glucose 381, 382, and 404).
regulation. SNF1 was first identified in screens for regulatory Interestingly, isolated PP1 molecules exhibit little substrate
factors in the glucose response (it was then named either CAT1 specificity and instead require regulatory subunits that alter the
or CCR1) (62, 102, 440). SNF1 was also identified in a screen conformation of the phosphatase and/or target it to its sub-
for mutants that could ferment glucose but not sucrose (46). strates (65, 97). This targeting hypothesis is supported by the
Since inactivation of SNF1 also impairs utilization of galactose, Glc7 localization pattern, which suggests dynamic changes
maltose, and nonfermentable carbon sources, soon after its throughout the yeast cell cycle (24). Glc7 binding proteins
identification it was recognized as an important regulatory specific to the following functions have been reported: glucose
gene mediating glucose derepression. The SNF1 gene was repression (Reg1) (380), glycogen metabolism (Gac1 and Pig1)
cloned and found to encode a serine-threonine protein kinase (59, 353, 426), cell cycle progression (Reg2 and Sds22) (120,
(51); the existing evidence also suggested a physical and func- 149, 288), actin organization (Scd5) (54), and sporulation
tional link to SNF4, another gene that had been identified in (Gip1) (357, 380).
the “sucrose nonfermenting” screen (49, 50, 259, 261). The snf4 A short motif present in most known regulatory subunits
and snf1 phenotypes are similar, including defective utilization ([RK]-X0-1-[VI]-X-[FW]) plays a critical role in the interaction
of the many carbon sources (e.g., sucrose, galactose, maltose, with a hydrophobic groove at the interface of two ␤-sheets in
raffinose, and lactate) that require expression of glucose-re- PP1 (97, 403). Thus, multiple regulatory subunits appear to
pressed genes. Increased SNF1 dosage and SNF1 mutations compete for Glc7 binding in vivo; for example, Gac1 overex-
(e.g., the SNF1-G53R allele, which produces a hyperactive pression affects glucose repression (426). In addition to bind-
kinase) can partially restore glucose derepression in the ab- ing regulatory subunits, the hydrophobic groove may mediate
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 261

changes in the structure and/or activity of the phosphatase. It sporulation (8, 45, 137, 196, 197, 297, 401). Clearly more work
also now seems likely that many if not all targeting subunits is needed before we will fully understand the respective rela-
interact with more than one site on the PP1 surface (425). In tionships between the distinct forms of the Snf1 kinase com-
the two-hybrid assay the F468R mutation in Reg1 dramatically plex and each of these regulatory mechanisms. However,
reduces its interaction with Glc7; the PP1 binding motif that Gal83 is likely to be a primary contributor to glucose regula-
contains this residue (RHIHF468) therefore appears to be one tion by the Snf1 complex, since Gal83 is the most abundant ␤
of the moieties required for formation of the Reg1/Glc7 com- subunit, it is the only ␤ subunit in the nucleus when cells are
plex (325). Importantly, Reg1 is the only regulatory subunit grown on a nonfermentable carbon source, and Snf1/Gal83 is
known to participate in glucose repression. the form responsible for most Snf1 kinase activity (144, 397).
The idea that Reg1 (first identified in reference 230 and in Another trio of factors that mediate Snf1 function are the
an earlier screen as Hex2 [101]) mediates the Glc7 role in redundantly acting Snf1 kinase kinases Sak1 (YER129W, for-
glucose repression arose because (i) reg1 and glc7 alleles were merly known as Pak1; the new name was chosen to avoid
both isolated in the screen for mutants with impaired repres- confusion with Prk1 or p21-activated kinases), Tos1, and Elm1,
sion of SUC2 (260, 378) (see above), (b) a combination of each of which shares sequence similarity with the human tumor

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


those defective alleles exhibited no synergy in relief from glu- suppressor and AMP-activated protein kinase LKB1 (155, 255,
cose repression of SUC2, (iii) SNF1 deletion is epistatic to both 354, 422). Removal of all three yeast activities confers an snf1
alleles, and (iv) Reg1 overexpression suppresses the repression phenotype. A one-to-one correspondence between these three
defect caused by glc7-T152K (379). Genetic and biochemical upstream kinases and the distinct forms that contain each of
experiments confirmed that Reg1 and Glc7 are linked both the three ␤ subunits has been ruled out (143). Since Sak1 is the
physically and functionally (325, 379). The results of alanine- most important of the trio of Snf1 kinase kinases in activating
scanning mutagenesis suggested that Reg1 interacts with at the Snf1/Gal83 form, it is likely also to be the most important
least two distinct areas of the resolved PP1 structure (14). regulator of Snf1-dependent glucose regulation (143). This
Satisfyingly, one of the Glc7 surfaces specific to its role in conclusion is further supported by the observation that dele-
glucose repression is adjacent to Thr152 (14); the glc7-T152K tion of SAK1 suppresses many of the Snf1-dependent pheno-
lesion had earlier been shown to impair Reg1-Glc7 interaction types observed in reg1⌬ cells (255).
(379). Like Glc7 and its regulatory subunits, the Snf1 kinase ap-
A functional link between Reg1 and Snf1 was suspected long pears to be localized by its ␤ subunits to distinct cellular sub-
ago, primarily because REG1 deletion results in derepression compartments, thereby targeting it to different substrates (331,
of glucose-repressed genes and SNF1 deletion results in a 397). In glucose-grown cells Snf1 is found exclusively in the
failure to derepress many (if not all) of those same genes. The cytoplasm; following a shift to a nonfermentable carbon
demonstration that Reg1 and Snf1 interact (218) left little source, Snf1 is targeted by Gal83 to the nucleus. The N termi-
doubt that the Reg1-targeted Glc7 phosphatase and Snf4- nus of Gal83, which is not shared by the other two ␤ subunits
activated Snf1 kinase are indeed interconnected compo- Sip1 and Sip2, is required for this function; the upstream ki-
nents of the glucose signaling pathway (106, 120, 162, 174, nase Sak1 also controls Snf1 localization to the nucleus in the
378, 379; see references 45, 121, and 178 for reviews) (Fig. 3). absence of glucose (144, 397). Upstream events in glucose
Since then numerous additional participants in this regulatory signaling may also be involved, since it has been reported that
duel have been discovered. For example, three ancillary com- PKA regulates nucleocytoplasmic shuttling of Snf1/Sip1 (144).
ponents of the Snf1/Snf4 kinase complex (Sip1, Sip2, and Gal83) If PKA also regulates shuttling of Snf1/Gal83, it appears to do
strengthen Snf1/Snf4 association (175, 429, 430). Each of these so redundantly with another signaling pathway.
three proteins contains an Snf1 binding internal region and an With respect to subnuclear localization, three as-yet-uncon-
Snf4 binding C-terminal region (their N-terminal regions are nected observations suggest the possibility that the Snf1 kinase
divergent); they therefore appear to bridge the interaction complex operates at least in part at the nuclear periphery: Snf1
between Snf1 and Snf4. By analogy with the heterotrimeric is concentrated in nuclear envelope fractions prepared from
mammalian Snf1 homolog, the AMP-activated protein kinase, pyruvate-grown (but not glucose-grown) cells (B. Menon and
SNF1 encodes the yeast ␣ subunit, SNF4 encodes the ␥ subunit, G. M. Santangelo, unpublished data), Glc7 has been impli-
and SIP1, SIP2, and GAL83 redundantly encode the ␤ subunit cated in mRNA processing as a component of the cleavage/
(136). polyadenylation complex (127, 256, 404), and Reg1 deletion
It was initially puzzling that little or no detectable phenotype causes an RNA processing defect (381, 382). Further elabora-
resulted from mutation of all three genes that encode the ␤ tion of this potential connection with perinuclear phenomena
subunits of the Snf1 kinase complex (105, 106, 121, 175). How- is needed and may provide critical information about the func-
ever, the construction of true null alleles revealed that, as tion and organization of the regulatory apparatus in the yeast
expected, the sip1⌬ sip2⌬ gal83⌬ triple mutant does indeed nucleus that responds to glucose (see below).
display an snf1 phenotype (331). Despite the observed redun- An overview of the regulatory duel between the Reg1/Glc7
dancy, the three ␤ subunits play distinct roles in vivo, in par- phosphatase and the Snf1 kinase complex is presented in Fig.
ticular with respect to Snf1 substrate specificity (143, 144, 175, 3. Snf1 has two domains, an N-terminal kinase domain and a
213, 254, 331, 396, 397, 430). C-terminal regulatory domain. In high concentrations of glu-
Besides its role in the response to glucose limitation, Snf1 cose the regulatory domain remains bound to the catalytic
has also been implicated in several other processes, including domain, maintaining Snf1 in an autoinhibited conformation
aging, thermotolerance, peroxisome biogenesis, filamentation, (labeled “inactive” in Fig. 3) (325). Upon depletion of glucose,
invasive growth, biofilm formation, meiosis, and (like Glc7) Snf4 counteracts autoinhibition of Snf1 by interacting with its
262 SANTANGELO MICROBIOL. MOL. BIOL. REV.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


FIG. 3. The opposing roles of Reg1/Glc7 phosphatase and the Snf1 kinase complex. The duel between Reg1/Glc7 and Snf1/Snf4/Gal83 turns
the Snf1 kinase on (ACTIVE) and off (INACTIVE) by determining whether the Snf1 regulatory domain (RD) binds to and autoinhibits its kinase
domain (KD). This switch plays a central role in determining the transcriptomic response to the presence or absence of glucose. The ␤ subunit
Gal83 acts as a scaffolding factor for the nuclear form of the Snf1 (␣) and Snf4 (␥) subunits of the kinase. Addition of glucose to yeast cells growing
on an alternative carbon source results in the dephosphorylation of active Snf1 kinase by Reg1/Glc7 (no. 1); this returns Snf1 to the autoinhibited
state (no. 2) and results in export of Snf1/Gal83 to the cytoplasm (not shown). Once glucose is depleted, Sak1 phosphorylates Snf1 (no. 3),
Snf1/Gal83 enters the nucleus (not shown), and Snf4 binds to the Snf1 regulatory domain, thereby releasing the catalytic domain (no. 4); Reg1 then
undergoes rapid Snf1-dependent phosphorylation (no. 6), which stabilizes the interaction between Snf1 and the Reg1/Glc7 phosphatase (no. 7) and
primes the latter to repress Snf1 anew should glucose reappear. Std1 interacts with the Snf1 catalytic domain and stimulates kinase activity (no.
5), although it is stoichiometrically underrepresented in Snf1 complexes. A dynamic equilibrium (purple arrows between no. 6 and 7), in which Glc7
appears to counteract Snf1 phosphorylation of Reg1 and promote its own dissociation from active kinase complexes, is at the heart of this
regulatory duel. Sip5 may stabilize the Reg1/Snf1 interaction in no. 6 (not shown). Hxk2 interacts with Reg1 (not shown), and Hxk2 may respond
to PKA signaling by dimerizing and mediating the switch in Glc7 phosphatase substrate selection (from Reg1 [no. 7] to Snf1 [no. 1]; see the text).

regulatory domain; deletion of the latter bypasses the require- localization to the nucleus is essentially carbon source inde-
ment for SNF4 in derepression of glucose-repressed genes (52, pendent, in glucose-grown cells nuclear Snf4 is apparently not
174, 204). Snf4 binding to Snf1 therefore produces an active, associated with other subunits of the Snf1 kinase complex
open conformation of the complex (labeled “active” in Fig. 3) (397). Thus, Snf4 apparently does not contribute to the cyto-
(174). Phosphorylation of a conserved threonine (T210) in the plasmic roles of Snf1 that are distinct from its mediation of
activation loop of Snf1 is essential for activation of the kinase; glucose derepression.
the upstream kinase Sak1 has been shown to phosphorylate Glucose repression involves Reg1/Glc7-facilitated conver-
this residue and is required for full Snf1 kinase activity (255). sion of the kinase complex from the active to the autoinhibited
Blocking phosphorylation at this position via a T210A muta- state, presumably via dephosphorylation of Snf1 (325). Reg1/
tion impairs Snf4 binding to the regulatory domain and pre- Glc7 appears to associate only with active kinase complexes,
vents the release of Snf1 autoinhibition (174, 198, 218). The since the catalytic domain of Snf1 (phosphorylated at T210) is
T210A lesion also interferes with normal nuclear enrichment required for the interaction with Reg1 (218). Upon activation
of Snf1 after a shift from high to low glucose (144). Since Snf4 in a reg1 or glc7-T152K background, the Snf1 complex becomes
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 263

trapped in the active conformation. Overexpression of Reg1, Equally mysterious is the apparent capacity of the glucose
but not overexpression of the mutated form that fails to bind signal to change the target of Glc7 phosphatase from Reg1
Glc7 (ReglF468R; see above), interferes with the Snf1/Snf4 in- to Snf1 (no. 7 and 1 in Fig. 3, respectively), which in the
teraction in low glucose (325). Sip5, the first protein shown to absence of Snf1 phosphorylation (no. 3) traps Snf1 in the
bind to both Reg1/Glc7 and the Snf1 kinase complex, may autoinhibited inactive state. Interestingly, PKA signal trans-
stabilize the interaction between them and thereby facilitate duction might alter Glc7 targeting through the negatively
glucose repression, since the two-hybrid interaction between acting regulator Hxk2. As mentioned above, PKA signaling
Reg1 and Snf1 is reduced threefold in a sip5⌬ mutant (326). appears to cause reduced phosphorylation of Hxk2, which
The critical regulatory interaction between Reg1 and the favors its dimeric form; the phosphatase that dephosphory-
Snf1 complex was presumed to occur in the nucleus, especially lates Hxk2 in response to glucose is known to be Reg1/Glc7
since that is where active Snf1 is predominantly located prior (4, 305, 398). Hxk2 (presumably its underphosphorylated
to being switched off and exported following the appearance of dimeric form) in turn interferes with dephosphorylation of
glucose, and Glc7 is known to be largely nuclear in growing Reg1 by Glc7 (325). Since only the phosphorylated form of
cells (24, 437). However, in contrast with a previous report that Reg1 promotes dissociation of Snf4 from Snf1 and inactiva-

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


found Reg1 in the nucleus (265), fluorescently tagged Reg1 tion of the kinase, the PKA-mediated transition from mo-
appears to be exclusively cytoplasmic (90, 91). To resolve this nomeric to dimeric Hxk2 is potentially a direct glucose trig-
discrepancy it was proposed that the activated form of Snf1 ger that stabilizes the repressing form of Reg1/Glc7 and
cycles rapidly between the nuclear and cytoplasmic compart- thereby inactivates Snf1. Reg1 and Hxk2 were reported to
ments, allowing inactivation by Reg1/Glc7 to occur in the cy- interact as predicted by this model (325). This is further
toplasm following the appearance of glucose (91). This hypoth- supported by a wealth of additional data: Reg1 overexpres-
esis has not yet been tested, but it may explain the apparent sion suppresses the glucose repression defect of an hxk2⌬
role of Bmh1 and Bmh2 in glucose-regulated gene expression mutant (325), the association between Reg1 and Glc7 is
(168). Bmh1 and Bmh2 are the yeast homologs of the some- unaffected by removal of Hxk2, and the Snf1-Snf4 interac-
what mysterious 14-3-3 proteins (33, 388), which may serve as tion is glucose insensitive in the absence of Hxk2 (174).
mediators of Reg1 function (90) and are known to respond to A near-complete chain of causality describing the glucose
Ras/PKA signaling (124, 232). response can thus be stated as follows (Fig. 3): early events
Additional information concerning the connection between in glucose signaling stimulate PKA to inhibit phosphoryla-
upstream glucose signaling events and the Glc7/Snf1 regula- tion of Hxk2, thereby targeting it to Reg1/Glc7, where Hxk2
tory duel will be the next topic of discussion, followed by a is dephosphorylated, dimerizes, and blocks dephosphoryla-
review of the downstream DNA-bound phosphoproteins that tion of Reg1 by Glc7; Glc7 then switches targets and inac-
are known Snf1 regulatory targets and that account for much tivates the Snf1 kinase, which can no longer phosphorylate
of the transcriptomic response to glucose in yeast cells. its DNA-bound substrates, resulting in altered expression of
Transmission of the Ras/cAMP-dependent glucose signal to the relevant target genes. Ambiguity unfortunately accom-
Reg1/Glc7 and Snf1/Snf4. Although the regulatory duel be- panies even this parsimonious view of the glucose regulatory
tween Snf1 kinase and Glc7 phosphatase has now largely been cascade, since the relationship between subcellular distribu-
explained at a molecular level, the input by glucose signaling tion and function awaits clarification for several of the fac-
that shifts the balance in favor of the closed, inactive form of tors involved (most notably Hxk2 and Reg1).
the kinase remains poorly characterized. A proposal that Snf1 DNA-bound repressors controlled by Snf1. Several down-
responds to the AMP/ATP ratio (421), which drops dramati- stream repressors are known to be phosphorylated and inacti-
cally upon removal of glucose (7, 421), has been difficult to vated by the Snf1 kinase in the absence of glucose. The first of
substantiate, since Snf1 kinase activity does not respond to these to be discovered was Mig1, originally identified as a
these nucleotides in vitro (136). In contrast, the ample evi- multicopy inhibitor of GAL gene expression (257). Deletion of
dence that PKA and Snf1 act antagonistically in affecting a the MIG1 gene relieves glucose repression of numerous target
similar set of cellular functions (7, 164, 366) is reinforced by genes (115, 131, 180, 189, 257, 258, 335, 336, 406), and its
the similarity of the transcriptome-wide response of Snf1 target product is thought to interact with the intergenic regions of at
genes following either the addition of glucose or the expression least 90 different genes in vivo (221, 251). Mig1 is presumed to
of activated G proteins in the absence of glucose (408). function downstream of Snf1, because deletion of MIG1 sup-
Sak1, the recently identified upstream activator of Snf1 that presses snf1 defects in glucose derepression (180, 386).
regulates both Snf1 kinase activity and its nuclear localization Mig1 is a Cys2His2 zinc finger protein that binds to a GC-rich
(144, 255), is an obvious potential intermediary between PKA core sequence (219). The consensus Mig1 binding site deter-
and Snf1 (question mark in Fig. 3). If the gap between the mined by in vitro selection is 5⬘-ATAAAATGCGGGGAA-3⬘
earliest events in glucose signaling and Sak1 could be bridged, (221). At the time of its discovery, other zinc finger proteins
it would represent a complete chain of causality between glu- were known to be capable of functioning as either activators or
cose sensing at the cellular periphery and the response of repressors, depending upon the chromosomal and cellular
DNA-bound regulators in the nucleus. Not surprisingly, how- context (207). This was therefore also considered a possibility
ever, given that its role as an Snf1 activator was only recently for Mig1 (257), a suggestion that later proved to be correct:
uncovered, little is yet known about regulation of Sak1 and both Mig1 and the related Cys2His2 zinc finger-containing
whether or not the PKA pathway might be involved. repressor Mig3 (Yer028) can also function as activators (220,
264 SANTANGELO MICROBIOL. MOL. BIOL. REV.

221, 303, 376, 423). Although this has complicated the clear export in response to glucose removal (86). These and
interpretation of Mig1 function (37, 160, 189, 376, 384, 386, other data suggest that Snf1 phosphorylation of Mig1 inacti-
423), clearly the primary physiological role of Mig1 seems to be vates it by changing its subcellular localization.
that of a negative regulator of glucose-repressed genes (e.g., According to this Mig1 nucleocytoplasmic shuttling hypoth-
SUC2 and GAL1). esis, Hxk2- and Reg1/Glc7-dependent inactivation of Snf1 in
Interestingly, the Mig1 N-terminal zinc fingers are very sim- the presence of glucose results in hypophosphorylation of Mig1,
ilar to those in the C terminus of WT1 (257), a human tumor which causes it to relocate rapidly to the nucleus; subsequent
suppressor that collaborates with p53 to repress transcription binding of Mig1 to its recognition sites upstream from glucose-
and (like p53) also activates transcription (133, 224, 235, 248, regulated genes mediates transcriptional repression (86, 87).
264, 268, 306, 348). Mig1 and WT1 are both phosphoproteins, Although this model is attractive, it is difficult to reconcile with
and PKA phosphorylation of WT1 appears to make an impor- two strikingly contrasting results. First, the Mig1 target gene
tant contribution to its repressor function (323); the signifi- GAL1 is derepressed normally in an msn5 mutant grown on
glycerol, despite the constitutive presence of Mig1 in the nu-
cance of a potential PKA target site found in the internal
cleus (86). Second, Mig1 remains bound to the GAL1 pro-
regulatory region of Mig1 is not yet known (257, 270).

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


moter in vivo under these same derepressing conditions (278).
Glucose-dependent repression by a LexA-Mig1 fusion is
Both of these results suggest that Mig1 transcriptional repres-
readily detected by a heterologous reporter gene with up-
sion of target genes is regulated via a mechanism other than
stream LexA operator sites, suggesting that Mig1 can repress
nuclear export and that the key function of Snf1 phosphoryla-
transcription without the assistance of other promoter-bound tion in relieving Mig1-dependent glucose repression is to in-
factors (376, 377). Analysis of LexA-Mig1 repression also led activate the relatively small amount of Mig1 that remains in the
to the discovery that the Mig1 polypeptide, which has a pre- nucleus and is bound upstream of target genes in derepressed
dicted molecular mass of 55 kDa (504 residues), is phosphor- cells (87, 270). How this is accomplished remains obscure.
ylated to different extents in glucose-repressed and dere- Mig3, Nrg1, and Nrg2 are three additional Snf1-regulated
pressed cells and might be a downstream target of Snf1 kinase repressors in S. cerevisiae. Mig3 is somewhat mysterious; it was
(87, 376). Indeed, three consensus Snf1 kinase sites (Ser278, identified because it contains Cys2His2 zinc fingers that are
Ser311, and Ser381) map within a region of Mig1 required for very similar to those of Mig1 and Mig2 (26, 220). Several
glucose regulation (77, 87, 269). All three of these sites, and a additional results seem to suggest that Mig3, like Mig1 but not
fourth one that matches the Snf1 consensus imperfectly Mig2, is an Snf1-controlled, DNA-bound glucose regulator
(Ser222), are phosphorylated by Snf1 in vitro (343). Mutation (182, 221). It binds to the Mig1 binding sites upstream from
of these serine residues to alanine, including a Mig1 mutant SUC2, and a LexA-Mig3 chimera functions as a glucose-de-
in which all four residues were mutated (S222A⫹S278A⫹ pendent repressor of reporter gene expression when bound to
S311A⫹S381A), impaired but did not eliminate Mig1 phos- upstream LexA operators. When cells are shifted from glucose
phorylation and glucose repression (377). Nevertheless, in vivo to galactose, Mig3 is extensively phosphorylated (at least in
phosphorylation of Mig1 is dramatically reduced in an snf1 part by Snf1) and rapidly subjected to Snf1-dependent prote-
mutant, and immunoprecipitated Snf1 (but not the kinase- olysis (94). Nevertheless, physiologically significant regulation
dead Snf1-K84R) phosphorylates coprecipitated Mig1. Also, of target genes by Mig3 is yet to be demonstrated; in wild-type
as expected based upon their capacity to mediate glucose re- cells under standard conditions, it seems unlikely to be more
pression by inhibiting Snf1 kinase activity, deletion of either than a minor regulator of glucose-responsive gene expression
REG1 or HXK2 results in Mig1 hyperphosphorylation under (220, 221). Recently the MIG3 gene was also isolated in a
repressing conditions (377). Taken together, the simplest in- screen for multicopy suppressors of Rad53-GFP toxicity, but
terpretation of these data is that phosphorylation of Mig1 by the relationship between Mig3 function and the DNA damage
Snf1 is at least partly responsible for inactivation of Mig1 response awaits further clarification (94). It is hoped that tran-
scriptomic analysis of the effects of an as-yet-untested environ-
repressor function.
mental condition or genetic background will eventually reveal
Deletions that map within an internal region containing
the Mig3 role in glucose regulation and explain the results
most of the Snf1 phosphorylation sites (residues 261 to 400)
obtained to date.
converts Mig1 into a constitutive repressor that is not inacti-
Nrg1 and Nrg2 contain Cys2His2 zinc fingers that are very
vated when glucose is depleted (270). Interestingly, deletions
similar both to each other and to those in Mig1, Mig2, and
in Mig1 that remove all or part of the internal region from Mig3 (400). Nrg1 was first identified in two separate screens
residue 261 to 400 cause it to persist in the nucleus in the for factors mediating repression of glucose-regulated genes (1,
absence of glucose, whereas under the same conditions wild- 282), and Nrg2 was discovered because of its two-hybrid inter-
type Mig1 is largely exported to the cytoplasm (87). Fusion of action with Snf1 (400). Outside of their DNA binding domains
Mig1 residues 261 to 400 to a GFP–␤-galactosidase chimera Nrg1 and Nrg2 share only 27% identity, and this is thought to
confers glucose-regulated nuclear import and export upon the account for their somewhat distinct regulatory roles in glucose
resulting fusion protein, and its import in the presence of repression, filamentous invasive growth, and biofilm formation
glucose requires both REG1 and HXK2. In glycerol-grown cells (18, 196, 282, 400, 438). In the absence of glucose, neither
Mig1 becomes increasingly nuclear as more Snf1 kinase sites factor is degraded, nor is their capacity to bind target sites in
are removed (86). Msn5, an importin ␤ homolog originally vivo impaired. Both Nrg1 and Nrg2 exhibit glucose-dependent
identified due to its suppression of an snf1 mutation when repression of a heterologous reporter gene; they are also both
overexpressed (107), interacts with Mig1 and mediates its nu- capable of interacting with Snf1 kinase, but (unlike Mig1 and
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 265

Mig3) neither appears to be phosphorylated by it (18, 400). genes whose expression is repressed in the presence of glucose.
Although Snf1 appears to modulate Nrg2 levels and is clearly Our clearest picture is of glucose induction by the Snf3/Rgt2
required for normal Nrg1 function, the regulatory relationship sensing mechanism, a pathway that appears to operate in a
between these factors is complex and in need of further study. largely PKA-independent fashion and (at minimum) contrib-
Interestingly, like Mig1 and Mig3 (see above), Nrg1 can func- utes the critical regulatory trim that allows S. cerevisiae to grow
tion as an activator in some circumstances (18). well on a broad range of glucose concentrations (from micro-
DNA-bound activators controlled by Snf1. The Snf1 protein molar to molar concentrations) (272). The integral plasma
kinase also regulates a number of positively acting transcrip- membrane proteins Snf3 and Rgt2 mediate both transcrip-
tion factors required for the utilization of nonfermentable car- tional repression and derepression of four of the genes that
bon sources. Two of these factors, Cat8 and Sip4, both contain encode glucose transporters (HXT1 to HXT4). This pathway
zinc finger DNA binding domains similar to that in Gal4 (a C6 establishes a regulatory link between glucose concentration
zinc cluster) (145, 206) and bind specifically to a set of carbon and expression of the transporters, and it operates in a largely
source responsive elements (CSRE) under derepressing con- Snf1-independent fashion (for reviews, see references 179 and
ditions (300, 395). Cat8 and Sip4 are closely related structurally 272). Snf3 and Rgt2 are putative glucose sensors that appear to

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


and appear to have slightly different affinities for variants of the act redundantly with the Ras2/Gpa2/PKA pathway, since the
CSRE sequence (319). Although Sip4-dependent transcrip- latter seems sufficient for glucose signaling through both the
tional activation has been shown to require phosphorylation of Snf1-dependent and Snf1-independent branches (408) (see
Sip4 by Snf1 kinase, removal of Sip4 has little or no effect on above). Interestingly, however, like activating mutations in
CSRE-regulated genes (141, 145, 206, 299). Also, unlike dele- RAS2 and GPA2, at least part of the glucose signal can be at
tion of CAT8, which causes defective growth on nonferment- least partially mimicked in the absence of glucose via dominant
able carbon sources, deletion of SIP4 results in no obvious mutations in SNF3 or RGT2 (228, 272, 273). This may mean
phenotype (206). that the Snf3/Rgt2 regulatory pathway, in addition to modu-
Cat8 regulates the expression of a wide array of genes in- lating expression of key glucose transporters, signals PKA or a
volved in the metabolism of nonfermentable carbon sources, PKA-independent glucose response pathway and thus aug-
including genes necessary for ethanol utilization (e.g., ACS1 ments or tailors the transcriptomic response beyond its well-
and ACR1/YJR095W) (28, 192), gluconeogenesis (141, 145), established effect on transporter genes.
the glyoxylate cycle (141), lactate utilization (JEN1) (27), and
isocitrate metabolism (IDP2) (27); Cat8 also regulates expres- Snf3/Rgt2 Signaling of the Rgt1 Repressor Complex
sion of SIP4 (141, 395). Cat8 activity is regulated on two levels:
its expression is Mig1 repressed (145, 257), and its capacity to Upon transmission to the cytoplasmic side of the plasma
activate transcription requires a functional Snf1 kinase (303). membrane, the Snf3/Rgt2-mediated glucose signal appears to
Snf1 phosphorylates Cat8 in ethanol-grown cells (56, 303); operate through YCK1 and its 77% identical paralog, YCK2, an
following the addition of glucose, it is dephosphorylated in a essential gene pair encoding casein kinase (Fig. 4) (310). Like
Glc7-independent manner (303). Interestingly, constitutively Ras (see above), Yck1 and Yck2 are tethered to the membrane
expressed Cat8 fused to the Ino2 activation domain bypasses via palmitate moieties attached to C-terminal Cys-Cys se-
the need for a functional Snf1 in cells growing on ethanol quences (9, 318). Membrane anchoring of the Yck proteins is
(300). thought to facilitate an interaction with Snf3 and Rgt2 that
Cat8 works together with another Snf1-regulated transcrip- activates the kinases following the glucose-induced conforma-
tional activator, Adr1, for maximal expression of a small group tion change in the sensors; Yck1 has been shown to interact
of shared target genes (including ADH2, ACS1, and ALD6) with Rgt2 in vivo (179, 247). The targets of activated Yck
under derepressing conditions (25, 98, 192, 356, 405, 432). Like kinases in this pathway are Std1 and Mth1 (247), the corepres-
the Mig1, Mig3, Nrg1, and Nrg2 repressors, Adr1 has a sors that interact with the Rgt1 repressor; the latter is a C6 zinc
Cys2His2-type DNA binding domain (283), and its function cluster protein that binds directly to DNA sites upstream from
requires Snf1 kinase (84), although the mechanism of Adr1 target genes (122, 163, 202, 272, 289, 332). Mutations in Rgt1
regulation remains unclear. It has been suggested that Snf1 restore HXT gene expression and were originally isolated via
and Reg1/Glc7 contribute to regulation of Adr1 by affecting its suppression of defective glucose regulation in the snf3⌬ and
binding to promoters of target genes. Snf1 promotes Adr1 grr1⌬ backgrounds (106, 227). Note that the Rgt1 repressor
binding under derepressing conditions, while Reg1/Glc7 inhib- blocks transcription of glucose-induced genes, unlike the Mig
its Adr1 binding to promoters in the presence of glucose (91, and Nrg repressors, which block transcription of glucose-re-
433); it is currently unknown whether Adr1 is a direct target of pressed genes (see above). Thus, the Mig and Nrg proteins act
Snf1 and Reg1/Glc7 or whether this effect occurs through as repressors only in the presence of glucose, and Rgt1 acts as
intermediary factors. Adr1 activity has also been reported to be a repressor only when glucose is absent (Fig. 4).
negatively regulated by PKA (60) in a manner independent of The large (⬎200-residue) cytoplasmic C-terminal tails of
its DNA binding (364). Snf3 and Rgt2 are an important feature of this signaling path-
way. Deletion of the tail domains impairs Snf3- and Rgt2-
dependent gene regulation in response to glucose, and fusing
SIGNAL RECEPTION: GLUCOSE INDUCTION
the tail to Hxt1 or Hxt2 complements those defects, as does
The connection between PKA function and up-regulation of expression of the Snf3 tail domain by itself (69, 89, 228, 247,
the ⬃1,000 glucose-induced genes is even more mysterious 272, 274). Although they were originally thought to play an
than the mechanisms discussed above that control the ⬃1,000 important role in receiving the glucose signal, recent evidence
266 SANTANGELO MICROBIOL. MOL. BIOL. REV.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


FIG. 4. The Snf3/Rgt2 signaling pathway. Transcription of hexose transporter genes (HXT) is repressed by Rgt1 in the absence of glucose; in
contrast, Mig1 represses transcription of its target genes in the presence of glucose and cross-regulates the Snf3/Rgt2 pathway as shown. Relief
from the repressor function of Rgt1 occurs via glucose signaling of the Snf3/Rgt2 sensors, which stimulate phosphorylation of Mth1 and Std1 by
Yck kinases; the C-terminal tails of Snf3 and Rgt2 also appear to facilitate this event by interacting with both the kinases and their substrates. The
Yck polypeptides are tethered to the plasma membrane via palmitate (red squiggles). Yck-phosphorylated Mth1 and Std1 are subjected to
SCFGrr1-mediated ubiquitination and degradation by the proteasome. This releases Rgt1 from its upstream DNA binding sites and results in
derepression of downstream HXT target genes. Removal of Mth1 and Std1 converts Rgt1 into a transcriptional activator that may stimulate HXT
expression in the absence of specific DNA binding activity.

suggests instead that a dynamic interaction with Mth1 and Std1 (117, 179, 247). Mutations in GRR1 were shown long ago to
(201, 332) is the important function of the Snf3 and Rgt2 impair the regulatory response to glucose (11) and have since
C-terminal tails in glucose signaling (247). been associated with a broad array of phenotypes, including
The membrane anchoring of Mth1 and Std1 by the cytoplas- cell elongation, decreased divalent cation transport, defects in
mic tails is thought to mediate phosphorylation of these Rgt1 sporulation, and slow growth or lethality in combination with
corepressors by Snf3/Rgt2-associated Yck kinases (Fig. 4) defective amino acid biosynthetic pathways (21, 114, 158, 215,
(247). Both Std1 and Mth1 can be phosphorylated by affinity- 427, 431).
purified Yck1 in vitro, and membrane fractions of crude ex- Interestingly, the F-box motif of Grr1, which mediates its
tracts yield maximal Mth1 phosphorylation; mutant forms of interaction with Skp1 (187), may explain the pleiotropic phe-
Yck kinase were used to demonstrate that the production of notype of grr1⌬ cells (10, 209). Skp1 is a component of several
phosphoforms of these regulators is Yck dependent (247). The structurally distinct but functionally related ubiquitin ligase
likely site of Yck phosphorylation is a region conserved in both complexes that target a wide variety of regulators to the 26S
Std1 (residues 129 to 147) and Mth1 (residues 118 to 136), proteasome for degradation (for a review, see reference 414).
each of which contains several matches with the consensus These complexes are termed SCF to indicate their two com-
target sequence of casein kinase I (SXXS) (247). mon components, Skp1 and the cullin Cdc53, and a third ex-
Once Std1 and Mth1 are phosphorylated by the Yck kinases, changeable component, the F-box protein. In S. cerevisiae a
they are degraded via a Grr1-dependent mechanism (Fig. 4) total of 21 F-box proteins have been identified by sequence
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 267

alignment (414). Further analysis is urgently needed, since contribution to these regulatory loops involve three zinc finger-
most of these factors have neither been verified as SCF com- containing repressors, Mig2, Mig3, and Rgt1, as well as the
ponents nor characterized as to their function, and each bona Rgt1 corepressors Std1 and Mth1. For example, glucose sig-
fide SCF subunit is expected to confer a distinct substrate naling through Snf3/Rgt2 induces expression of the MIG2-
specificity upon its respective complex. Like Grr1, any one of encoded repressor, and in the absence of glucose Std1 binds to
these F-box proteins could contribute to nutrient signaling by and activates Snf1 kinase (Fig. 3) (see above). The Snf1-Mig1
targeting kinase substrates in response to the appearance or pathway in turn represses expression of Snf3 and Mth1. Im-
disappearance of glucose. portantly, despite these links to Snf1 kinase activity, signaling
Two F-box proteins that target cell cycle regulators for deg- by Snf3/Rgt2 does appear to operate as an independent glu-
radation were first identified as a result of their important roles cose response pathway (Fig. 4) (179, 182, 228, 272, 273). Thus,
in cell cycle progression: Cdc4, which targets the cyclin-depen- Snf3 and Rgt2 initiate a component of the glucose response
dent kinase inhibitor Sic1, and Grr1, which (long after its that overlaps at least partially with both the PKA pathway and
discovery as a regulator of the glucose response) was found to its Snf1 kinase branch but that can also operate independently
target the G1 cyclins Cln1 and Cln2 (16, 110, 158, 209, 341, 393, of either.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


414). Grr1 was shown subsequently to also target Gic2, a An important remaining aspect of this glucose regulatory
Cdc42 effector that mediates actin polarization at bud emer- pathway that awaits clarification is the apparent capacity of
gence (170, 414). A breakthrough in understanding the initially Rgt1, under conditions (high glucose concentration) in which it
puzzling GRR1 contribution to the glucose response (11, 209, no longer interacts with its recognition sites in DNA, to reverse
385, 387) came with the discovery that SCFGrr1, in addition to roles and function as an activator of HXT transcription and of
targeting the aforementioned cell cycle regulators, mediates a heterologous reporter gene (186, 250, 271, 275) (however,
the ubiquitination and degradation of the Rgt1 corepressors see also reference 117). Although its functional significance
Std1 and Mth1 (117, 209, 347). remains unclear, transcriptional activation by Rgt1 requires
Grr1-mediated ubiquitination and degradation of Mth1 and the glucose sensors Snf3 and Rgt2 and the F-box protein Grr1
Std1 appear to require, in addition to the Skp1 interaction with (275).
the Grr1 F box, an LRR domain comprised of 12 leucine-rich
repeats that form the substrate-targeting domain in Grr1 (158,
187). This is consistent with our knowledge of structure-func- The Cell Cycle Connection
tion relationships in other F-box proteins, which invariably
possess a second motif that mediates substrate targeting (414). Eukaryotic cells grow and divide at maximal rates only when
For example, in Cdc4 a WD40 repeat domain is sufficient for glucose is freely available. This requires sophisticated regula-
high-affinity interaction with its substrates (253). tory coordination between the metabolic engine of growth and
Degradation of Mth1 in glucose-grown cells has been shown the wide variety of structural events required for cellular re-
to require both phosphorylation by Yck kinase (247) and the duplication. With respect to glucose signaling it has long
LRR domain of Grr1, although Grr1 residues identified as seemed likely that there is a connection with the essential
essential for Cln2 interaction appear to be distinct from those cyclin-dependent kinase Cdc28; the latter acts in concert with
required for interaction with Mth1 (117, 158, 347). No infor- the G1 cyclins Cln1 to -3 at the primary gating event (Start) in
mation is yet available regarding the structural requirements of the cell division cycle (3, 116, 132, 165, 262, 263, 276, 285, 286,
the Grr1-Std1 interaction; this will likely be more difficult to 321, 371, 416, 424; see references 83, 365, and 389 for reviews).
determine, since Std1 apparently does not disappear as com- Unfortunately many of the critical relationships between the
pletely as Mth1 in the presence of glucose (182). growth and cell cycles have resisted elucidation. The slow
In the absence of glucose Rgt1 binds to a consensus sequence progress on this front is evident when one considers that Hart-
(5⬘-CGGANNA-3⬘) (186) found in multiple copies upstream of well first codified the basic questions over 30 years ago (138):
the HXT target genes (250, 275). Transcriptional repression by how does macromolecular synthesis communicate with the
Rgt1 requires Mth1, which causes a conformation change that CDC28 gene product, and how are all of these diverse inputs
allows Rgt1 to bind to its recognition sites in DNA (117, 202, (e.g., the pathways for carbon, nitrogen, sulfur, phosphorus,
289). Std1 also interacts with Rgt1 in the absence of glucose and potassium assimilation) integrated at Start?
(289, 372), although it does not regulate the Rgt1 DNA bind- Two mechanisms of apparent coordination between glucose
ing activity (179). More work is needed to determine the signaling and cell cycle progression seem particularly worthy of
contribution of Std1 to Rgt1-mediated repression in the pres- further attention. The first involves Grr1, the aforementioned
ence of glucose; an interesting possibility is that its interaction F-box protein that both relieves glucose repression as a com-
with Rgt1 shifts the activator/repressor equilibrium (see below) ponent of the Snf3/Rgt2 pathway and regulates Cln-dependent
in favor of repressor function (289). Elimination of both Mth1 Cdc28 function (209). Little is yet known about how Grr1
and Std1 appears to be required for maximal derepression of might integrate glucose signaling with cell cycle regulation, but
Rgt1 target genes (202). it now seems clear that it responds directly to glucose sensing
The central events in the Snf3/Rgt2 pathway, from glucose by Snf3 and Rgt2 at the plasma membrane. Second is the PKA
signal generation at the plasma membrane to signal reception pathway, which has long been considered a general regulator
by a DNA-bound repressor (179, 247), are depicted in Fig. 4. of cell growth in any carbon source and as such is thought to
As identified by global profiling (182), Snf3/Rgt2 signaling also participate in the dramatic increase in the rate of cell division
initiates feedback and feedforward regulatory loops that are upon the appearance of fermentable sugars, including glucose.
intertwined with the Snf1-dependent pathway. The Snf3/Rgt2 As discussed above, many components of the PKA pathway
268 SANTANGELO MICROBIOL. MOL. BIOL. REV.

that were originally identified in connection with their role in participates directly in an activation mechanism shared by this
glucose signaling have now been thoroughly characterized. common set of target genes. First, Gcr1 activation of glycolytic
Progress has also been made in understanding the ultimate and RP genes is eliminated by point mutations or small dele-
recipients of the glucose response in the yeast nucleus; PKA tions that eliminate the Rap1 binding site(s) in the correspond-
signaling increases transcription of Rap1 target genes and ap- ing promoters (374, 375). Thus, the capacity of Gcr1 to stim-
pears to operate through the newly identified Rap1 coregula- ulate transcription of most if not all of its target genes is
tors Fhl1/Ifh1 as well as the Rap1/Gcr1/Gcr2 transcriptional entirely dependent upon Rap1 binding upstream of those
activation assemblage (81, 83, 188, 320, 328, 402, 416). Target genes. Second, mutational inactivation of leucine zipper-me-
genes activated by Rap1 fall into two main groups, glycolytic diated Gcr1 homodimerization eliminates both the GCR1 con-
and ribosomal protein (RP) genes (211). Not surprisingly, tribution to activation of Rap1 target genes and complemen-
these are the genes most abundantly transcribed by RNA poly- tation of the gcr1⌬ phenotype (81, 82). Indeed, loss of Gcr1
merase II in S. cerevisiae cultures growing rapidly on glucose; function and reduced transcriptional activation of Rap1 target
26 of the 30 most highly expressed transcription units are in genes exhibit a tight correlation across an entire spectrum of
one of these two groups (392). Overall the Rap1 activation lesions that cause mild to severe impairment. Finally, fluores-

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


mechanism(s) accounts for 60 to 75% of RNA polymerase II cently tagged Rap1 and Gcr1 colocalize to foci in the yeast
transcription in rapidly growing cells (324, 410). nucleus in live cells (236), and epitope-tagged Rap1 and Gcr1
coimmunoprecipitate (375) and can be detected in a large
(⬎1-MDa) complex by size affinity chromatography; this Rap1
Transcriptional Induction of Glycolytic and
complex exhibits faster migration in the absence of Gcr1, in-
Ribosomal Protein Genes
dicating that it is not an aggregation artifact (H. Zhou and
Rap1 is an essential DNA binding protein required for both G. M. Santangelo, unpublished data).
transcriptional repression and activation of target genes (128, Despite these data and other experimental support for the
339); interestingly, it also binds to telomeres and negatively hypothesis that Rap1 and Gcr1 collaborate directly to stimu-
regulates telomere length (for a review, see reference 344). late transcription of both glycolytic and RP genes, it has been
The promoters of most genes encoding glycolytic enzymes difficult to rule out a competing idea, that the effect of GCR1
or ribosomal proteins contain sites occupied by Rap1 in vivo deletion on RP gene expression is indirect. This alternative
(211). The Rap1 site appears to mediate the response of hypothesis is based on the observation of coordinate regulation
these genes to the appearance or disappearance of glucose; of the 138 RP genes in S. cerevisiae. Coordinate regulation
for example, their expression is coordinately down-regu- links transcription of RP and other translational component
lated upon depletion of glucose at the diauxic shift (85). The genes to the growth rate of yeast cells (93, 185, 409). The idea
PKA and TOR signaling pathways are thought to regulate that removal of Gcr1 indirectly causes a drop in RP transcrip-
this response of Rap1-activated genes to nutritional status tion involves the following reasoning: the reduction in glyco-
(328, 402, 410, 441). lytic gene transcription, which is a direct result of GCR1 dele-
Recent reports suggest that the PKA and TOR pathways tion, causes a severe defect (slow growth relative to wild-type
intersect with respect to their shared role in stimulating RP cells); this low growth rate in turn causes reduced RP gene
expression. The transcription factors Sfp1, Fhl1, Ifh1, and Crf1 transcription via a Gcr1-independent mechanism of coordinate
appear to be key downstream targets of this signaling in the regulation. According to this hypothesis, the rationale for as-
yeast nucleus, and various unified models suggest how PKA signing the “sick phenotype” label to the gcr1⌬ mutant is the
and TOR signaling might intersect to regulate cell cycle pro- supposition that a bottleneck in energy generation takes place:
gression by controlling the rate of ribosome production (181, reduced glycolytic flux in glucose-grown gcr1⌬ cells impairs
226, 292). Although additional work is needed to clarify the anaerobic metabolism, while glucose repression of aerobic me-
roles of these newly identified transcriptional regulators, to- tabolism remains intact. This unfortunate combination would
gether they appear to comprise the downstream targets of leave cells with no good way to generate energy, thereby ex-
nutrient sensing through the TOR pathway and thus are the plaining the slow growth and indirect reduction in RP tran-
most likely recipients of signaling by the anticancer agent rapa- scription.
mycin. In contrast, the Rap1 coactivator Gcr1 appears to re- Three independent lines of evidence demonstrate that this
spond only to PKA; gcr1⌬ cells exhibit no alteration in rapa- alternative hypothesis is inviable. First, it is now clear that
mycin sensitivity or rapamycin-induced down-regulation of RP glucose repression is not intact in gcr1⌬ cells (327, 383). Sec-
gene expression (S. J. Deminoff, K. A. Willis, and G. M. San- ond, analysis of strains that harbor partially functional GCR1
tangelo, unpublished data). Like Fhl1/Ifh1, Gcr1 makes an alleles identified several that grow and ferment glucose almost
important contribution to nutrient sensing, since its removal as well as the wild type but nevertheless exhibit RP transcrip-
causes a long delay in the glucose response (as measured by tion as defective as that observed in gcr1⌬ cells (81). Third, the
upshift of RP gene transcription, polysome formation, trans- gcr1⌬ defect in RP transcription is equivalent in all ferment-
lation rate, and growth rate [416; see below]). able carbon sources tested thus far (glucose, sucrose, raffinose,
Transcriptional activation by Rap1 was shown long ago to and galactose), which, significantly (see above), includes non-
exhibit sensitivity to disruption of GCR1 (81, 324, 435). Ge- repressing sugars and pyruvate, a nonfermentable carbon
nome-wide analysis has now demonstrated for the entire ge- source (Fig. 5). Together these data eliminate the key under-
nome that genes with a Rap1 binding site in their promoter are lying assumption of the alternative hypothesis, i.e., that in the
also Gcr1 target genes (81, 211, 324, 375, 435). A wealth of absence of Gcr1, glucose repression and/or slow growth causes
additional evidence suggests that the Rap1/Gcr1 assemblage an indirect effect on RP expression.
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 269

is whether (and to what extent) the respective Sfp1/Fhl1/Ifh1


and Gcr1/Gcr2 contributions to Rap1 transcriptional activa-
tion and cell cycle control are coordinated.

The Repressor/Activator Theme in Glucose Regulation


Although some information has been gleaned regarding the
all-important events downstream of the transcriptional regula-
tory factors that mediate repression and derepression of glu-
cose-responsive target genes, a more than rudimentary under-
standing of the central mechanism(s) of gene regulation in S.
cerevisiae and other eukaryotes has thus far eluded our grasp.
Like Mig1, Mig3, Nrg1, and Rgt1, many of the factors that bind
DNA either directly or indirectly are capable of both activating

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


and repressing transcription. This puzzling property of glucose
repressors is also true of positively acting regulators such as
Rap1 and Gcr1, as well as RNA polymerase II holoenzyme
components such as Med1, Ssn8, Sin4, and Rgr1 (15, 58, 210,
242, 346).
How a distinction is made between these diametrically op-
posite activities of activation and repression based on chromo-
somal and/or environmental context, as well as the molecular
mechanisms that execute this decision, awaits clarification. For
example, an unambiguous finding regarding the mechanism of
FIG. 5. Transcriptomic analysis of defective gene expression in glucose repression in yeast cells is that the Ssn6/Tup1 core-
gcr1⌬ cells. Microarray hybridization was done with RNA from iso- pressor complex plays an important role downstream of most
genic gcr1⌬ versus wild-type cultures grown to mid-logarithmic phase of the factors that bind DNA directly, such as the MIG-, NRG-,
(a density of 1 ⫻ 106 cells/ml) on each of the carbon sources indicated and RGT1-encoded repressors. However, upon derepression
(2% glucose, 3% sucrose, or 3% pyruvate). Total RNA was extracted
both Mig1 and Ssn6/Tup1 remain associated with glucose-
(408) and labeled with either Cy3-CTP or Cy5-CTP by using a low-
RNA-input fluorescent linear amplification kit (Agilent Technologies, repressed promoters (29, 278, 279). Further, even Ssn6 appears
Palo Alto, CA). Labeled RNA was purified with the RNeasy MinElute to be capable of stimulating transcription under some circum-
kit (QIAGEN) and hybridized to yeast 60-mer oligonucleotide arrays stances (68). Thus, most if not all of the relevant negatively
(Agilent Technologies, Palo Alto, CA) according to the manufactur- acting regulators remain associated with target promoters but
er’s recommendations. Array slides were scanned at 10-␮m resolution
with two-line averaging by using an Axon GenePix 4200A scanner and are somehow inactivated (or their activities are reversed) un-
GenePix 6.0 software. Hierarchical clustering was done with Acuity der derepressing conditions. This is a much more subtle regu-
4.0, with a centered Pearson similarity metric. The data shown here latory mechanism, and it operates on a much more static pro-
represent an average of two replicate arrays, including a dye-swap tein-DNA complex, than was previously believed to exist. The
control. The scale shows color intensity proportional to the log ratio of
final sections of this review will describe a new paradigm for
expression in gcr1⌬ cells (relative to the corresponding wild-type val-
ues). The cluster diagram shows the results for the 192 genes (11 eukaryotic gene activation and regulation that may provide a
glycolytic, 110 ribosomal protein, 4 translational component, and 67 superior context in which to understand these and other inter-
other genes) that, in all three carbon sources, exhibit at least a twofold esting new observations about glucose regulation in the yeast
decrease in expression in gcr1⌬ cells. nucleus.

Reverse Recruitment: Rap1/Gcr1 Activation


Therefore, especially when combined with new insights con- at the Nuclear Periphery
cerning the shared participation of Rap1, Gcr1, and Gcr2 in
perinuclear gene regulation (see below), it is now apparent Beyond its interesting regulatory role in yeast cell physiol-
that these global regulators work together to stimulate tran- ogy, a second mystery has shrouded the Rap1/Gcr1 mechanism
scription of both glycolytic and RP genes, the two largest and of transcriptional activation: it conforms poorly with the re-
most abundantly transcribed classes of glucose-induced target cruitment paradigm of gene regulation (36, 70, 295, 296, 352,
genes in yeast cells. Much work remains in deciphering the 434). For example, when a Rap1 binding site is placed up-
physical organization and relative contributions of the Rap1/ stream from heterologous reporter genes, or when the activity
Fhl1/Ifh1 and Rap1/Gcr1/Gcr2 assemblages and how they are of Rap1-Gal4 or Rap1-LexA fusion proteins is measured,
signaled by the PKA and TOR pathways in response to the Rap1 activation is surprisingly weak (in some contexts it is
appearance of glucose. Significantly in this regard, the conse- undetectable) (38, 88, 349, 415). In contrast, mutational inac-
quences of removing Gcr1 include a dramatic drop in transla- tivation of Rap1 binding sites in the very strong glycolytic
tion rate, a decrease in CLN transcription, a cln3⌬-like in- promoters typically results in a 10- to 20-fold decrease in tran-
crease in cell size, and conditional cell cycle arrest and scription, suggesting that Rap1 is a powerful activator (see,
elongated morphology in the cln3⌬ and cln1⌬ cln2⌬ back- e.g., reference 374). This discrepancy, with identical Rap1
grounds (83, 416). An important topic for future investigation binding elements yielding very different results depending on
270 SANTANGELO MICROBIOL. MOL. BIOL. REV.

whether they are in their native location or upstream from a chemical mutagens have failed. Further, every mutation in this
reporter, is paradoxical because no yeast protein other than region that inactivates GCR1 function also eliminates Gcr1-
Rap1 binds to its recognition motif in DNA. Moreover, weak LexA activation of a LexA-driven reporter gene. Since Gcr1-
Rap1 activation of reporter genes is not due to a structural dependent transcriptional activation of target genes is elimi-
perturbation resulting from fusion to a heterologous DNA nated by removal of the Rap1 binding site in the corresponding
binding domain; Rap1-LexA fusion proteins that can fully promoters, this was originally interpreted to mean that contact
complement a deletion of endogenous RAP1 show little or no between Rap1 and Gcr1 is a requirement for the latter to
heterologous activity as transcriptional activators. This was recruit the transcriptional machinery. However, targeted mu-
originally thought to indicate a requirement for additional tagenesis of RAP1 has been equally unsuccessful in generating
DNA-bound proteins (such as chromatin-remodeling factors) detectable suppression of lesions in Gcr1 that impair activa-
for full activation by Rap1 and its coactivators, but to date no tion. Furthermore, activation of a heterologous reporter by
arrangement of DNA motifs has been described that allows an Gcr1 persists long after the disappearance of Rap1 in RAP1ts
isolated Rap1 site to promote transcription as efficiently as its strains. Together these data suggest that when Gcr1 is bound
native counterparts in glycolytic promoters. upstream from a reporter gene, its Rap1 interaction domain is

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


An alternative explanation, which remains untested, is that required but (paradoxically) Rap1 itself is not. The large body
cooperativity resulting from coupling of the various steps in of existing data regarding the Gcr2 contribution to the Rap1/
transcription (i.e., activation, elongation, termination, process- Gcr1 activation mechanism has likewise been difficult to rec-
ing, and export of the final products to the cytoplasm [225]) oncile with the recruitment paradigm (81, 435). Thus, all at-
explains the impressive strength of Rap1 activation when the tempts to resist the straightforward interpretation of an
protein is bound at its native sites. Inclusion of mRNA export activation domain that overlaps with transmembrane domains,
in such a holistic mechanism of gene expression implies a i.e., that Gcr1 association with the nuclear membrane is an
functional and/or physical connection with nuclear pores. This important feature of its transcriptional activation function,
“gene expression machine” (GEM) explanation for the dis- have failed.
crepancy in Rap1 activation efficiency is plausible because a A recent genome-wide genetic screen (synthetic genetic ar-
pas de deux between chromatin and GEMs, an arrangement
ray analysis; see above) of gcr1⌬ and gcr2⌬ query strains
that could have evolved to optimize expression of naturally
yielded a surprising result that has finally provided an im-
occurring promoter-open reading frame combinations whose
proved context for resolving these disparities and elucidating
products are in high demand, would be difficult to reproduce
the mechanism of Rap1/Gcr1 transcriptional activation. Both
with a heterologous gene. (A similar discrepancy has been
GCR1 and GCR2 were found to be members of a large genetic
observed for transcriptional repression by Ssn6/Tup1, which is
network of nucleoporin genes (236). More specifically, dele-
much stronger in natural promoters than in artificial constructs
tion of each nonessential gene encoding a component of the
[194, 203]). Since most GEMs in glucose-grown cells appear
Nup84 subcomplex exhibits a synthetic genetic defect in com-
to be occupied with Rap1-regulated transcription units (see
bination with GCR1 deletion. Follow-up biochemical, cell bi-
above) (392, 416), this would explain why Rap1 activation (and
ological, and gene expression analyses confirmed that Rap1/
perhaps all glucose-regulated transcription) is unusually diffi-
Gcr1 activation occurs at the nuclear periphery and is impaired
cult to reproduce via the ubiquitous heterologous reporter
assay. An important corollary of this idea is that an accurate by disruption of genes encoding components of the Nup84
working model for gene regulation will require detailed map- subcomplex. For example, Rap1 and Gcr1 are 100% and 71%
ping of nuclear architecture (169, 200, 238–240, 280, 281). as enriched in nuclear envelope fractions as the integral nu-
Interestingly, the “gene gating” hypothesis proposed by clear membrane protein Pom152, unlike control proteins such
Günther Blobel in 1985 (22) may prove to have been a partic- as the nucleolar factor Nop1 (⬍1% enriched) (236). Impor-
ularly perceptive early suggestion for how three-dimensional tantly, perinuclear localization of Rap1/Gcr1 target genes has
structure in the nucleus is used to mediate (and regulate) gene been observed via chromatin immunoprecipitation of nuclear
expression. pore-associated genes (48), which provides independent sup-
A second prominent aspect of the Rap1 activation mecha- port for the conclusion that there is a physical and functional
nism that conforms poorly with the recruitment model of gene link between the Rap1/Gcr1 assemblage and the nuclear rim.
activation is the nature of the Gcr1 activation domain. This Most surprisingly, not only do Rap1 and its coactivators
domain is irreducibly large (280 residues) and can be subdi- Gcr1 and Gcr2 activate transcription at the nuclear periphery,
vided into at least four distinct hypomutable regions (82, 83); but many yeast nucleoporins function as transcriptional acti-
two of these hypomutable regions coincide with predicted vators; two of these were shown to do so at their normal
transmembrane domains (236), and transmembrane-disrupt- perinuclear location (236). Transcriptional activation by nu-
ing lesions in these domains are known to inactivate both cleoporins is severely impaired by GCR1 deletion, suggest-
GCR1 function and Gcr1-LexA activation of a heterologous ing that disruption of the Rap1 activation assemblage inter-
reporter gene (82). If, as predicted by the recruitment model, feres with the normal organization of GEMs at the yeast
these transmembrane motifs are required for incorporation of nuclear periphery. This result may recapitulate a phenomenon
Gcr1 into its activation complex, it should have been possible relevant to human cancer. Perinuclear gene activation appears
to make compensatory extragenic lesions that would suppress to be oncogenic in acute myeloid leukemia and related syn-
at least one of the inactivating mutations. Exhaustive screens dromes in humans, in which chromosomal rearrangements
for such suppressing lesions by using either irradiation or fuse Hox DNA binding domains to the nucleoporin hNup98
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 271

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


FIG. 6. Both Gcr1 and Rpo21 are related to the RNA polymerase II large-subunit gene from M. invertens. The N terminus and carboxy-terminal
domain (CTD) of Rpo21 exhibit strong homology with their M. invertens counterparts, whereas the internal region of the M. invertens protein
exhibits weaker (although highly significant) homology with both Rpo21 and Gcr1. The transmembrane domains of Gcr1 (TM1 and TM2), as well
as its primary homodimerization domain (1LZ), all of which make a critical contribution to Gcr1 function (see the text), exhibit ⬎80% similarity
with the M. invertens RNA polymerase large subunit, as indicated.

(126, 183). Interestingly, human Nup98 also activates tran- discovery of a functional connection between the Rap1/Gcr1/
scription in yeast cells, and its ability to do so is impaired in the Gcr2 activation assemblage and GEMs at the nuclear rim, the
absence of Gcr1 (236). Integral GEM components, including reverse recruitment model does not imply that all activation
several Mediator subunits (Ssn8, Sin4, and Med1) and Rpo21, occurs at the nuclear periphery. Indeed, promyelocytic leuke-
also associate with the nuclear periphery and participate in mia protein bodies in the nuclear lumina of mammalian cells
perinuclear activation. (61, 290, 399) could represent nodes of GEM organization that
These data indicate the existence of an unusually intimate parallel what we have observed to occur at nuclear pores in
relationship between the Rap1/Gcr1 assemblage and GEM yeast cells. In this regard, the reverse recruitment idea is con-
components, a conclusion that is further suggested by protein sistent with an earlier proposal by Hutchison and Weintraub
sequence similarity searches. For example, PSI-BLAST analy- that active eukaryotic genes are localized along interchromatin
sis of Gcr1 identified homology with the RNA polymerase II channels that communicate with the nuclear periphery (167).
large subunit in the free-living protist Mastigamoeba invertens Thus, the central feature of the reverse recruitment idea is that
(Fig. 6) (351). The highly significant E value of this pairing is genes become transcriptionally active by docking with com-
2.0 ⫻ 10⫺30 (marginal significance is attained at an E value of partmentalized GEMs. Although they are relatively abundant
at the periphery, it seems likely that at least some GEMs are
0.05 [5]). M. invertens and other primitively amitochondriate
luminal; these internal GEMs may nevertheless maintain func-
members of the order Pelobiontida may represent the earliest
tional and/or physical communication with their perinuclear
eukaryotic lineages (351). Thus, the RNA polymerase II large-
counterparts.
subunit gene in M. invertens could represent a composite pre-
The reverse recruitment idea explains much or all of the
cursor of both Rpo21 and Gcr1 (Fig. 6). This potential primor-
data that seemed paradoxical when attempting to fit them
dial connection between nuclear pores and Rap1/Gcr1/Gcr2
within the recruitment model. It presumes that Rap1 and Gcr1
function may help to explain the lack of evolutionary conser-
are separate cogs in a large and multifaceted GEM and that
vation of the latter; divergence of the nucleoporin Nup96 has the site of Gcr1 attachment to this megalith is an extensive
been shown to cause hybrid inviability in Drosophila and may hydrophobic interaction with either the Nup84 subcomplex, an
therefore contribute to speciation (293). adjacent site on the nuclear envelope itself, or both (Fig. 7).
These new data begin to place the Rap1/Gcr1 activation Thus, there may be two types of reverse recruitment in S.
assemblage within its proper architectural context in the yeast cerevisiae, i.e., transient occupation of GEMs by tightly regu-
nucleus and (at least where this unusual regulatory complex is lated (and often weakly expressed) genes and constitutive oc-
concerned) suggest an interesting alternative to the recruit- cupation by strongly expressed genes that bind directly to a
ment model of transcriptional activation. We have termed this GEM component; the Rap1/Gcr1 mechanism is in the latter
alternative paradigm “reverse recruitment” to highlight its cen- category. According to this idea, regulation of Rap1/Gcr1 tar-
tral feature, that Rap1/Gcr1 target genes are recruited to pe- get genes, which clearly does occur (e.g., upshift upon the
rinuclear GEMs; the term references the opposite presump- appearance of glucose), might involve a novel type of regula-
tion of the recruitment model, that the requisite factors are tory control: alteration of the functional or physical relation-
drawn to (or assembled on) the relevant promoters. Impor- ship between perinuclear GEMs and the Rap1/Gcr1 assem-
tantly, although this mechanism was uncovered through the blage.
272 SANTANGELO MICROBIOL. MOL. BIOL. REV.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


FIG. 7. Perinuclear transcriptional activation of Rap1/Gcr1 target genes via reverse recruitment. Known components of the Rap1 activation
(Gcr1/Gcr2) or silencing (Sir complex) assemblages are shown. Essential nucleoporins are indicated with an asterisk. Dashed lines highlight
presumptive perinuclear tethering interactions; nuclear pore complex-associated factors shown to interact with Gcr1 are underlined. The
representation shown here is not intended to rule out the existence of a unified complex that can switch between activation and repression of
transcription. (Reprinted from reference 236 with permission of the publisher. Copyright 2005 National Academy of Sciences, U.S.A.)

GENE REGULATION VIA REVERSE RECRUITMENT Rap1 repression of target genes upon disruption of the secre-
tory pathway (208) and Rgt1 activation of HXT1 (see above).
A New Model for Eukaryotic Transcriptional Activation
Ghost regulation cannot be reconciled with a competitor of the
and Repression
reverse recruitment idea that postulates an alternative expla-
As described above, the existing data suggest that reverse nation for the perinuclear location of activated GAL and INO
recruitment accounts for the bulk of transcriptional activation genes, i.e., that transcription initiation complexes form as pre-
in growing yeast cells. What about more conventional regula-
tory systems: does reverse recruitment explain all types of
transcriptional activation and regulation in eukaryotic nuclei?
Data favoring this conclusion are already available for two
conventional regulatory mechanisms, INO and GAL (32, 48)
(Fig. 8). With respect to the well-studied GAL system, tran-
scriptional activation in galactose-grown cells is accompanied
by association between GAL genes and the nuclear periphery,
which is dramatically reduced in glucose-grown cells (48). This
result is particularly significant because analyses of galactose-
regulated gene expression and the Gal4 activator were used
initially to establish the recruitment paradigm. The link be-
tween transcriptional activation and perinuclear association
has also been reported for INO1 (32). Thus, a growing body of
evidence supports the ideas that a link exists between the
nuclear periphery and transcriptional activation and that re-
versible association with the nuclear rim may be an integral
feature of all eukaryotic gene regulation (see also a recent
analysis of var genes in Plasmodium falciparum [301]).
FIG. 8. Perinuclear transcriptional activation of GAL genes via re-
Universal application of the reverse recruitment paradigm verse recruitment. The glucose-regulated transcriptional activator
may provide a context in which to understand numerous per- Gal4 associates with its DNA binding sites whether the downstream
plexing phenomena that have heretofore defied explanation, target genes are inactive or active, as shown. The GAL1, GAL7, and
most importantly the capacity of regulators to exert influence GAL10 genes are not detectably associated with the nuclear periphery
under conditions (in glucose-grown cells) in which their transcription is
on gene transcription under circumstances where either direct repressed (UNINDUCED, left side). When cells are grown in the
or indirect contact with the DNA is thought to be unlikely. The presence of galactose, these same GAL genes associate with the nu-
best-characterized examples of such “ghost regulation” are clear periphery (INDUCED, right side).
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 273

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


FIG. 9. A model for glucose regulation at the nuclear periphery of S. cerevisiae. Numerous regulators involved in the transcriptomic response
to glucose are components of a large nuclear assemblage (gene expression machine) that is tethered to nuclear pores (dashed lines) via multiple
interactions with the Nup84 subcomplex. Red arrows depict repressed target genes, and green arrows depict activated targets. Inactivation of
DNA-bound regulators (Mig1, Mig3, and Nrg1) by the Snf1 kinase is predicted to involve a subtle conformational alteration that remains obscure.
Many of these glucose regulatory factors, including components of the RNA polymerase II Mediator, are bifunctional factors that can either
repress or activate transcription in a context-dependent fashion (shown here for Rap1). This facile switching between transcriptional readouts
underscores the mechanistic subtlety of regulatory partnering and/or conformational specificity. Association between nuclear pores and the
Gcr1/Gcr2 complex is required for transcriptional activation by both Rap1 and Nup84 subcomplex components (229). The perinuclear Rap1/
Gcr1/Gcr2 assemblage thus allows productive access to GEMs by glucose-induced (glycolytic and RP) genes tethered at the nuclear periphery.

dicted by the recruitment model and move to the periphery cruitment model requires them to assemble in the proper
only during the final stages of gene expression (i.e., during sequence (i.e., in a temporally flawless fashion) at each pro-
mRNA processing and export). moter, or to preassemble and then bind as a complex or series
of complexes, in order to obtain the proper transcriptional
readout for each corresponding gene. Indeed, a recent ge-
Glucose May Signal a Highly Networked and Structurally
nome-wide analysis of quiescence revealed that, in contrast
Stable Perinuclear Machine
with the recruitment model, RNA polymerase II is already
Another recent development favoring the reverse recruit- positioned upstream of many inactive genes prior to their in-
ment idea is that proteins capable of both induction and re- duction upon exit from stationary phase (298).
pression of transcription, including many involved in glucose With respect to glucose regulation in particular, it is also
regulation (e.g., Rap1, Gcr1, Snf1, Mig1, Mig3, Nrg1, Rgt1, now apparent that DNA-bound transcriptional activators,
Ssn6, Med1, Ssn8, Sin4, Rgr1, and Fhl1), appear to be the rule readily identified in some systems (e.g., Gal4), may be curi-
rather than the exception. We have now shown that many of ously missing from others. Given that SUC2 has been a model
these factors are concentrated at the periphery of the yeast target gene in the study of glucose repression, it is reasonable
nucleus (236; B. B. Menon and G. M. Santangelo, unpublished to have expected by now the identification of a DNA-bound
data). It is increasingly clear that subtle allosteric alteration of SUC2 activator. (Although it seems unlikely, it remains a for-
bifunctional activator/repressor-type regulators is a common mal possible that the gene encoding the SUC2 activator protein
mechanism responsible for major regulatory shifts in the tran- has simply eluded detection.) The very successful screens for
scriptome (e.g., at Snf1/Mig1/Ssn6 targets). Reverse recruit- non-sucrose-fermenting mutants (46, 259, 385) have instead
ment has the advantage of allowing a straightforward and par- identified the critically important kinase subunits Snf1 and
simonious depiction of this type of subtle gene regulation Snf4, a putative glucose sensor localized to the plasma mem-
mechanism (Fig. 9), a task that has become more and more brane (Snf3), endosomal factors (Snf7 and Snf8 [148, 287]),
difficult and convoluted when one adheres to the recruitment and components of the Swi/Snf chromatin remodeling complex
paradigm. This is in part because the latter requires so many (Snf2, Snf5, Snf6, Snf11, and Snf12). None of these proteins
factors to assemble in the correct configuration at each pro- are thought to bind DNA directly. Subsequent screens for
moter. It is especially so when also considering that the re- suppressors of Snf1 (47, 386) identified two critical down-
274 SANTANGELO MICROBIOL. MOL. BIOL. REV.

stream factors in glucose repression, Mig1 (Ssn1) and Ssn6; the invertens homolog, to Kellie Barbara for graciously contributing the
products of the other six SSN alleles associate biochemically microarray data shown in Fig. 5, to Kellie Barbara and Nicole Thomp-
son for help with literature searches, and to Kristine Willis, Jeong-Ho
with RNA polymerase II and participate in the Mediator- Kim, Kelly Tatchell, Lucy Robinson, and Brenda Andrews for review-
based mechanism of glucose repression by the Ssn6/Tup1 core- ing the manuscript, as well as for their keen insight and tolerance. Most
pressor: Ssn2, Ssn3, Sin4 (Ssn4), Srb8 (Ssn5), Rox3 (Ssn7), importantly I thank the current and past members of the Santangelo
and Ssn8. laboratory for their perspiration, inspiration, and helpful suggestions.
Thus, of all these Snf and Ssn loci, the only protein identified Finally, kudos and thanks to the many fine biomedical researchers in
Mississippi with whom I have collaborated during my tenure as Direc-
to date that exhibits specific DNA binding activity is the zinc tor of the Mississippi Functional Genomics Network (mfgn.usm.edu
finger protein Mig1. Mig1 (and its paralogs Mig2 and Mig3) /mfgn).
bind to a GC core element upstream from SUC2 and other Work in my laboratory is supported by National Institutes of Health
glucose-regulated genes. Mig1 and Mig3, like most of the other award RR16476.
transcription factors identified in Snf and Ssn screens, are REFERENCES
known to function as both repressors and activators. Never-
1. Ahn, J. H., S. H. Park, and H. S. Kang. 1995. Inactivation of the UAS1 of
theless, Mig1 is presumed not to participate in activation of STA1 by glucose and STA10 and identification of two loci, SNS1 and MSS1,

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


SUC2, which it binds to and represses in the presence of glu- involved in STA10-dependent repression in Saccharomyces cerevisiae. Mol.
Gen. Genet. 246:529–537.
cose. This conclusion is based on three assumptions that are 2. Ahuatzi, D., P. Herrero, T. de la Cera, and F. Moreno. 2004. The glucose-
well worth revisiting. First, SUC2 is derepressed normally in regulated nuclear localization of hexokinase 2 in Saccharomyces cerevisiae is
the absence of MIG1 (and/or MIG2). However, the redun- Mig1-dependent. J. Biol. Chem. 279:14440–14446.
3. Alberghina, L., R. L. Rossi, L. Querin, V. Wanke, and M. Vanoni. 2004. A
dancy of factors that bind to similar GC recognition sites in cell sizer network involving Cln3 and Far1 controls entrance into S phase in
DNA makes it difficult to construct a strain in which all regu- the mitotic cycle of budding yeast. J. Cell Biol. 167:433–443.
lators that could bind and activate transcription are known to 4. Alms, G. R., P. Sanz, M. Carlson, and T. A. Haystead. 1999. Reg1p targets
protein phosphatase 1 to dephosphorylate hexokinase II in Saccharomyces
be eliminated. Thus, it is quite possible that MIG deletion cerevisiae: characterizing the effects of a phosphatase subunit on the yeast
uncovers a cryptic activator function in the SUC2 promoter. proteome. EMBO J. 18:4157–4168.
5. Altschul, S. F., T. L. Madden, A. A. Schaffer, J. Zhang, Z. Zhang, W. Miller,
Second, Mig1 relocates to the cytoplasm in the absence of and D. J. Lipman. 1997. Gapped BLAST and PSI-BLAST: a new genera-
glucose. However, the Mig1 molecules remaining in the nu- tion of protein database search programs. Nucleic Acids Res. 25:3389–3402.
cleus are sufficient to occupy binding sites upstream of Mig1 6. Andrews, P. D., and M. J. Stark. 2000. Type 1 protein phosphatase is
required for maintenance of cell wall integrity, morphogenesis and cell
target genes (see above). Third, removal of the Mig1 recogni- cycle progression in Saccharomyces cerevisiae. J. Cell Sci. 113:507–520.
tion motif in the promoter does not impair SUC2 derepression. 7. Ashe, M. P., S. K. De Long, and A. B. Sachs. 2000. Glucose depletion
However, this argument rests on the assumption that the re- rapidly inhibits translation initiation in yeast. Mol. Biol. Cell 11:833–848.
8. Ashrafi, K., S. S. Lin, J. K. Manchester, and J. I. Gordon. 2000. Sip2p and
cruitment model of transcriptional activation is correct, and its partner Snf1p kinase affect aging in S. cerevisiae. Genes Dev. 14:1872–
that once binding of those factors to their respective DNA 1885.
9. Babu, P., J. D. Bryan, H. R. Panek, S. L. Jordan, B. M. Forbrich, S. C.
recognition sites is blocked, they no longer remain in the vi- Kelley, R. T. Colvin, and L. C. Robinson. 2002. Plasma membrane local-
cinity of (i.e., tethered to) their respective target genes. In ization of the Yck2p yeast casein kinase 1 isoform requires the C-terminal
contrast, the reverse recruitment hypothesis postulates that a extension and secretory pathway function. J. Cell Sci. 115:4957–4968.
10. Bai, C., P. Sen, K. Hofmann, L. Ma, M. Goebl, J. W. Harper, and S. J.
given regulator may remain in the vicinity of a “tethered” Elledge. 1996. SKP1 connects cell cycle regulators to the ubiquitin prote-
target gene and continue to influence its expression by altering olysis machinery through a novel motif, the F-box. Cell 86:263–274.
the behavior of associated GEMs. This leaves open the possi- 11. Bailey, R. B., and A. Woodword. 1984. Isolation and characterization of a
pleiotropic glucose repression resistant mutant of Saccharomyces cerevisiae.
bility that Mig1 and other factors originally identified as re- Mol. Gen. Genet. 193:507–512.
pressors (e.g., Rgt1) might, in contrast to previous expecta- 12. Bailis, J. M., and G. S. Roeder. 2000. Pachytene exit controlled by reversal
of Mek1-dependent phosphorylation. Cell 14:211–221.
tions, undergo a repressor-to-activator switch and activate 13. Baker, H. V. 1986. Glycolytic gene expression in Saccharomyces cerevisiae:
their target genes under derepressing conditions (Fig. 9). nucleotide sequence of GCR1, null mutants, and evidence for expression.
Reverse recruitment thus provides a viable alternative to an Mol. Cell. Biol. 6:3774–3784.
14. Baker, S. H., D. L. Frederick, A. Bloecher, and K. Tatchell. 1997. Alanine-
increasingly untenable prediction of the diffusion-based re- scanning mutagenesis of protein phosphatase type 1 in the yeast Saccharo-
cruitment model that a disparate collection of activators, re- myces cerevisiae. Genetics 145:615–626.
pressors, coactivators, corepressors, and GEM components are 15. Balciunas, D., C. Galman, H. Ronne, and S. Bjorklund. 1999. The Med1
subunit of the yeast mediator complex is involved in both transcriptional
brought to each promoter as needed to provide the appropri- activation and repression. Proc. Natl. Acad. Sci. USA 96:376–381.
ate regulatory input. An improved understanding of nuclear 16. Barral, Y., S. Jentsch, and C. Mann. 1995. G1 cyclin turnover and nutrient
uptake are controlled by a common pathway in yeast. Genes Dev. 9:399–
substructure and its reception of regulatory signals from the 409.
plasma membrane and cytoplasm is therefore a critical prereq- 17. Belinchon, M. M., and J. M. Gancedo. 2003. Xylose and some non-sugar
uisite to constructing an accurate global wiring diagram for carbon sources cause catabolite repression in Saccharomyces cerevisiae.
Arch. Microbiol. 180:293–297.
glucose signaling in S. cerevisiae and other eukaryotes. Reas- 18. Berkey, C. D., V. K. Vyas, and M. Carlson. 2004. Nrg1 and Nrg2 transcrip-
sessment of the regulatory relationships in other systems will tional repressors are differently regulated in response to carbon source.
likewise become necessary if reverse recruitment is as wide- Eukaryot. Cell 3:311–317.
19. Bhattacharya, S., L. Chen, J. R. Broach, and S. Powers. 1995. Ras mem-
spread an explanation for gene induction and repression as brane targeting is essential for glucose signaling but not for viability in
now seems plausible. yeast. Proc. Natl. Acad. Sci. USA 92:2984–2988.
20. Bisson, L. F., and V. Kunathigan. 2003. On the trail of an elusive flux
sensor. Res. Microbiol. 154:603–610.
ACKNOWLEDGMENTS 21. Blacketer, M. J., P. Madaule, and A. M. Myers. 1995. Mutational analysis
of morphologic differentiation in Saccharomyces cerevisiae. Genetics 140:
I am extremely grateful to Kristine Willis for figure design and 1259–1275.
preparation, to Youping Deng and Venkata Thodima for the graphical 22. Blobel, G. 1985. Gene gating: a hypothesis. Proc. Natl. Acad. Sci. USA
depiction of the ClustalW alignment of Gcr1 and Rpo21 with their M. 82:8527–8529.
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 275

23. Bloecher, A., and K. Tatchell. 1999. Defects in Saccharomyces cerevisiae 52. Celenza, J. L., and M. Carlson. 1989. Mutational analysis of the Saccharo-
protein phosphatase type I activate the spindle/kinetochore checkpoint. myces cerevisiae SNF1 protein kinase and evidence for functional interac-
Genes Dev. 13:517–522. tion with the SNF4 protein. Mol. Cell. Biol. 9:5034–5044.
24. Bloecher, A., and K. Tatchell. 2000. Dynamic localization of protein phos- 53. Celenza, J. L., F. J. Eng, and M. Carlson. 1989. Molecular analysis of the
phatase type 1 in the mitotic cell cycle of Saccharomyces cerevisiae. J. Cell SNF4 gene of Saccharomyces cerevisiae: evidence for physical association of
Biol. 149:125–140. the SNF4 protein with the SNF1 protein kinase. Mol. Cell. Biol. 9:5045–
25. Blumberg, H., A. Eisen, A. Sledziewski, D. Bader, and E. T. Young. 1987. 5054.
Two zinc fingers of a yeast regulatory protein shown by genetic evidence to 54. Chang, J. S., K. Henry, B. L. Wolf, M. Geli, and S. K. Lemmon. 2002.
be essential for its function. Nature 328:443–445. Protein phosphatase-1 binding to Scd5p is important for regulation of actin
26. Bohm, S., D. Frishman, and H. W. Mewes. 1997. Variations of the C2H2 organization and endocytosis in yeast. J. Biol. Chem. 277:48002–48008.
zinc finger motif in the yeast genome and classification of yeast zinc finger 55. Chang, Y. W., S. C. Howard, and P. K. Herman. 2004. The Ras/PKA
proteins. Nucleic Acids Res. 25:2464–2469. signaling pathway directly targets the Srb9p protein, a component of the
27. Bojunga, N., and K. D. Entian. 1999. Cat8p, the activator of gluconeogenic general RNA polymerase II transcription apparatus. Mol. Cell 15:107–116.
genes in Saccharomyces cerevisiae, regulated carbon source-dependent ex- 56. Charbon, G., K. D. Breunig, R. Wattiez, J. Vandenhaute, and I. Noel-
pression of NADP-dependent cytosolic isocitrate dehydrogenase (Idp2p) Georis. 2004. Key role of Ser562/661 in Snf1-dependent regulation of Cat8p
and lactate permease (Jen1p). Mol. Gen. Genet. 262:869–875. in Saccharomyces cerevisiae and Kluyveromyces lactis. Mol. Cell. Biol. 24:
28. Bojunga, N., P. Kotter, and K. D. Entian. 1998. The succinate/fumarate 4083–4091.
transporter Acr1p of Saccharomyces cerevisiae is part of the gluconeogenic 57. Chaves, R. S., P. Herrero, and F. Moreno. 1999. Med8, a subunit of the
pathway and its expression is regulated by Cat8p. Mol. Gen. Genet. 260: mediator CTD complex of RNA polymerase II, directly binds to regulatory
453–461.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


elements of SUC2 and HXK2 genes. Biochem. Biophys. Res. Commun.
29. Boukaba, A., E. I. Georgieva, F. A. Myers, A. W. Thorne, G. Lopez-Rodas, 254:345–350.
C. Crane-Robinson, and L. Franco. 2004. A short-range gradient of histone 58. Chen, S., R. W. West Jr., S. L. Johnson, H. Gans, B. Kruger, and J. Ma.
H3 acetylation and Tup1p redistribution at the promoter of the Saccharo- 1993. TSF3, a global regulatory protein that silences transcription of yeast
myces cerevisiae SUC2 gene. J. Biol. Chem. 279:7678–7684. GAL genes, also mediates repression by ␣2 repressor and is identical to
30. Boy-Marcotte, E., M. Perrot, F. Bussereau, H. Boucherie, and M. Jacquet. SIN4. Mol. Cell. Biol. 13:831–840.
1998. Msn2p and Msn4p control a large number of genes induced at the 59. Cheng, C., D. Huang, P. J. Roach. 1997. Yeast PIG genes: PIG1 encodes a
diauxic transition which are repressed by cyclic AMP in Saccharomyces putative type 1 phosphatase subunit that interacts with the yeast glycogen
cerevisiae. J. Bacteriol. 180:1044–1052. synthase Gsy2p. Yeast 13:1–8.
31. Boy-Marcotte, E., P. Ikonomi, and M. Jacquet. 1996. SDC25, a dispensable 60. Cherry, J. R., T. R. Johnson, C. Dollard, J. R. Shuster, and C. L. Denis.
Ras guanine nucleotide exchange factor of Saccharomyces cerevisiae differs 1989. Cyclic AMP-dependent protein kinase phosphorylates and inactivates
from CDC25 by its regulation. Mol. Biol. Cell 7:529–539. the yeast transcriptional activator ADR1. Cell 56:409–419.
32. Brickner, J. H., and P. Walter. 2004. Gene recruitment of the activated 61. Ching, R. W., G. Dellaire, C. H. Eskiw, and D. P. Bazett-Jones. 2005. PML
INO1 locus to the nuclear membrane. PLoS Biol. 2:1843–1853. bodies: a meeting place for genomic loci? J. Cell Sci. 118:847–854.
33. Bridges, D., and G. B. Moorhead. 2004. 14-3-3 proteins: a number of 62. Ciriacy, M. 1977. Isolation and characterization of yeast mutants defective
functions for a numbered protein. Sci. STKE 2004:re10. in intermediary carbon metabolism and in carbon catabolite derepression.
34. Broek, D., T. Toda, T. Michaeli, L. Levin, C. Birchmeier, M. Zoller, S. Mol. Gen. Genet. 154:213–220.
Powers, and M. Wigler. 1987. The S. cerevisiae CDC25 gene product reg-
63. Clifton, D., and D. G. Fraenkel. 1981. The gcr (glycolysis regulation) mu-
ulates the RAS/adenylate cyclase pathway. Cell 48:789–799.
tation of Saccharomyces cerevisiae. J. Biol. Chem. 256:13074–13078.
35. Bruckner, R., and F. Titgemeyer. 2002. Carbon catabolite repression in
64. Clifton, D., S. B. Weinstock, and D. B. Fraenkel. 1978. Glycolysis mutants
bacteria: choice of the carbon source and autoregulatory limitation of sugar
in Saccharomyces cerevisiae. Genetics 88:1–11.
utilization. FEMS Microbiol. Lett. 209:141–148.
65. Cohen, P., and P. T. Cohen. 1989. Protein phosphatases come of age.
36. Bryant, G. O., and M. Ptashne. 2003. Independent recruitment in vivo by
J. Biol. Chem. 264:21435–21438.
Gal4 of two complexes required for transcription. Mol. Cell 11:1301–1309.
37. Bu, Y., and M. C. Schmidt. 1998. Identification of cis-acting elements in the 66. Colombo, S., D. Ronchetti, J. M. Thevelein, J. Winderickx, and E. Marte-
SUC2 promoter of Saccharomyces cerevisiae required for activation of tran- gani. 2004. Activation state of the Ras2 protein and glucose-induced sig-
scription. Nucleic Acids Res. 26:1002–1009. naling in Saccharomyces cerevisiae. J. Biol. Chem. 279:46715–46722.
38. Buchman, A. R., N. F. Lue, and R. D. Kornberg. 1988. Connections between 67. Colombo, S., P. Ma, L. Cauwenberg, J. Winderickx, M. Crauwels, A.
transcriptional activators, silencers, and telomeres as revealed by functional Teunissen, D. Nauwelaers, J. H. de Winde, M. F. Gorwa, D. Colavizza, and
analysis of a yeast DNA-binding protein. Mol. Cell. Biol. 8:5086–5099. J. M. Thevelein. 1998. Involvement of distinct G-proteins, Gpa2 and Ras, in
39. Cameron, S., L. Levin, M. Zoller, and M. Wigler. 1988. cAMP-independent glucose- and intracellular acidification-induced cAMP signalling in the
control of sporulation, glycogen metabolism, and heat shock resistance in yeast Saccharomyces cerevisiae. EMBO J. 17:3326–3341.
S. cerevisiae. Cell 53:555–566. 68. Conlan, R. S., N. Gounalaki, P. Hatzis, and D. Tzamarias. 1999. The
40. Cannon, J. F., and K. Tatchell. 1987. Characterization of Saccharomyces Tup1-Cyc8 protein complex can shift from a transcriptional co-repressor to
cerevisiae genes encoding subunits of cyclic AMP-dependent protein kinase. a transcriptional co-activator. J. Biol. Chem. 274:205–210.
Mol. Cell. Biol. 7:2653–2663. 69. Coons, D. M., P. Vagnoli, and L. F. Bisson. 1997. The C-terminal domain
41. Cannon, J. F., J. B. Gibbs, and K. Tatchell. 1986. Suppressors of the ras2 of Snf3p is sufficient to complement the growth defect of snf3 null muta-
mutation of Saccharomyces cerevisiae. Genetics 113:247–264. tions in Saccharomyces cerevisiae: SNF3 functions in glucose recognition.
42. Cannon, J. F., J. R. Pringle, A. Fiechter, and M. Khalil. 1994. Character- Yeast 13:9–20.
ization of glycogen-deficient glc mutants of Saccharomyces cerevisiae. Ge- 70. Cosma, M. P. 2002. Ordered recruitment: gene-specific mechanism of tran-
netics 136:485–503. scription activation. Mol. Cell 10:227–236.
43. Carlson, M. 1997. Genetics of transcriptional regulation in yeast: connec- 71. Crabtree, H. G. 1929. Observation of the carbohydrate metabolism of
tions to the RNA polymerase II CTD. Annu. Rev. Cell Dev. Biol. 13:1–23. tumors. Biochem. J. 23:536–545.
44. Carlson, M. 1998. Regulation of glucose utilization in yeast. Curr. Opin. 72. Crauwels, M., M. C. Donaton, M. B. Pernambuco, J. Winderickx, J. H. de
Genet. Dev. 8:560–564. Winde, and J. M. Thevelein. 1997. The Sch9 protein kinase in the yeast
45. Carlson, M. 1999. Glucose repression in yeast. Curr. Opin. Microbiol. Saccharomyces cerevisiae controls cAPK activity and is required for nitrogen
2:202–207. activation of the fermentable-growth-medium-induced (FGM) pathway.
46. Carlson, M., B. C. Osmond, and D. Botstein. 1981. Mutants of yeast de- Microbiology 143:2627–2637.
fective in sucrose utilization. Genetics 98:25–40. 73. Crechet, J. B., E. Jacquet, A. Bernardi, and A. Parmeggiani. 2000. Analysis
47. Carlson, M., B. C. Osmond, L. Neigeborn, and D. Botstein. 1984. A sup- of the role of the hypervariable region of yeast Ras2p and its farnesylation
pressor of SNF1 mutations causes constitutive high-level invertase synthesis in the interaction with exchange factors and adenylyl cyclase. J. Biol. Chem.
in yeast. Genetics 107:19–32. 275:17754–17761.
48. Casolari, J. M., C. R. Brown, S. Komili, J. West, H. Hieronymus, and P. A. 74. Crechet, J. B., R. H. Cool, E. Jacquet, and J. Y. Lallemand. 2003. Charac-
Silver. 2004. Genome-wide localization of the nuclear transport machinery terization of Saccharomyces cerevisiae Ras1p and chimaeric constructs of
couples transcriptional status and nuclear organization. Cell 117:427–439. Ras proteins reveals the hypervariable region and farnesylation as critical
49. Celenza, J. L., and M. Carlson. 1984. Cloning and genetic mapping of elements in the adenylyl cylase signaling pathway. Biochemistry 42:14903–
SNF1, a gene required for expression of glucose-repressible genes in Sac- 14912.
charomyces cerevisiae. Mol. Cell. Biol. 4:49–53. 75. Crespo, J. L., and M. N. Hall. 2002. Elucidating TOR signaling and rapa-
50. Celenza, J. L., and M. Carlson. 1984. Structure and expression of the SNF1 mycin action: lessons from Saccharomyces cerevisiae. Microbiol. Mol. Biol.
gene of Saccharomyces cerevisiae. Mol. Cell. Biol. 4:54–60. Rev. 66:579–591.
51. Celenza, J. L., and M. Carlson. 1986. A yeast gene that is essential for 76. Cui, D. Y., C. R. Brown, and H. L. Chiang. 2004. The type 1 phosphatase
release from glucose repression encodes a protein kinase. Science 233: Reg1p-Glc7p is required for the glucose-induced degradation of fructose-
1175–1180. 1,6-biphosphatase in the vacuole. J. Biol. Chem. 279:9713–9724.
276 SANTANGELO MICROBIOL. MOL. BIOL. REV.

77. Dale, S., W. A. Wilson, A. M. Edelman, and D. G. Hardie. 1995. Similar 102. Entian, K. D., and F. K. Zimmermann. 1982. New genes involved in carbon
substrate recognition motifs for mammalian AMP-activated protein kinase, catabolite repression and derepression in the yeast Saccharomyces cerevi-
higher plant HMG-CoA reductase kinase-A, yeast SNF1, and mammalian siae. J. Bacteriol. 151:1123–1128.
calmodulin-dependent protein kinase I. FEBS Lett. 361:191–195. 103. Entian, K. D., and J. A. Barnett. 1992. Regulation of sugar utilization by
78. Dalley, B. K., and J. F. Cannon. 1996. Novel, activated RAS mutations alter Saccharomyces cerevisiae. Trends Biochem. Sci. 17:506–510.
protein-protein interactions. Oncogene 13:1209–1220. 104. Entian, K. D., F. K. Zimmermann, and I. Scheel. 1977. A partial defect in
79. Damak, F., E. Boy-Marcotte, D. Le-Roscouet, R. Guilbaud, and M. Jacquet. carbon catabolite repression in mutants of Saccharomyces cerevisiae with
1991. SDC25, a CDC25-like gene which contains a RAS-activating domain reduced hexose phosphorylation. Mol. Gen. Genet. 156:99–105.
and is a dispensable gene of Saccharomyces cerevisiae. Mol. Cell. Biol. 105. Erickson, J. R., and M. Johnston. 1993. Genetic and molecular character-
11:202–212. ization of GAL83: its interaction and similarities with other genes involved
80. de la Cera, T., P. Herrero, F. Moreno-Herrero, R. S. Chaves, and F. in glucose repression in Saccharomyces cerevisiae. Genetics 135:655–664.
Moreno. 2002. Mediator factor Med8p interacts with the hexokinase 2: 106. Erickson, J. R., and M. Johnston. 1994. Suppressors reveal two classes of
implication in the glucose signalling pathway of Saccharomyces cerevisiae. J. glucose repression genes in the yeast Saccharomyces cerevisiae. Genetics
Mol. Biol. 319:703–714. 136:1271–1278.
81. Deminoff, S. J., and G. M. Santangelo. 2001. Rap1p requires Gcr1p and 107. Estruch, F., and M. Carlson. 1990. Increased dosage of the MSN1 gene
Gcr2p homodimers to activate ribosomal protein and glycolytic genes, re- restores invertase expression in yeast mutants defective in the SNF1 protein
spectively. Genetics 158:133–143. kinase. Nucleic Acids Res. 18:6959–6964.
82. Deminoff, S. J., J. Tornow, and G. M. Santangelo. 1995. Unigenic evolution: 108. Estruch, F., M. A. Treitel, X. Yang, and M. Carlson. 1992. N-terminal
a novel genetic method localizes a putative leucine zipper that mediates mutations modulate yeast SNF1 protein kinase function. Genetics 132:639–
650.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


dimerization of the Saccharomyces cerevisiae regulator Gcr1p. Genetics
141:1263–1274. 109. Fabrizio, P., F. Pozza, S. D. Pletcher, C. M. Gendron, and V. D. Longo.
83. Deminoff, S. J., K. Willis, and G. M. Santangelo. 2003. Coordination be- 2001. Regulation of longevity and stress resistance by Sch9 in yeast. Science
tween eukaryotic growth and cell cycle progression: RAP1/GCR1 transcrip- 292:288–290.
tional activation mediates glucose-dependent CLN function. Recent Res. 110. Feldman, R. M., C. C. Correll, K. B. Kaplan, and R. J. Deshaies. 1997. A
Dev. Genet. 3:1–16. complex of Cdc4p, Skp1p, and Cdc53p/cullin catalyzes ubiquitination of the
84. Denis, C. L. 1984. Identification of new genes involved in the regulation of phosphorylated CDK inhibitor Sic1p. Cell 91:221–230.
yeast alcohol dehydrogenase II. Genetics 108:833–844. 111. Feng, Z. H., S. E. Wilson, Z. Y. Peng, K. K. Schlender, E. M. Reimann, and
85. DeRisi, J. L., V. R. Iyer, and P. O. Brown. 1997. Exploring the metabolic R. J. Trumbly. 1991. The yeast GLC7 gene required for glycogen accumu-
and genetic control of gene expression on a genomic scale. Science 278: lation encodes a type 1 protein phosphatase. J. Biol. Chem. 266:23796–
680–686. 23801.
86. DeVit, M. J., and M. Johnston. 1999. The nuclear exportin Msn5 is required 112. Ferguson, S. B., E. S. Anderson, R. B. Harshaw, T. Thate, N. L. Craig, and
for nuclear export of the Mig1 glucose repressor of Saccharomyces cerevi- H. C. Nelson. 2005. Protein kinase A regulates constitutive expression of
siae. Curr. Biol. 9:1231–1241. small heat-shock genes in an Msn2/4p-independent and Hsf1p-dependent
87. DeVit, M. J., J. A. Waddle, and M. Johnston. 1997. Regulated nuclear manner in Saccharomyces cerevisiae. Genetics 169:1203–1214.
translocation of the Mig1 glucose repressor. Mol. Biol. Cell 8:1603–1618. 113. Fields, S., and O. Song. 1989. A novel genetic system to detect protein-
protein interactions. Nature 340:245–246.
88. Devlin, C., K. Tice-Baldwin, D. Shore, and K. T. Arndt. 1991. RAP1 is
114. Flick, J. S., and M. Johnston. 1991. GRR1 of Saccharomyces cerevisiae is
required for BAS1/BAS2- and GCN4-dependent transcription of the yeast
required for glucose repression and encodes a protein with leucine-rich
HIS4 gene. Mol. Cell. Biol. 11:3642–3651.
repeats. Mol. Biol. Cell 11:5101–5112.
89. Dlugai, S., S. Hippler, R. Wieczorke, and E. Boles. 2001. Glucose-depen-
115. Flick, J. S., and M. Johnston. 1992. Analysis of URSG-mediated glucose
dent and -independent signalling functions of the yeast glucose sensor Snf3.
repression of the GAL1 promoter of Saccharomyces cerevisiae. Genetics
FEBS Lett. 505:389–392.
130:295–304.
90. Dombek, K. M., N. Kacherovsky, and E. T. Young. 2004. The Reg1-inter-
116. Flick, K., D. Chapman-Shimshoni, D. Stuart, M. Guaderrama, and C.
acting proteins, Bmh1, Bmh2, Ssb1, and Ssb2, have roles in maintaining
Wittenberg. 1998. Regulation of cell size by glucose is exerted via repression
glucose repression in Saccharomyces cerevisiae. J. Biol. Chem. 279:39165–
of the CLN1 promoter. Mol. Cell. Biol. 18:2492–2501.
39174.
117. Flick, K. M., N. Spielewoy, T. I. Kalashnikova, M. Guaderrama, Q. Zhu,
91. Dombek, K. M., V. Voronkova, A. Raney, and E. T. Young. 1999. Functional H. C. Chang, and C. Wittenberg. 2003. Grr1-dependent inactivation of
analysis of the yeast Glc7-binding protein Reg1 identifies a protein phos- Mth1 mediates glucose-induced dissociation of Rgt1 from HXT gene pro-
phatase type 1-binding motif as essential for repression of ADH2 expres- moters. Mol. Cell. Biol. 14:3230–3241.
sion. Mol. Cell. Biol. 19:6029–6040. 118. Forsberg, H., and P. O. Ljungdahl. 2001. Sensors of extracellular nutrients
92. Dong, X., D. A. Mitchell, S. Lobo, L. Zhao, D. J. Bartels, and R. J. in Saccharomyces cerevisiae. Curr. Genet. 40:91–109.
Deschenes. 2003. Palmitoylation and plasma membrane localization of 119. Francois, J., and J. L. Parrou. 2001. Reserve carbohydrates metabolism in
Ras2p by a nonclassical trafficking pathway in Saccharomyces cerevisiae. the yeast Saccharomyces cerevisiae. FEMS Microbiol. Rev. 25:125–145.
Mol. Cell. Biol. 23:6574–6584. 120. Frederick, D. L., and K. Tatchell. 1996. The REG2 gene of Saccharomyces
93. Donovan, D. M., and N. J. Pearson. 1986. Transcriptional regulation of cerevisiae encodes a type 1 protein phosphatase-binding protein that func-
ribosomal proteins during a nutritional upshift in Saccharomyces cerevisiae. tions with Reg1p and the Snf1 protein kinase to regulate growth. Mol. Cell.
Mol. Cell. Biol. 6:2429–2435. Biol. 16:2922–2931.
94. Dubacq, C., A. Chevalier, and C. Mann. 2004. The protein kinase Snf1 is 121. Gancedo, J. M. 1998. Yeast carbon catabolite repression. Microbiol. Mol.
required for tolerance to the ribonucleotide reductase inhibitor hydroxy- Biol. Rev. 62:334–361.
urea. Mol. Cell. Biol. 24:2560–2572. 122. Ganster, R. W., W. Shen, and M. C. Schmidt. 1993. Isolation of STD1, a
95. Dubacq, C., R. Guerois, R. Courbeyrette, K. Kitagawa, and C. Mann. 2002. high-copy-number suppressor of a dominant negative mutation in the yeast
Sgt1p contributes to cyclic AMP pathway activity and physically interacts TATA-binding protein. Mol. Cell. Biol. 13:3650–3659.
with the adenylyl cyclase Cyr1p/Cdc35p in budding yeast. Eukaryot. Cell 123. Gelade, R., S. Van de Velde, P. Van Dijck, and J. M. Thevelein. 2003.
1:568–582. Multi-level response of the yeast genome to glucose. Genome Biol. 4:233.
96. Durchschlag, E., W. Reiter, G. Ammerer, and C. Schuller. 2004. Nuclear 124. Gelperin, D., J. Weigle, K. Nelson, P. Roseboom, K. Irie, K. Matsumoto,
localization destabilizes the stress-regulated transcription factor Msn2. and S. Lemmon. 1995. 14-3-3 proteins: potential roles in vesicular transport
J. Biol. Chem. 279:55425–55432. and Ras signaling in Saccharomyces cerevisiae. Proc. Natl. Acad. Sci. USA
97. Egloff, M. P., D. F. Johnson, G. Moorhead, P. T. Cohen, P. Cohen, and D. 92:11539–11543.
Barford. 1997. Structural basis for the recognition of regulatory subunits by 125. Geymonat, M., L. Wang, H. Garreau, and M. Jacquet. 1998. Ssa1p chap-
the catalytic subunit of protein phosphatase 1. EMBO J. 16:1876–1887. erone interacts with the guanine nucleotide exchange factor of ras Cdc25p
98. Eisen, A., W. E. Taylor, H. Blumberg, and E. T. Young. 1988. The yeast and controls the cAMP pathway in Saccharomyces cerevisiae. Mol. Micro-
regulatory protein ADR1 binds in a zinc-dependent manner to the up- biol. 30:855–864.
stream activating sequence of ADH2. Mol. Cell. Biol. 8:4552–4556. 126. Ghannam, G., A. Takeda, T. Camarata, M. A. Moore, A. Viale, and N. R.
99. Entian, K. D. 1980. Genetic and biochemical evidence for hexokinase PII as Yaseen. 2004. The oncogene Nup98-HOXA9 induces gene transcription in
a key enzyme involved in carbon catabolite repression in yeast. Mol. Gen. myeloid cells. J. Biol. Chem. 279:866–875.
Genet. 178:633–637. 127. Gilbert, W., and C. Guthrie. 2004. The Glc7p nuclear phosphatase pro-
100. Entian, K. D., and D. Mecke. 1982. Genetic evidence for a role of hexoki- motes mRNA export by facilitating association of Mex67p with mRNA.
nase isozyme PII in carbon catabolite repression in Saccharomyces cerevi- Mol. Cell 13:201–212.
siae. J. Biol. Chem. 257:870–874. 128. Gotta, M., and S. M. Gasser. 1996. Nuclear organization and transcrip-
101. Entian, K. D., and F. K. Zimmermann. 1980. Glycolytic enzymes and tional silencing in yeast. Experientia 52:1136–1147.
intermediates in carbon catabolite repression mutants of Saccharomyces 129. Griffioen, G., and J. M. Thevelein. 2002. Molecular mechanisms controlling
cerevisiae. Mol. Gen. Genet. 177:345–350. the localization of protein kinase A. Curr. Genet. 41:199–207.
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 277

130. Griffioen, G., P. Anghileri, E. Imre, M. D. Baroni, and H. Ruis. 2000. 156. Horak, J., J. Regelmann, and D. H. Wolf. 2002. Two distinct proteolytic
Nutritional control of nucleocytoplasmic localization of cAMP-dependent systems responsible for glucose-induced degradation of fructose-1,6-
protein kinase catalytic and regulatory subunits in Saccharomyces cerevisiae. bisphosphatase and the Gal2p transporter in the yeast Saccharomyces
J. Biol. Chem. 275:1449–1456. cerevisiae share the same protein components of the glucose signaling path-
131. Griggs, D. W., and M. Johnston. 1991. Regulated expression of the GAL4 way. J. Biol. Chem. 277:8248–8254.
activator gene in yeast provides a sensitive genetic switch for glucose re- 157. Howard, S. C., Y. V. Budovskaya, Y. W. Chang, and P. K. Herman. 2002.
pression. Proc. Natl. Acad. Sci. USA 88:8597–8601. The C-terminal domain of the largest subunit of RNA polymerase II is
132. Hall, D. D., D. D. Markwardt, F. Parviz, and W. Heideman. 1998. Regu- required for stationary phase entry and functionally interacts with the
lation of the Cln3-Cdc28 kinase by cAMP in Saccharomyces cerevisiae. Ras/PKA signaling pathway. J. Biol. Chem. 277:19488–19497.
EMBO J. 17:4370–4378. 158. Hsiung, Y. G., H. C. Chang, J. Pellequer, R. La Valle, S. Lanker, and C.
133. Han, Y., S. San-Marina, J. Liu, and M. D. Minden. 2004. Transcriptional Wittenberg. 2001. F-box protein Grr1 interacts with phosphorylated targets
activation of c-myc proto-oncogene by WT1 protein. Oncogene 23:6933– via the cationic surface of its leucine-rich repeat. Mol. Cell. Biol. 21:2506–
6941. 2520.
134. Haney, S. A., and J. R. Broach. 1994. Cdc25p, the guanine nucleotide 159. Hsu, J. Y., Z. W. Sun, X. Li, M. Reuben, K. Tatchell, D. K. Bishop, J. M.
exchange factor for the Ras proteins of Saccharomyces cerevisiae, promotes Grushcow, C. J. Brame, J. A. Caldwell, D. F. Hunt, R. Lin, M. M. Smith,
exchange by stabilizing Ras in a nucleotide-free state. J. Biol. Chem. 269: and C. D. Allis. 2000. Mitotic phosphorylation of histone H3 is governed
16541–16548. by Ip11/aurora kinase and Glc7/PP1 phosphatase in budding yeast and
135. Harashima, T., and J. Heitman. 2002. The G␣ protein Gpa2 controls yeast nematodes. Cell 102:279–291.
differentiation by interacting with kelch repeat proteins that mimic G␤ 160. Hu, Z., J. O. Nehlin, H. Ronne, and C. A. Michels. 1995. MIG1-dependent
and MIG1-independent glucose regulation of MAL gene expression in

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


subunits. Mol. Cell 10:163–173.
136. Hardie, D. G., D. Carling, and M. Carlson. 1998. The AMP-activated/SNF1 Saccharomyces cerevisiae. Curr. Genet. 28:258–266.
protein kinase subfamily: metabolic sensors of the eukaryotic cell? Annu. 161. Hu, Z., Y. Yue, H. Jiang, B. Zhang, P. W. Sherwood, and C. A. Michels.
Rev. Biochem. 67:821–855. 2000. Analysis of the mechanism by which glucose inhibits maltose induc-
137. Harkness, T. A., K. A. Shea, C. Legrand, M. Brahmania, and G. F. Davies. tion of MAL gene expression in Saccharomyces. Genetics 154:121–132.
2004. A functional analysis reveals dependence on the anaphase-promoting 162. Huang, D., K. T. Chun, M. G. Goebl, and P. J. Roach. 1996. Genetic
complex for prolonged life span in yeast. Genetics 168:759–774. interactions between REG1/HEX2 and GLC7, the gene encoding the pro-
138. Hartwell, L. H. 1974. Saccharomyces cerevisiae cell cycle. Bacteriol. Rev. tein phosphatase type 1 catalytic subunit in Saccharomyces cerevisiae. Ge-
38:164–198. netics 143:119–127.
139. Hartwell, L. H., R. K. Mortimer, J. Culotti, and M. Culotti. 1973. Genetic 163. Hubbard, E. J., R. Jiang, and M. Carlson. 1994. Dosage-dependent mod-
control of the cell division cycle in yeast. V. Genetic analysis of cdc mutants. ulation of glucose repression by MSN3 (STD1) in Saccharomyces cerevisiae.
Genetics 74:267–286. Mol. Cell. Biol. 14:1972–1978.
140. Haurie, V., F. Sagliocco, and H. Boucherie. 2004. Dissecting regulatory 164. Hubbard, E. J., X. L. Yang, and M. Carlson. 1992. Relationship of the
networks by means of two-dimensional gel electrophoresis: application to cAMP-dependent protein kinase pathway to the SNF1 protein kinase and
the study of the diauxic shift in the yeast Saccharomyces cerevisiae. Pro- invertase expression in Saccharomyces cerevisiae. Genetics 130:71–80.
teomics 4:364–373. 165. Hubler, L., J. Bradshaw-Rouse, and W. Heideman. 1993. Connections be-
tween the Ras-cyclic AMP pathway and G1 cyclin expression in the budding
141. Haurie, V., M. Perrot, T. Mini, P. Jeno, F. Sagliocco, and H. Boucherie.
yeast Saccharomyces cerevisiae. Mol. Cell. Biol. 13:6274–6282.
2001. The transcriptional activator Cat8p provides a major contribution
166. Hung, G. C., C. R. Brown, A. B. Wolfe, J. Liu, and H. L. Chiang. 2004.
to the reprogramming of carbon metabolism during the diauxic shift in
Degradation of the gluconeogenic enzymes fructose-1,6-bisphosphatase
Saccharomyces cerevisiae. J. Biol. Chem. 276:76–85.
and malate dehydrogenase is mediated by distinct proteolytic pathways and
142. Hazbun, T. R., and S. Fields. 2002. A genome-wide screen for site-specific
signaling events. J. Biol. Chem. 279:49138–49150.
DNA-binding proteins. Mol. Cell Proteomics 1:538–543.
167. Hutchison, N., and H. Weintraub. 1985. Localization of DNAase I-sensitive
143. Hedbacker, K., R. Townley, and M. Carlson. 2004. Cyclic AMP-dependent
sequences to specific regions of interphase nuclei. Cell 43:471–482.
protein kinase regulates the subcellular localization of Snf1-Sip1 protein
168. Ichimura, T., H. Kubota, T. Goma, N. Mizushima, Y. Ohsumi, M. Iwago, K.
kinase. Mol. Cell. Biol. 24:1836–1843.
Kakiuchi, H. U. Shekhar, T. Shinkawa, M. Taoka, T. Ito, and T. Isobe.
144. Hedbacker, K., S. P. Hong, and M. Carlson. 2004. Pak1 protein kinase 2004. Transcriptomic and proteomic analysis of a 14-3-3 gene-deficient
regulates activation and nuclear localization of Snf1-Gal83 protein kinase. yeast. Biochemistry 43:6149–6158.
Mol. Cell. Biol. 24:8255–8263. 169. Isogai, Y., and R. Tjian. 2003. Targeting genes and transcription factors to
145. Hedges, D., M. Proft, and K. D. Entian. 1995. CAT8, a new zinc cluster- segregated nuclear compartments. Curr. Opin. Cell Biol. 15:296–303.
encoding gene necessary for depression of gluconeogenic enzymes in the 170. Jaquenoud, M., M. P. Gulli, K. Peter, and M. Peter. 1998. The Cdc42p
yeast Saccharomyces cerevisiae. Mol. Cell. Biol. 15:1915–1922. effector Gic2p is targeted for ubiquitin-dependent degradation by the SCF-
146. Herrero, P., C. Martinez-Campa, and F. Moreno. 1998. The hexokinase 2 Grr1 complex. EMBO J. 17:5360–5373.
protein participates in regulatory DNA-protein complexes necessary for 171. Jiang, H., I. Medintz, and C. A. Michels. 1997. Two glucose sensing/signal-
glucose repression of the SUC2 gene in Saccharomyces cerevisiae. FEBS ing pathways stimulate glucose-induced inactivation of maltose permease in
Lett. 434:71–76. Saccharomyces. Mol. Biol. Cell 8:1293–1304.
147. Herrero, P., J. Galindez, N. Ruiz, C. Martinez-Campa, and F. Moreno. 172. Jiang, H., I. Medintz, B. Zhang, and C. A. Michels. 2000. Metabolic signals
1995. Transcription regulation of the Saccharomyces cerevisiae HXK1, trigger glucose-induced inactivation of maltose permease in Saccharomyces.
HXK2 and GLK1 genes. Yeast 11:137–144. J. Bacteriol. 182:647–654.
148. Hierro, A., J. Sun, A. S. Rusnak, J. Kim, G. Prag, S. E. Emr, and J. H. 173. Jiang, H., K. Tatchell, S. Liu, and C. A. Michels. 2000. Protein phosphatase
Hurley. 2004. Structure of the ESCRT-II endosomal trafficking complex. type-1 regulatory subunits Reg1p and Reg2p act as signal transducers in the
Nature 431:221–225. glucose-induced inactivation of maltose permease in Saccharomyces cerevisiae.
149. Hisamoto, N., D. L. Frederick, K. Sugimoto, K. Tatchell, and K. Matsu- Mol. Gen. Genet. 263:411–422.
moto. 1995. The EGP1 gene may be a positive regulator of protein phos- 174. Jiang, R., and M. Carlson. 1996. Glucose regulates protein interactions
phatase type 1 in the growth control of Saccharomyces cerevisiae. Mol. Cell. within the yeast SNF1 protein kinase complex. Genes Dev. 10:3105–3115.
Biol. 15:3767–3776. 175. Jiang, R., and M. Carlson. 1997. The Snf1 protein kinase and its activating
150. Hisamoto, N., K. Sugimoto, and K. Matsumoto. 1994. The Glc7 type 1 subunit, Snf4, interact with distinct domains of the Sip1/Sip2/Gal83 com-
protein phosphatase of Saccharomyces cerevisiae is required for cell cycle ponent in the kinase complex. Mol. Cell. Biol. 17:2099–2106.
progression in G2/M. Mol. Cell. Biol. 14:3158–3165. 176. Jiang, Y., C. Davis, and J. R. Broach. 1998. Efficient transition to growth on
151. Hixson, C. S., and E. G. Krebs. 1980. Characterization of a cyclic AMP- fermentable carbon sources in Saccharomyces cerevisiae requires signaling
binding protein from bakers’ yeast. Identification as a regulatory subunit of through the Ras pathway. EMBO J. 17:6942–6951.
cyclic AMP-dependent protein kinase. J. Biol. Chem. 255:2137–2145. 177. Johnson, K. E., S. Cameron, T. Toda, M. Wigler, and M. J. Zoller. 1987.
152. Holland, M. J., T. Yokoi, J. P. Holland, K. Myambo, and M. A. Innis. 1987. Expression in Escherichia coli of BCY1, the regulatory subunit of cyclic
The GCR1 gene encodes a positive transcriptional regulator of the enolase AMP-dependent protein kinase from Saccharomyces cerevisiae. Purification
and glyceraldehydes-3-phosphate dehydrogenase gene families in Saccharo- and characterization. J. Biol. Chem. 262:8636–8642.
myces cerevisiae. Mol. Cell. Biol. 7:813–820. 178. Johnston, M. 1999. Feasting, fasting and fermenting. Glucose sensing in
153. Holzer, H. 1976. Catabolite inactivation in yeast. Trends Biochem. Sci. yeast and other cells. Trends Genet. 15:29–33.
1:178–181. 179. Johnston, M., and J. H. Kim. 2005. Glucose as a hormone: receptor-
154. Holzer, H. 1989. Proteolytic catabolite inactivation in Saccharomyces cer- mediated glucose sensing in the yeast Saccharomyces cerevisiae. Biochem.
evisiae. Revis Biol. Celular 21:305–319. Soc. Trans. 33:247–252.
155. Hong, S. P., F. C. Leiper, A. Woods, D. Carling, and M. Carlson. 2003. 180. Johnston, M., J. S. Flick, and T. Pexton. 1994. Multiple mechanisms pro-
Activation of yeast Snf1 and mammalian AMP-activated protein kinase by vide rapid and stringent glucose repression of GAL gene expression in
upstream kinases. Proc. Natl. Acad. Sci. USA 100:8839–8843. Saccharomyces cerevisiae. Mol. Cell. Biol. 14:3834–3841.
278 SANTANGELO MICROBIOL. MOL. BIOL. REV.

181. Jorgensen, P., I. Rupes, J. R. Sharom, L. Schneper, J. R. Broach, and M. 206. Lesage, P., X. Yang, and M. Carlson. 1996. Yeast SNF1 protein kinase
Tyers. 2004. A dynamic transcriptional network communicates growth po- interacts with SIP4, a C6 zinc cluster transcriptional activator: a new role for
tential to ribosome synthesis and critical cell size. Genes Dev. 18:2491– SNF1 in the glucose response. Mol. Cell. Biol. 16:1921–1928.
2505. 207. Levine, M., and J. L. Manley. 1989. Transcriptional repression of eukaryotic
182. Kaniak, A., Z. Xue, D. Macool, J. H. Kim, and M. Johnston. 2004. Regu- promoters. Cell 59:405–408.
latory network connecting two glucose signal transduction pathways in 208. Li, B., C. R. Nierras, and J. R. Warner. 1999. Transcriptional elements
Saccharomyces cerevisiae. Eukaryot. Cell 3:221–231. involved in the repression of ribosomal protein synthesis. Mol. Cell. Biol.
183. Kasper, L. H., P. K. Brindle, C. A. Schnabel, C. E. Pritchard, M. L. Cleary, 19:5393–5404.
and J. M. van Deursen. 1999. CREB binding protein interacts with nucleo- 209. Li, F. N., and M. Johnston. 1997. Grr1 of Saccharomyces cerevisiae is
porin-specific FG repeats that activate transcription and mediate NUP98- connected to the ubiquitin proteolysis machinery through Skp1: coupling
HOXA9 oncogenicity. Mol. Cell. Biol. 19:764–776. glucose sensing to gene expression and the cell cycle. EMBO J. 16:5629–
184. Kido, M., F. Shima, T. Satoh, T. Asato, K. Kariya, and T. Kataoka. 2002. 5638.
Critical function of the Ras-associating domain as a primary Ras-binding 210. Li, Y., S. Bjorklund, Y. W. Jiang, Y. J. Kim, W. S. Lane, D. J. Stillman, and
site for regulation of Saccharomyces cerevisiae adenylyl cyclase. J. Biol. R. D. Kornberg. 1995. Yeast global transcriptional regulators Sin4 and Rgr1
Chem. 277:3117–3123. are components of mediator complex/RNA polymerase II holoenzyme.
185. Kief, D. R., and J. R. Warner. 1981. Coordinate control of syntheses of Proc. Natl. Acad. Sci. USA 92:10864–10868.
ribosomal ribonucleic acid and ribosomal proteins during nutritional 211. Lieb, J. D., X. Liu, D. Botstein, and P. O. Brown. 2001. Promoter-specific
shift-up in Saccharomyces cerevisiae. Mol. Cell. Biol. 1:1007–1015. binding of Rap1 revealed by genome-wide maps of protein-DNA associa-
186. Kim, J. H., J. Polish, and M. Johnston. 2003. Specificity and regulation of tion. Nat. Genet. 28:327–334.
212. Lin, S. J., M. Kaeberlein, A. A. Andalis, L. A. Sturtz, P. A. Defossez, V. C.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


DNA binding in the yeast glucose transporter gene repressor Rgt1. Mol.
Cell. Biol. 23:5208–5216. Culotta, G. R. Fink, and L. Guarente. 2002. Calorie restriction extends
187. Kishi, T., and F. Yamao. 1998. An essential function of Grr1 for the Saccharomyces cerevisiae lifespan by increasing respiration. Nature 418:
degradation of Cln2 is to act as a binding core that links Cln2 to Skp1. 344–348.
J. Cell Sci. 111:3655–3661. 213. Lin, S. S., J. K. Manchester, and J. L. Gordon. 2003. Sip2, an N-myristoylated
188. Klein, C., and K. Struhl. 1994. Protein kinase A mediates growth-regulated beta subunit of Snf1 kinase, regulates aging in Saccharomyces cerevisiae by
expression of yeast ribosomal protein genes by modulating RAP1 transcrip- affecting cellular histone kinase activity, recombination at rDNA loci, and
tional activity. Mol. Cell. Biol. 14:1920–1928. silencing. J. Biol. Chem. 278:13390–13397.
189. Klein, C. J., L. Olsson, and J. Nielsen. 1998. Glucose control in Saccharo- 214. Lodi, T., C. Donnini, and I. Ferrero. 1991. Catabolite repression by galac-
myces cerevisiae: the role of Mig1 in metabolic functions. Microbiology tose in overexpressed GAL4 strains of Saccharomyces cerevisiae. J. Gen.
144:13–24. Microbiol. 137:1039–1044.
190. Kraakman, L., K. Lemaire, P. Ma, A. W. Teunissen, M. C. Donaton, P. Van 215. Loeb, J. D., T. A. Kerentseva, T. Pan, M. Sepulveda-Becerra, and H. Liu.
Dijck, J. Winderickx, J. H. de Winde, and J. M. Thevelein. 1999. A Sac- 1999. Saccharomyces cerevisiae G1 cyclins are differentially involved in in-
charomyces cerevisiae G-protein coupled receptor, Gpr1, is specifically re- vasive and pseudohyphal growth independent of the filamentation mitogen-
quired for glucose activation of the cAMP pathway during the transition to activated protein kinase pathway. Genetics 153:1535–1546.
growth on glucose. Mol. Microbiol. 32:1002–1012. 216. Longo, V. D. 2003. The Ras and Sch9 pathways regulate stress resistance
191. Kraakman, L. S., J. Winderickx, J. M. Thevelein, and J. H. de Winde. 1999. and longevity. Exp. Gerontol. 38:807–811.
Structure-function analysis of yeast hexokinase: structural requirements for 217. Lorenz, M. C., X. Pan, T. Harashima, M. E. Cardenas, Y. Xue, J. P. Hirsch,
triggering cAMP signalling and catabolite repression. Biochem. J. 343:159– and J. Heitman. 2000. The G protein-coupled receptor Gpr1 is a nutrient
168. sensor that regulates pseudohyphal differentiation in Saccharomyces cerevisiae.
Genetics 154:609–622.
192. Kratzer, S., and H. J. Schuller. 1997. Transcriptional control of the yeast
218. Ludin, K., R. Jiang, and M. Carlson. 1998. Glucose-regulated interaction of
acetyl-CoA synthase gene, ACS1, by the positive regulators CAT8 and
a regulatory subunit of protein phosphatase 1 with the Snf1 protein kinase
ADR1 and the pleiotropic repressor UME6. Mol. Microbiol. 26:631–641.
in Saccharomyces cerevisiae. Proc. Natl. Acad. Sci. USA 95:6245–6250.
193. Kriegel, T. M., J. Rush, A. B. Vojtek, D. Clifton, and D. G. Fraenkel. 1994.
219. Lundin, M., J. O. Nehlin, and H. Ronne. 1994. Importance of a flanking
In vivo phosphorylation site of hexokinase 2 in Saccharomyces cerevisiae.
AT-rich region in target site recognition by the GC box-binding zinc finger
Biochemistry 33:149–152.
protein MIG1. Mol. Cell. Biol. 14:1979–1985.
194. Kuchin, S., and M. Carlson. 1998. Functional relationships of Srb10-Srb11
220. Lutfiyya, L. L., and M. Johnston. 1996. Two zinc-finger-containing repres-
kinase, carboxy-terminal domain kinase CTDK-I, and transcriptional core-
sors are responsible for glucose repression of SUC2 expression. Mol. Cell.
pressor Ssn6-Tup1. Mol. Cell. Biol. 18:1163–1171.
Biol. 16:4790–4797.
195. Kuchin, S., I. Treich, and M. Carlson. 2000. A regulatory shortcut between 221. Lutfiyya, L. L., V. R. Iyer, J. DeRisi, M. J. DeVit, P. O. Brown, and M.
the Snf1 protein kinase and RNA polymerase II holoenzyme. Proc. Natl. Johnston. 1998. Characterization of three related glucose repressors and
Acad. Sci. USA 97:7916–7920. genes they regulate in Saccharomyces cerevisiae. Genetics 150:1377–1391.
196. Kuchin, S., V. K. Vyas, and M. Carlson. 2002. Snf1 protein kinase and the 222. Ma, H., and D. Botstein. 1986. Effects of null mutations in the hexokinase
repressors Nrg1 and Nrg2 regulate FLO11, haploid invasive growth, and genes of Saccharomyces cerevisiae on catabolite repression. Mol. Cell. Biol.
diploid pseudohyphal differentiation. Mol. Cell. Biol. 22:3994–4000. 6:4046–4052.
197. Kuchin, S., V. K. Vyas, and M. Carlson. 2003. Role of the yeast Snf1 protein 223. MacKelvie, S. H., P. D. Andrews, and M. J. Stark. 1995. The Saccharomyces
kinase in invasive growth. Biochem. Soc. Trans. 31:175–177. cerevisiae gene SDS22 encodes a potential regulator of the mitotic function
198. Kuchin, S., V. K. Vyas, E. Kanter, S. P. Hong, and M. Carlson. 2003. Std1p of yeast type 1 protein phosphatase. Mol. Cell. Biol. 15:3777–3785.
(Msn3p) positively regulates the Snf1 kinase in Saccharomyces cerevisiae. 224. Maheswaran, S., S. Park, A. Bernard, J. F. Morris, F. J. Rauscher III, D. E.
Genetics 163:507–514. Hill, and D. A. Haber. 1993. Physical and functional interaction between
199. Kuroda, Y., N. Suzuki, and T. Kataoka. 1993. The effect of posttranslational WT1 and p53 proteins. Proc. Natl. Acad. Sci. USA 90:5100–5104.
modifications on the interactions of Ras2 with adenylyl cyclase. Science 225. Maniatis, T., and R. Reed. 2002. An extensive network of coupling among
259:683–686. gene expression machines. Nature 416:499–506.
200. Laemmli, U. K., and R. Tjian. 1996. A nuclear traffic jam: unraveling 226. Marion, R. M., A. Regev, E. Segal, Y. Barash, D. Koller, N. Friedman, and
multicomponent machines and compartments. Curr. Opin. Cell Biol. 8:299– E. K. O’Shea. 2004. Sfp1 is a stress- and nutrient-sensitive regulator of
302. ribosomal protein gene expression. Proc. Natl. Acad. Sci. USA 101:14315–
201. Lafuente, M. J., C. Gancedo, J. C. Jauniaux, and J. M. Gancedo. 2000. 14322.
Mth1 receives the signal given by the glucose sensors Snf3 and Rgt2 in 227. Marshall-Carlson, L., J. L. Celenza, B. C. Laurent, and M. Carlson. 1990.
Saccharomyces cerevisiae. Mol. Microbiol. 35:161–172. Mutational analysis of the SNF3 glucose transporter of Saccharomyces
202. Lakshmanan, J., A. L. Mosley, and S. Ozcan. 2003. Repression of tran- cerevisiae. Mol. Cell. Biol. 10:1105–1115.
scription by Rgt1 in the absence of glucose requires Std1 and Mth1. Curr. 228. Marshall-Carlson, L., L. Neigeborn, D. Coons, L. Bisson, and M. Carlson.
Genet. 44:19–25. 1991. Dominant and recessive suppressors that restore glucose transport in
203. Lee, M., S. Chatterjee, and K. Struhl. 2000. Genetic analysis of the role of a yeast Snf3 mutant. Genetics 128:505–512.
Pol II holoenzyme components in repression by the Cyc8-Tup1 corepressor 229. Matsumoto, K., I. Uno, Y. Oshima, and T. Ishikawa. 1982. Isolation and
in yeast. Genetics 155:1535–1542. characterization of yeast mutants deficient in adenylate cyclase and cAMP-
204. Leech, A., N. Nath, R. R. McCartney, and M. C. Schmidt. 2003. Isolation of dependent protein kinase. Proc. Natl. Acad. Sci. USA 79:2355–2359.
mutations in the catalytic domain of the Snf1 kinase that render its activity 230. Matsumoto, K., T. Yoshimatsu, and Y. Oshima. 1983. Recessive mutations
independent of the Snf4 subunit. Eukaryot. Cell 2:265–273. conferring resistance to carbon catabolite repression of galactokinase syn-
205. Lemaire, K., S. Van de Velde, P. Van Dijck, and J. M. Thevelein. 2004. thesis in Saccharomyces cerevisiae. J. Bacteriol. 153:1405–1414.
Glucose and sucrose act as agonist and mannose as antagonist ligands of the 231. Mayordomo, I., and P. Sanz. 2001. Human pancreatic glucokinase (GlkB)
G protein-coupled receptor Gpr1 in the yeast Saccharomyces cerevisiae. complements the glucose signalling defect of Saccharomyces cerevisiae hxk2
Mol. Cell 16:293–299. mutants. Yeast 18:1309–1316.
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 279

232. Mayordomo, I., J. Regelmann, J. Horak, and P. Sanz. 2003. Saccharomyces 257. Nehlin, J. O., and H. Ronne. 1990. Yeast MIG1 repressor is related to the
cerevisiae 14-3-3 proteins Bmh1 and Bmh2 participate in the process of mammalian early growth response and Wilms’ tumour finger proteins.
catabolite inactivation of maltose permease. FEBS Lett. 544:160–164. EMBO J. 9:2891–2898.
233. Mbonyi, K., L. van Aelst, J. C. Arguelles, A. W. Jans, and J. M. Thevelein. 258. Nehlin, J. O., M. Carlberg, and H. Ronne. 1991. Control of yeast GAL
1990. Glucose-induced hyperaccumulation of cyclic AMP and defective genes by MIG1 repressor: a transcriptional cascade in the glucose response.
glucose repression in yeast strains with reduced activity of cyclic AMP- EMBO J. 10:3373–3377.
dependent protein kinase. Mol. Cell. Biol. 10:4518–4523. 259. Neigeborn, L., and M. Carlson. 1984. Genes affecting the regulation of
234. McEwen, B. S., V. G. Allfrey, and A. E. Mirsky. 1963. Studies on energy- SUC2 gene expression by glucose repression in Saccharomyces cerevisiae.
yielding reactions in thymus nuclei. II. Pathways of aerobic carbohydrate Genetics 108:845–858.
catabolism. J. Biol. Chem. 238:2571–2578. 260. Neigeborn, L., and M. Carlson. 1987. Mutations causing constitutive inver-
235. McKay, L. M., B. Carpenter, and S. G. Roberts. 1999. Regulation of the tase synthesis in yeast: genetic interactions with snf mutations. Genetics
Wilms’ tumour suppressor protein transcriptional activation domain. On- 115:247–253.
cogene 18:6546–6554. 261. Neigeborn, L., K. Rubin, and M. Carlson. 1986. Suppressors of SNF2
236. Menon, B. B., N. J. Sarma, S. Pasula, S. J. Deminoff, K. A. Willis, K. E. mutations restore invertase derepression and cause temperature-sensitive
Barbara, B. Andrews, and G. M. Santangelo. 2005. Reverse recruitment: lethality in yeast. Genetics 112:741–753.
the Nup84 nuclear pore subcomplex mediates Rap1/Gcr1/Gcr2 transcrip- 262. Newcomb, L. L., D. D. Hall, and W. Heideman. 2002. AZF1 is a glucose-
tional activation. Proc. Natl. Acad. Sci. USA 102:5749–5754. dependent positive regulator of CLN3 transcription in Saccharomyces cerevi-
siae. Mol. Cell. Biol. 22:1607–1614.
237. Mintzer, K. A., and J. Field. 1999. The SH3 domain of the S. cerevisiae
263. Newcomb, L. L., J. A. Diderich, M. G. Slattery, and W. Heideman. 2003.
Cdc25p binds adenylyl cyclase and facilitates Ras regulation of cAMP
Glucose regulation of Saccharomyces cerevisiae cell cycle genes. Eukaryot.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


signalling. Cell Signal. 11:127–135.
Cell 2:143–149.
238. Misteli, T. 2004. Spatial positioning; a new dimension in genome function.
264. Nichols, K. E., G. G. Re, Y. X. Yan, A. J. Garvin, and D. A. Haber. 1995.
Cell 119:153–156.
WT1 induces expression of insulin-like growth factor 2 in Wilms’ tumor
239. Misteli, T. 2005. Concepts in nuclear architecture. Bioassays 27:477–487. cells. Cancer Res. 55:4540–4543.
240. Misteli, T., and D. L. Spector. 1998. The cellular organization of gene 265. Niederacher, D., and K. D. Entian. 1991. Characterization of Hex2 protein,
expression. Curr. Opin. Cell Biol. 10:323–331. a negative regulatory element necessary for glucose repression in yeast.
241. Mitts, M. R., J. Bradshaw-Rouse, and W. Heideman. 1991. Interactions Eur. J. Biochem. 200:311–319.
between adenylate cyclase and the yeast GTPase-activating protein IRA1. 266. Nikawa, J., P. Sass, and M. Wigler. 1987. Cloning and characterization
Mol. Cell. Biol. 11:4591–4598. of the low-affinity cyclic AMP phosphodiesterase gene of Saccharomyces
242. Mizuno, T., and S. Harashima. 2000. Activation of basal transcription by a cerevisiae. Mol. Cell. Biol. 7:3629–3636.
mutation in SIN4, a yeast global repressor, occurs through a mechanism 267. Nishida, Y., F. Shima, H. Sen, Y. Tanada, C. Yanagihara, Y. Yamawaki-
different from activator-mediated transcriptional enhancement. Mol. Gen. Kataoka, K. Kariya, and T. Kataoka. 1998. Coiled-coil interaction of N-
Genet. 263:48–59. terminal 36 residues of cyclase-associated protein with adenylyl cyclase is
243. Monod, J. 1942. Recherches sur la croissance des cultures bacteriennes. sufficient for its function in Saccharomyces cerevisiae ras pathway. J. Biol.
Ph.D. thesis. University of Paris, Paris, France. Chem. 273:28019–28024.
244. Moreno, F., and P. Herrero. 2002. The hexokinase 2-dependent glucose 268. Oren, M. 2003. Decision making by p53: life, death and cancer. Cell Death
signal transduction pathway of Saccharomyces cerevisiae. FEMS Microbiol. Differ. 10:431–442.
Rev. 26:83–90. 269. Ostling, J., and H. Ronne. 1998. Negative control of the Mig1p repressor by
245. Moreno, F., D. Ahuatzi, A. Riera, C. A. Palomino, and P. Herrero. 2005. Snf1p-dependent phosphorylation in the absence of glucose. Eur. J. Bio-
Glucose sensing through the Hxk2-dependent signalling pathway. Biochem. chem. 252:162–168.
Soc. Trans. 33:265–268. 270. Ostling, J., M. Carlberg, and H. Ronne. 1996. Functional domains in the
246. Moreno-Herrero, F., P. Herrero, J. Colchero, A. M. Baro, and F. Moreno. Mig1 repressor. Mol. Cell. Biol. 16:753–761.
1999. Analysis by atomic force microscopy of Med8 binding to cis-acting 271. Ozcan, S., and M. Johnston. 1995. Three different regulatory mechanisms
regulatory elements of the SUC2 and HXK2 genes of Saccharomyces cerevisiae. enable yeast hexose transporter (HXT) genes to be induced by different
FEBS Lett. 459:427–432. levels of glucose. Mol. Cell. Biol. 15:1564–1572.
247. Moriya, H., and M. Johnston. 2004. Glucose sensing and signaling in 272. Ozcan, S., and M. Johnston. 1999. Function and regulation of yeast hexose
Saccharomyces cerevisiae through the Rgt2 glucose sensor and casein kinase transporters. Microbiol. Mol. Biol. Rev. 63:554–569.
I. Proc. Natl. Acad. Sci. USA 101:1572–1577. 273. Ozcan, S., J. Dover, A. G. Rosenwald, S. Wolfi, and M. Johnston. 1996. Two
248. Morris, J. F., S. L. Madden, O. E. Tournay, D. M. Cook, V. P. Sukhatme, glucose transporters in Saccharomyces cerevisiae are glucose sensors that
and F. J. Rauscher III. 1991. Characterization of the zinc finger protein generate a signal for induction of gene expression. Proc. Natl. Acad. Sci.
encoded by the WT1 Wilms’ tumor locus. Oncogene 6:2339–2348. USA 93:12428–12432.
249. Mosch, H. U., E. Kubler, S. Krappmann, G. R. Fink, and G. H. Braus. 1999. 274. Ozcan, S., J. Dover, and M. Johnston. 1998. Glucose sensing and signaling
Crosstalk between the Ras2p-controlled mitogen-activated protein kinase by two glucose receptors in the yeast Saccharomyces cerevisiae. EMBO J.
and cAMP pathways during invasive growth of Saccharomyces cerevisiae. 17:2566–2573.
Mol. Biol. Cell 10:1325–1335. 275. Ozcan, S., T. Leong, and M. Johnston. 1996. Rgt1p of Saccharomyces
cerevisiae, a key regulator of glucose-induced genes, is both an activator and
250. Mosley, A. L., J. Lakshmanan, B. K. Aryal, and S. Ozcan. 2003. Glucose-
a repressor of transcription. Mol. Cell. Biol. 16:6419–6426.
mediated phosphorylation converts the transcription factor Rgt1 from a
276. Paalman, J. W., R. Verwaal, S. H. Slofstra, A. J. Verkleij, J. Boonstra, and
repressor to an activator. J. Biol. Chem. 278:10322–10327.
C. T. Verrips. 2003. Trehalose and glycogen accumulation is related to the
251. Mukherjee, S., M. F. Berger, G. Jona, X. S. Wang, D. Muzzey, M. Snyder, duration of the G1 phase of Saccharomyces cerevisiae. FEMS Yeast Res.
R. A. Young, and M. L. Bulyk. 2004. Rapid analysis of the DNA-binding 3:261–268.
specificities of transcription factors with DNA microarrays. Nat. Genet. 277. Pan, X., and J. Heitman. 1999. Cyclic AMP-dependent protein kinase
36:1331–1339. regulates pseudohyphal differentiation in Saccharomyces cerevisiae. Mol.
252. Nakafuku, M., T. Obara, K. Kaibuchi, I. Miyajima, A. Miyajima, H. Itoh, Cell. Biol. 19:4874–4887.
S. Nakamura, K. Arai, K. Matsumoto, and Y. Kaziro. 1988. Isolation of a 278. Papamichos-Chronakis, M., T. Gligoris, and D. Tzamarias. 2004. The Snf1
second yeast Saccharomyces cerevisiae gene (GPA2) coding for guanine kinase controls glucose repression in yeast by modulating interactions be-
nucleotide-binding regulatory protein: studies on its structure and possible tween the Mig1 repressor and the Cyc8-Tup1 co-repressor. EMBO Rep.
functions. Proc. Natl. Acad. Sci. USA 85:1374–1378. 5:368–372.
253. Nash, P., X. Tang, S. Orlicky, Q. Chen, F. B. Gertler, M. D. Mendenhall, F. 279. Papamichos-Chronakis, M., T. Petrakis, E. Ktistaki, I. Topalidou, and D.
Sicheri, T. Pawson, and M. Tyers. 2001. Multisite phosphorylation of a Tzamarias. 2002. Cti6, a PHD domain protein, bridges the Cyc8-Tup1
CDK inhibitor sets a threshold for the onset of DNA replication. Nature corepressor and the SAGA coactivator to overcome repression at GAL1.
414:514–521. Mol. Cell 9:1297–1305.
254. Nath, N., R. R. McCartney, and M. C. Schmidt. 2002. Purification and 280. Parada, L. A., S. Sotiriou, and T. Misteli. 2004. Spatial genome organiza-
characterization of Snf1 kinase complexes containing a defined ␤ subunit tion. Exp. Cell Res. 296:64–70.
composition. J. Biol. Chem. 277:50403–50408. 281. Parada, L., and T. Misteli. 2002. Chromosome positioning in the interphase
255. Nath, N., R. R. McCartney, and M. C. Schmidt. 2003. Yeast Pak1 kinase nucleus. Trends Cell Biol. 12:425–432.
associates with and activates Snf1. Mol. Cell. Biol. 23:3909–3917. 282. Park, S. H., S. S. Koh, J. H. Chun, H. J. Hwang, and H. S. Kang. 1999. Nrg1
256. Nedea, E., X. He, M. Kim, J. Pootoolal, G. Zhong, V. Canadien, T. Hughes, is a transcriptional repressor for glucose repression of STA1 gene expres-
S. Buratowski, C. L. Moore, and J. Greenblatt. 2003. Organization and sion in Saccharomyces cerevisiae. Mol. Cell. Biol. 19:2044–2050.
function of APT, a subcomplex of the yeast cleavage and polyadenylation 283. Parraga, G., S. J. Horvath, A. Eisen, W. E. Taylor, L. Hood, E. T. Young,
factor involved in the formation of mRNA and small nucleolar RNA 3⬘- and R. E. Klevit. 1988. Zinc-dependent structure of a single-finger domain
ends. J. Biol. Chem. 278:33000–33010. of yeast ADR1. Science 241:1489–1492.
280 SANTANGELO MICROBIOL. MOL. BIOL. REV.

284. Parrou, J. L., B. Enjalbert, and J. Francois. 1999. STRE- and cAMP- 309. Robertson, L. S., H. C. Causton, R. A. Young, and G. R. Fink. 2000. The
independent transcriptional induction of Saccharomyces cerevisiae GSY2 yeast A kinases differentially regulate iron uptake and respiratory function.
encoding glycogen synthase during diauxic growth on glucose. Yeast 15: Proc. Natl. Acad. Sci. USA 97:5984–5988.
1471–1484. 310. Robinson, L. C., E. J. A. Hubbard, P. R. Graves, A. A. DePaoli-Roach, P. J.
285. Parviz, F., and W. Heideman. 1998. Growth-independent regulation of Roach, C. Kung, D. W. Haas, C. H. Hagedorn, M. Goebl, M. R. Culbertson,
CLN3 mRNA levels by nutrients in Saccharomyces cerevisiae. J. Bacteriol. and M. Carlson. 1992. Yeast casein kinase I homologues: an essential gene
180:225–230. pair. Proc. Natl. Acad. Sci. USA 89:28–32.
286. Parviz, F., D. D. Hall, D. D. Markwardt, and W. Heideman. 1998. Tran- 311. Rodriguez, A., T. De La Cera, P. Herrero, and F. Moreno. 2001. The
scriptional regulation of CLN3 expression by glucose in Saccharomyces hexokinase 2 protein regulates the expression of GLK1, HXK1 and HXK2
cerevisiae. J. Bacteriol. 180:4508–4515. genes of Saccharomyces cerevisiae. Biochem. J. 355:625–631.
287. Peck, J. W., E. T. Bowden, and P. D. Burbelo. 2004. Structure and function 312. Rodriguez, C., P. Sanz, and C. Gancedo. 2003. New mutations of Saccharo-
of human Vps20 and Snf7 proteins. Biochem. J. 377:693–700. myces cerevisiae that partially relieve both glucose and galactose repression
288. Peggie, M. W., S. H. MacKelvie, A. Bloecher, E. V. Knatko, K. Tatchell, and activate the protein kinase Snf1. FEMS Yeast Res. 3:77–84.
M. J. Stark. 2002. Essential functions of Sds22p in chromosome localization 313. Rolland, F., J. H. de Winde, K. Lemaire, E. Boles, J. M. Thevelein, and J.
of PP1. J. Cell Sci. 115:195–206. Winderickx. 2000. Glucose-induced cAMP signalling in yeast requires both
289. Polish, J. A., J. H. Kim, and M. Johnston. 2005. How the Rgt1 transcription a G-protein coupled receptor system for extracellular glucose detection and
factor of Saccharomyces cerevisiae is regulated by glucose. Genetics 169: a separable hexose kinase-dependent sensing process. Mol. Microbiol. 38:
348–358.
583–594.
314. Rolland, F., J. Winderickx, and J. M. Thevelein. 2001. Glucose-sensing
290. Pombo, A., P. Cuello, W. Schul, J. B. Yoon, R. G. Roeder, P. R. Cook, and
mechanisms in eukaryotic cells. Trends Biochem. Sci. 26:310–317.

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


S. Murphy. 1998. Regional and temporal specialization in the nucleus: a
315. Rolland, F., J. Winderickx, and J. M. Thevelein. 2002. Glucose-sensing and
transcriptionally-active nuclear domain rich in PTF, Oct1 and PIKA anti-
-signalling mechanisms in yeast. FEMS Yeast Res. 2:183–201.
gens associates with specific chromosomes early in the cell cycle. EMBO J.
316. Rolland, F., V. Wanke, L. Cauwenberg, P. Ma, E. Boles, M. Vanoni, J. H. de
17:1768–1778.
Winde, J. M. Thevelein, and J. Winderickx. 2001. The role of hexose
291. Powers, S., T. Kataoka, O. Fasano, M. Goldfarb, J. Strathern, J. Broach, transport and phosphorylation in cAMP signalling in the yeast Saccharo-
and M. Wigler. 1984. Genes in S. cerevisiae encoding proteins with domains myces cerevisiae. FEMS Yeast Res. 1:33–45.
homologous to the mammalian ras proteins. Cell 36:607–612. 317. Rose, M., W. Albig, and K. D. Entian. 1991. Glucose repression in Saccharo-
292. Powers, T. 2004. Ribosome biogenesis: giant steps for a giant problem. Cell myces cerevisiae is directly associated with hexose phosphorylation by hexo-
119:901–902. kinases PI and PII. Eur. J. Biochem. 199:511–518.
293. Presgraves, D. C., L. Balagopalan, S. M. Abmayr, and H. A. Orr. 2003. 318. Roth, A. F., Y. Feng, L. Chen, and N. G. Davis. 2002. The yeast DHHC
Adaptive evolution drives divergence of a hybrid inviability gene between cysteine-rich domain protein Akr1p is a palmitoyl transferase. J. Cell Biol.
two species of Drosophila. Nature 423:715–719. 159:23–28.
294. Prieto, S., B. J. de la Cruz, and I. E. Scheffler. 2000. Glucose-regulated 319. Roth, S., J. Kumme, and H. J. Schuller. 2004. Transcriptional activators
turnover of mRNA and the influence of poly(A) tail length on half-life. Cat8 and Sip4 discriminate between sequence variants of the carbon
J. Biol. Chem. 275:14155–14166. source-responsive promoter element in the yeast Saccharomyces cerevisiae.
295. Ptashne, M. 2003. Regulated recruitment and cooperativity in the design of Curr. Genet. 45:121–128.
biological regulatory systems. Phil. Trans. R. Soc. London 361:1223–1234. 320. Rudra, D., Y. Zhao, and J. R. Warner. 2005. Central role of Ifh1p-Fhl1p
296. Ptashne, M., and A. Gann. 1997. Transcriptional activation by recruitment. interaction in the synthesis of yeast ribosomal proteins. EMBO J. 24:533–542.
Nature 386:569–577. 321. Russell, M., J. Bradshaw-Rouse, D. Markwardt, and W. Heideman. 1993.
297. Purnapatre, K., S. Piccirillo, B. L. Schneider, and S. M. Honigberg. 2002. Changes in gene expression in the Ras/adenylate cyclase system of Saccharo-
The CLN3/SWI6/CLN2 pathway and SNF1 act sequentially to regulate myces cerevisiae: correlation with cAMP levels and growth arrest. Mol. Cell.
meiotic initiation in Saccharomyces cerevisiae. Genes Cells 7:675–691. Biol. 4:757–765.
298. Radonjic, M., J.-C. Andrau, P. Lijnzaad, P. Kemmeren, T. T. J. P. Kock- 322. Saier, M. H., Jr. 1996. Cyclic AMP-independent catabolite repression in
elkorn, D. van Leenen, N. L. van Berkum, and F. C. P. Holstege. 2005. bacteria. FEMS Microbiol. Lett. 138:97–103.
Genome-wide analyses reveal RNA polymerase II located upstream of 323. Sakamoto, Y., M. Yoshida, K. Semba, and T. Hunter. 1997. Inhibition of the
genes poised for rapid response upon S. cerevisiae stationary phase exit. DNA-binding and transcriptional repression activity of the Wilms’ tumor
Mol. Cell 18:171–183. gene product, WT1, by cAMP-dependent protein kinase-mediated phos-
299. Rahner, A., A. Scholer, E. Martens, G. Gollwitzer, and H. J. Schuller. 1996. phorylation of Ser-365 and Ser-393 in the zinc finger domain. Oncogene
Dual influence of the yeast Cat1p (Snf1p) protein kinase on carbon source- 15:2001–2012.
dependent transcriptional activation of gluconeogenic genes by the regula- 324. Santangelo, G. M., and J. Tornow. 1990. Efficient transcription of the
tory gene. CAT8. Nucleic Acids Res. 24:2331–2337. glycolytic gene ADH1 and three translational component genes requires the
300. Rahner, A., M. Hiesinger, and H. J. Schuller. 1999. Deregulation of glu- GCR1 product, which can act through TUF/GRF/RAP binding sites. Mol.
coneogenic structural genes by variants of the transcriptional activator Cell. Biol. 10:859–862.
Cat8p of the yeast Saccharomyces cerevisiae. Mol. Microbiol. 34:146–156. 325. Sanz, P., G. R. Alms, T. A. Haystead, and M. Carlson. 2000. Regulatory
301. Ralph, S. A., C. Scheidig-Benatar, and A. Scherf. 2005. Antigenic variation interactions between the Reg1-Glc7 protein phosphatase and the Snf1
protein kinase. Mol. Cell. Biol. 20:1321–1328.
in Plasmodium falciparum is associated with movement of var loci between
subnuclear locations. Proc. Natl. Acad. Sci. USA 102:5414–5419. 326. Sanz, P., K. Ludin, and M. Carlson. 2000. Sip5 interacts with both the
Reg1/Glc7 protein phosphatase and the Snf1 protein kinase of Saccharo-
302. Ramaswamy, N. T., L. Li, M. Khalil, and J. F. Cannon. 1998. Regulation of
myces cerevisiae. Genetics 154:99–107.
yeast glycogen metabolism and sporulation by Glc7p protein phosphatase.
327. Sasaki, H., and H. Uemura. 2005. Influence of low glycolytic activities in
Genetics 149:57–72.
gcr1 and gcr2 mutants on the expression of other metabolic pathway genes
303. Randez-Gil, F., N. Bojunga, M. Proft, and K. D. Entian. 1997. Glucose in Saccharomyces cerevisiae. Yeast 22:111–127.
derepression of gluconeogenic enzymes in Saccharomyces cerevisiae corre- 328. Schawalder, S. B., M. Kabani, I. Howard, U. Choudhury, M. Werner, and
lates with phosphorylation of the gene activator Cat8p. Mol. Cell. Biol. D. Shore. 2004. Growth-regulated recruitment of the essential yeast ribo-
17:2502–2510. somal protein gene activator Ifh1. Nature 432:1058–1061.
304. Randez-Gil, F., P. Herrero, P. Sanz, J. A. Prieto, and F. Moreno. 1998. 329. Scheffler, I. E., B. J. de la Cruz, and S. Prieto. 1998. Control of mRNA
Hexokinase PII has a double cytosolic-nuclear localisation in Saccharomyces turnover as a mechanism of glucose repression in Saccharomyces cerevisiae.
cerevisiae. FEBS Lett. 425:475–478. Int. J. Biochem. Cell. Biol. 30:1175–1193.
305. Randez-Gil, F., P. Sanz, K. D. Entian, and J. A. Prieto. 1998. Carbon 330. Schmelzle, T., T. Beck, D. E. Martin, and M. N. Hall. 2004. Activation of
source-dependent phosphorylation of hexokinase PII and its role in the the RAS/cyclic AMP pathway suppresses a TOR deficiency in yeast. Mol.
glucose-signaling response in yeast. Mol. Cell. Biol. 18:2940–2948. Cell. Biol. 24:338–351.
306. Rauscher, F. J., III. 1993. The WT1 Wilms tumor gene product: a devel- 331. Schmidt, M. C., and R. R. McCartney. 2000. ␤-Subunits of Snf1 kinase are
opmentally regulated transcription factor in the kidney that functions as a required for kinase function and substrate definition. EMBO J. 19:4936–
tumor suppressor. FASEB J. 7:896–903. 4943.
307. Regelmann, J., T. Schule, F. S. Josupeit, J. Horak, M. Rose, K. D. Entian, 332. Schmidt, M. C., R. R. McCartney, X. Zhang, T. S. Tillman, H. Solimeo, S.
M. Thumm, and D. H. Wolf. 2003. Catabolite degradation of fructose-1,6- Wolfl, C. Almonte, and S. C. Watkins. 1999. Std1 and Mth1 proteins
bisphosphatase in the yeast Saccharomyces cerevisiae: a genome-wide screen interact with the glucose sensors to control glucose-regulated gene expres-
identifies eight novel GID genes and indicates the existence of two degra- sion in Saccharomyces cerevisiae. Mol. Cell. Biol. 19:4561–4571.
dation pathways. Mol. Biol. Cell 14:1652–1663. 333. Schneper, L., A. Krauss, R. Miyamoto, S. Fang, and J. R. Broach. 2004. The
308. Reifenberger, E., K. Freidel, and M. Ciriacy. 1995. Identification of novel Ras/protein kinase A pathway acts in parallel with the Mob2/Cbk1 pathway
HXT genes in Saccharomyces cerevisiae reveals the impact of individual to effect cell cycle progression and proper bud site selection. Eukaryot. Cell
hexose transporters on glycolytic flux. Mol. Microbiol. 16:157–167. 3:108–120.
VOL. 70, 2006 GLUCOSE SIGNALING IN S. CEREVISIAE 281

334. Schork, S. M., G. Bee, M. Thumm, and D. H. Wolf. 1994. Site of catabolite IRA2 encode proteins that may be functionally equivalent to mammalian
inactivation. Nature 369:283–284. ras GTPase activating protein. Cell 60:803–807.
335. Schuller, H. J. 2003. Transcriptional control of nonfermentative metabo- 361. Tatchell, K. 1986. RAS genes and growth control in Saccharomyces cerevisiae.
lism in the yeast Saccharomyces cerevisiae. Curr. Genet. 43:139–160. J. Bacteriol. 166:364–367.
336. Schuller, H. J., and K. D. Entian. 1991. Extragenic suppressors of yeast 362. Tatchell, K., D. T. Chaleff, D. DeFeo-Jones, and E. M. Scolnick. 1984.
glucose derepression mutants leading to constitutive synthesis of several Requirement of either of a pair of ras-related genes of Saccharomyces
glucose-repressible enzymes. J. Bacteriol. 173:2045–2052. cerevisiae for spore viability. Nature 309:523–527.
337. Schwartz, D., C. J. Decker, and R. Parker. 2003. The enhancer of decapping 363. Tatchell, K., L. C. Robinson, and M. Breitenbach. 1985. RAS2 of Saccharo-
proteins, Edc1p and Edc2p, bind RNA and stimulate the activity of the myces cerevisiae is required for gluconeogenic growth and proper response
decapping enzyme. RNA 9:239–251. to nutrient limitation. Proc. Natl. Acad. Sci. USA 82:3785–3789.
338. Shima, F., Y. Yamawaki-Kataoka, C. Yanagihara, M. Tamada, T. Okada, 364. Taylor, W. E., and E. T. Young. 1990. cAMP-dependent phosphorylation
K. Kariya, and T. Kataoka. 1997. Effect of association with adenylyl cyclase- and inactivation of yeast transcription factor ADR1 does not affect DNA
associated protein on the interaction of yeast adenylyl cyclase with Ras binding. Proc. Natl. Acad. Sci. USA 87:4098–4102.
protein. Mol. Cell. Biol. 17:1057–1064. 365. Thevelein, J. M., and J. H. de Winde. 1999. Novel sensing mechanisms and
339. Shore, D. 1994. RAP1: a protean regulator in yeast. Trends Genet. 10:408– targets for the cAMP-protein kinase A pathway in the yeast Saccharomyces
412. cerevisiae. Mol. Microbiol. 33:904–918.
340. Siebert, G. 1961. Enzyme and substrate der glykolyse in isolierten 366. Thompson-Jaeger, S., J. Francois, J. P. Gaughran, and K. Tatchell. 1991.
zellkernen. Biochem. Z. 334:369. Deletion of SNF1 affects the nutrient response of yeast and resembles
341. Skowyra, D., K. L. Craig, M. Tyers, S. J. Elledge, and J. W. Harper. 1997. mutations which activate the adenylate cyclase pathway. Genetics 129:697–
F-box proteins are receptors that recruit phosphorylated substrates to the

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


706.
SCF ubiquitin-ligase complex. Cell 91:209–219. 367. Toda, T., I. Uno, T. Ishikawa, S. Powers, T. Kataoka, D. Broek, S. Cameron,
342. Smith, A., M. P. Ward, and S. Garrett. 1998. Yeast PKA represses Msn2p/ J. Broach, K. Matsumoto, and M. Wigler. 1985. In yeast, RAS proteins are
Msn4p-dependent gene expression to regulate growth, stress response and controlling elements of adenylate cyclase. Cell 40:27–36.
glycogen accumulation. EMBO J. 17:3556–3564. 368. Toda, T., S. Cameron, P. Sass, and M. Wigler. 1988. SCH9, a gene of
343. Smith, F. C., S. P. Davies, W. A. Wilson, D. Carling, and D. G. Hardie. 1999. Saccharomyces cerevisiae that encodes a protein distinct from, but function-
The SNF1 kinase complex from Saccharomyces cerevisiae phosphorylates ally and structurally related to, cAMP-dependent protein kinase catalytic
the transcriptional repressor protein Mig1p in vitro at four sites within or subunits. Genes Dev. 2:517–527.
near regulatory domain 1. FEBS Lett. 453:219–223. 369. Toda, T., S. Cameron, P. Sass, M. Zoller, and M. Wigler. 1987. Three
344. Smogorzewska, A., and T. de Lange. 2004. Regulation of telomerase by different genes in S. cerevisiae encode the catalytic subunits of the cAMP-
telomeric proteins. Annu. Rev. Biochem. 73:177–208. dependent protein kinase. Cell 50:277–287.
345. Sobering, A. K., M. J. Romeo, H. A. Vay, and D. E. Levin. 2003. A novel Ras 370. Toda, T., S. Cameron, P. Sass, M. Zoller, J. D. Scott, B. McMullen, M.
inhibitor, Eri1, engages yeast Ras at the endoplasmic reticulum. Mol. Cell. Hurwitz, E. G. Krebs, and M. Wigler. 1987. Cloning and characterization of
Biol. 23:4983–4990. BCY1, a locus encoding a regulatory subunit of the cyclic AMP-dependent
346. Song, W., I. Treich, N. Qian, S. Kuchin, and M. Carlson. 1996. SSN genes protein kinase in Saccharomyces cerevisiae. Mol. Cell. Biol. 7:1371–1377.
that affect transcriptional repression in Saccharomyces cerevisiae encode
371. Tokiwa, G., M. Tyers, T. Volpe, and B. Futcher. 1994. Inhibition of G1
SIN4, ROX3, and SRB proteins associated with RNA polymerase II. Mol.
cyclin activity by the Ras/cAMP pathway in yeast. Nature 371:342–345.
Cell. Biol. 16:115–120.
372. Tomas-Cobos, L., and P. Sanz. 2002. Active Snf1 protein kinase inhibits
347. Spielewoy, N., K. Flick, T. I. Kalashnikova, J. R. Walker, and C. Witten-
expression of the Saccharomyces cerevisiae HXT1 glucose transporter gene.
berg. 2004. Regulation and recognition of SCFGrr1 targets in the glucose
Biochem. J. 398:657–663.
and amino acid signaling pathways. Mol. Cell. Biol. 24:8994–9005.
373. Tong, A. H., M. Evangelista, A. B. Parsons, H. Xu, G. D. Bader, N. Page, M.
348. Stanhope-Baker, P., P. M. Kessler, W. Li, M. L. Agarwal, and B. R. Wil-
Robinson, S. Raghibizadeh, C. W. Hogue, H. Bussey, B. Andrews, M. Tyers,
liams. 2004. The Wilms tumor suppressor-1 target gene podocalyxin is
and C. Boone. 2001. Systematic genetic analysis with ordered arrays of yeast
transcriptionally repressed by p53. J. Biol. Chem. 279:33575–33585.
deletion mutants. Science 294:2364–2368.
349. Stanway, C. A., A. Chambers, A. J. Kingsman, and S. M. Kingsman. 1989.
374. Tornow, J., and G. M. Santangelo. 1990. Efficient expression of the Sac-
Characterization of the transcriptional potency of sub-elements of the UAS
charomyces cerevisiae glycolytic gene ADH1 is dependent upon a cis-acting
of the yeast PGK gene in a PGK mini-promoter. Nucleic Acids Res. 17:
regulatory element (UASRPG) found initially in genes encoding ribosomal
9205–9218.
proteins. Gene 90:79–85.
350. Stark, M. J. 1996. Yeast protein serine/threonine phosphatases: multiple
roles and diverse regulation. Yeast 12:1647–1675. 375. Tornow, J., X. Zeng, W. Gao, and G. M. Santangelo. 1993. GCR1, a
351. Stiller, J. W., E. C. Duffield, and B. D. Hall. 1998. Amitochondriate amoebae transcriptional activator in Saccharomyces cerevisiae, complexes with RAP1
and the evolution of DNA-dependent RNA polymerase II. Proc. Natl. and can function without its DNA binding domain. EMBO J. 12:2431–2437.
Acad. Sci. USA 95:11769–11774. 376. Treitel, M. A., and M. Carlson. 1995. Repression by SSN6-TUP1 is directed
352. Struhl, K., D. Kadosh, M. Keaveney, L. Kuras, and Z. Moqtaderi. 1998. by MIG1, a repressor/activator protein. Proc. Natl. Acad. Sci. USA 92:
Activation and repression mechanisms in yeast. Cold Spring Harbor Symp. 3132–3136.
Quant. Biol. 63:413–421. 377. Treitel, M. A., S. Kuchin, and M. Carlson. 1998. Snf1 protein kinase regulates
353. Stuart, J. S., D. L. Frederick, C. M. Varner, and K. Tatchell. 1994. The phosphorylation of the Mig1 repressor in Saccharomyces cerevisiae. Mol. Cell.
mutant type 1 protein phosphatase encoded by glc7-1 from Saccharomyces Biol. 18:6273–6280.
cerevisiae fails to interact productively with the GAC1-encoded regulatory 378. Tu, J., and M. Carlson. 1994. The GLC7 type 1 protein phosphatase is
subunit. Mol. Cell. Biol. 14:896–905. required for glucose repression in Saccharomyces cerevisiae. Mol. Cell. Biol.
354. Sutherland, C. M., S. A. Hawley, R. R. McCartney, A. Leech, M. J. Stark, 14:6789–6796.
M. C. Schmidt, and D. G. Hardie. 2003. Elm1p is one of three upstream 379. Tu, J., and M. Carlson. 1995. REG1 binds to protein phosphatase type 1
kinases for the Saccharomyces cerevisiae SNF1 complex. Curr. Biol. 13: and regulates glucose repression in Saccharomyces cerevisiae. EMBO J.
1299–1305. 14:5939–5946.
355. Suzuki, N., H. R. Choe, Y. Nishida, Y. Yanawaki-Kataoka, S. Ohnishi, T. 380. Tu, J., W. Song, and M. Carlson. 1996. Protein phosphatase type 1 interacts
Tamaoki, and T. Kataoka. 1990. Leucine-rich repeats and carboxyl termi- with proteins required for meiosis and other cellular processes in Saccharo-
nus are required for interaction of yeast adenylate cyclase with RAS pro- myces cerevisiae. Mol. Cell. Biol. 16:4199–4206.
teins. Proc. Natl. Acad. Sci. USA 87:8711–8715. 381. Tung, K. S., and A. K. Hopper. 1995. The glucose repression and RAS-
356. Tachibana, C., J. Y. Yoo, J. B. Tagne, N. Kacherovsky, T. I. Lee, and E. T. cAMP signal transduction pathways of Saccharomyces cerevisiae each affect
Young. 2005. Combined global localization analysis and transcriptome data RNA processing and the synthesis of a reporter protein. Mol. Gen. Genet.
identify genes that are directly coregulated by Adr1 and Cat8. Mol. Cell. 247:48–54.
Biol. 25:2138–2146. 382. Tung, K. S., L. L. Norbeck, S. L. Nolan, N. S. Atkinson, and A. K. Hopper.
357. Tachikawa, H., A. Bloecher, K. Tatchell, and A. M. Neiman. 2001. A 1992. SRN1, a yeast gene involved in RNA processing, is identical to
Gip1p-Glc7p phosphatase complex regulates septin organization and spore HEX2/REG1, a negative regulator in glucose repression. Mol. Cell. Biol.
wall formation. J. Cell Biol. 155:797–808. 12:2673–2680.
358. Tanaka, K., B. K. Lin, D. R. Wood, and F. Tamanoi. 1991. IRA2, an 383. Turkel, S., T. Turgut, M. C. Lopez, H. Uemura, and H. V. Baker. 2003.
upstream negative regulator of RAS in yeast, is a RAS GTPase-activating Mutations in GCR1 affect SUC2 gene expression in Saccharomyces cerevisiae.
protein. Proc. Natl. Acad. Sci. USA 88:468–472. Mol. Genet. Genomics 268:825–831.
359. Tanaka, K., K. Matsumoto, and A. Toh-e. 1989. IRA1, an inhibitory regu- 384. Tzamarias, D., and K. Struhl. 1995. Distinct TPR motifs of Cyc8 are
lator of the RAS-cyclic AMP pathway in Saccharomyces cerevisiae. Mol. involved in recruiting the Cyc8-Tup1 corepressor complex to differentially
Cell. Biol. 9:757–768. regulated promoters. Genes Dev. 9:821–831.
360. Tanaka, K., M. Nakafuku, T. Satoh, M. S. Marshall, J. B. Gibbs, K. 385. Vallier, L. G., and M. Carlson. 1991. New SNF genes, GAL11 and GRR1
Matsumoto, Y. Kaziro, and A. Toh-e. 1990. S. cerevisiae genes IRA1 and affect SUC2 expression in Saccharomyces cerevisiae. Genetics 129:675–684.
282 SANTANGELO MICROBIOL. MOL. BIOL. REV.

386. Vallier, L. G., and M. Carlson. 1994. Synergistic release from glucose 414. Willems, A. R., M. Schwab, and M. Tyers. 2004. A hitchhiker’s guide to the
repression by mig1 and ssn mutations in Saccharomyces cerevisiae. Genetics cullin ubiquitin ligases: SCF and its kin. Biochim. Biophys. Acta 1695:133–170.
137:49–54. 415. Willett, C. E., C. M. Gelfman, and M. J. Holland. 1993. A complex regu-
387. Vallier, L. G., D. Coons, L. F. Bisson, and M. Carlson. 1994. Altered latory element from the yeast gene ENO2 modulates GCR1-dependent
regulatory responses to glucose are associated with a glucose transport transcriptional activation. Mol. Cell. Biol. 13:2623–2633.
defect in grr1 mutants of Saccharomyces cerevisiae. Genetics 136:1279–1285. 416. Willis, K. A., K. E. Barbara, B. B. Menon, J. Moffat, B. Andrews, and G. M.
388. van Hemert, M. J., G. P. van Heusden, and H. Y. Steensma. 2001. Yeast Santangelo. 2003. The global transcriptional activator of Saccharomyces cer-
14-3-3 proteins. Yeast 18:889–895. evisiae, Gcr1p, mediates the response to glucose by stimulating protein synthe-
389. Vanoni, M., R. L. Rossi, L. Querin, V. Zinzalla, and L. Alberghina. 2005. sis and CLN-dependent cell cycle progression. Genetics 165:1017–1029.
Glucose modulation of cell size in yeast. Biochem. Soc. Trans. 33:294–296. 417. Wills, C. 1990. Regulation of sugar and ethanol metabolism in Saccharo-
390. van Tuinen, E., and H. Riezman. 1987. Immunolocalization of glyceralde- myces cerevisiae. Crit. Rev. Biochem. Mol. Biol. 25:245–280.
hydes-3-phosphate dehydrogenase, hexokinase, and carboxypeptidase Y in 418. Willumsen, B. M., A. D. Cox, P. A. Solski, C. J. Der, and J. E. Buss. 1996.
yeast cells at the ultrastructural level. J. Histochem. Cytochem. 35:327–333. Novel determinants of H-Ras plasma membrane localization and transfor-
391. Vasudevan, S., and S. W. Peltz. 2001. Regulated ARE-mediated mRNA mation. Oncogene 13:1901–1909.
decay in Saccharomyces cerevisiae. Mol. Cell 7:1191–1200. 419. Wilson, B. A., M. Khalil, F. Tamanoi, and J. F. Cannon. 1993. New acti-
392. Velculescu, V. E., L. Zhang, W. Zhou, J. Vogelstein, M. A. Basrai, D. E. vated RAS2 mutations identified in Saccharomyces cerevisiae. Oncogene
Bassett Jr., P. Hieter, B. Vogelstein, and K. W. Kinzler. 1997. Character- 8:3441–3445.
ization of the yeast transcriptome. Cell 88:243–251. 420. Wilson, R. B., and K. Tatchell. 1988. SRA5 encodes the low-Km cyclic AMP
393. Verma, R., R. M. Feldman, and R. J. Deshaies. 1997. SIC1 is ubiquitinated phosphodiesterase of Saccharomyces cerevisiae. Mol. Cell. Biol. 8:505–510.
421. Wilson, W. A., S. A. Hawley, and D. G. Hardie. 1996. Glucose repression/

Downloaded from http://mmbr.asm.org/ on January 22, 2015 by guest


in vitro by a pathway that requires CDC4, CDC34, and cyclin/CDK activ-
ities. Mol. Biol. Cell 8:1427–1437. derepression in budding yeast: SNF1 protein kinase is activated by phos-
394. Versele, M., J. H. de Winde, and J. M. Thevelein. 1999. A novel regulator phorylation under derepressing conditions, and this correlates with a high
of G protein signaling in yeast, Rgs2, downregulates glucose-activation of AMP:ATP ratio. Curr. Biol. 6:1426–1434.
the cAMP pathway through direct inhibition of Gpa2. EMBO J. 18:5577– 422. Woods, A., S. R. Johnstone, K. Dickerson, F. C. Leiper, L. G. Fryer, D.
5591. Neumann, U. Schlattner, T. Wallimann, M. Carlson, and D. Carling. 2003.
395. Vincent, O., and M. Carlson. 1998. Sip4, a Snf1 kinase-dependent tran- LKB1 is the upstream kinase in the AMP-activated protein kinase cascade.
scriptional activator, binds to the carbon source-responsive element of Curr. Biol. 13:2004–2008.
gluconeogenic genes. EMBO J. 17:7002–7008. 423. Wu, J., and R. J. Trumbly. 1998. Multiple regulatory proteins mediate
396. Vincent, O., and M. Carlson. 1999. Gal83 mediates the interaction of the repression and activation by interaction with the yeast Mig1 binding site.
Snf1 kinase complex with the transcription activator Sip4. EMBO J. 18: Yeast 14:985–1000.
6672–6681. 424. Wu, M., L. Newcomb, and W. Heideman. 1999. Regulation of gene expres-
397. Vincent, O., R. Townley, S. Kuchin, and M. Carlson. 2001. Subcellular sion by glucose in Saccharomyces cerevisiae: a role for ADA2 and ADA3/
localization of the Snf1 kinase is regulated by specific ␤ subunits and a novel NGG1. J. Bacteriol. 181:4755–4760.
glucose signaling mechanism. Genes Dev. 15:1104–1114. 425. Wu, X., and K. Tatchell. 2001. Mutations in yeast protein phosphatase type
1 that affect targeting subunit binding. Biochemistry 40:7410–7420.
398. Vojtek, A. B., and D. G. Fraenkel. 1990. Phosphorylation of yeast hexoki-
426. Wu, X., H. Hart, C. Cheng, P. J. Roach, and K. Tatchell. 2001. Character-
nases. Eur. J. Biochem. 190:371–375.
ization of Gac1p, a regulatory subunit of protein phosphatase type I in-
399. von Mikecz, A., S. Zhang, M. Montminy, E. M. Tan, and P. Hemmerich. 2000.
volved in glycogen accumulation in Saccharomyces cerevisiae. Mol. Genet.
CREB-binding protein (CBP)/p300 and RNA polymerase II colocalize in tran-
Genomics 265:622–635.
scriptionally active domains in the nucleus. J. Cell Biol. 150:265–273.
427. Xu, X., J. D. Wightman, B. L. Geller, D. Avram, and A. T. Bakalinsky. 1994.
400. Vyas, V. K., S. Kuchin, and M. Carlson. 2001. Interaction of the repressors
Isolation and characterization of sulfite mutants of Saccharomyces cerevisiae.
Nrg1 and Nrg2 with the Snf1 protein kinase in Saccharomyces cerevisiae.
Curr. Genet. 25:488–496.
Genetics 158:563–572.
428. Xue, Y., M. Batlle, and J. P. Hirsch. 1998. GPR1 encodes a putative G
401. Vyas, V. K., S. Kuchin, C. D. Berkey, and M. Carlson. 2003. Snf1 kinases
protein-coupled receptor that associates with the Gpa2p G␣ subunit and
with different ␤-subunit isoforms play distinct roles in regulating haploid
functions in a Ras-independent pathway. EMBO J. 17:1996–2007.
invasive growth. Mol. Cell. Biol. 23:1341–1348.
429. Yang, X., E. J. Hubbard, and M. Carlson. 1992. A protein kinase substrate
402. Wade, J. T., D. B. Hall, and K. Struhl. 2004. The transcription factor Ifh1 identified by the two-hybrid system. Science 257:680–682.
is a key regulator of yeast ribosomal protein genes. Nature 432:1054–1058. 430. Yang, X., R. Jiang, and M. Carlson. 1994. A family of proteins containing
403. Wakula, P., M. Beullens, H. Ceulemans, W. Stalmans, and M. Bollen. 2003. a conserved domain that mediates interaction with the yeast SNF1 protein
Degeneracy and function of the ubiquitous RVXF motif that mediates kinase complex. EMBO J. 13:5878–5886.
binding to protein phosphatase-1. J. Biol. Chem. 278:18817–18823. 431. Yang, Z., and L. F. Bisson. 1996. The SKS1 protein kinase is a multicopy sup-
404. Walsh, E. P., D. J. Lamont, K. A. Beattie, and M. J. Stark. 2002. Novel pressor of the snf3 mutation of Saccharomyces cerevisiae. Yeast 12:1407–1419.
interactions of Saccharomyces cerevisiae type 1 protein phosphatase identi- 432. Young, E. T., K. M. Dombek, C. Tachibana, and T. Ideker. 2003. Multiple
fied by single-step affinity purification and mass spectrometry. Biochemistry pathways are co-regulated by the protein kinase Snf1 and the transcription
41:2409–2420. factors Adr1 and Cat8. J. Biol. Chem. 278:26146–26158.
405. Walther, K., and H. J. Schuller. 2001. Adr1 and Cat8 synergistically activate 433. Young, E. T., N. Kacherovsky, and K. Van Riper. 2002. Snf1 protein kinase
the glucose-regulated alcohol dehydrogenase gene ADH2 of the yeast Sac- regulates Adr1 binding to chromatin but not transcription activation.
charomyces cerevisiae. Microbiology 147:2037–2044. J. Biol. Chem. 277:38095–38103.
406. Wang, J., and R. Needleman. 1996. Removal of Mig1p binding site converts 434. Zaman, Z., A. Z. Ansari, L. Gaudreau, J. Nevado, and M. Ptashne. 1998.
a MAL63 constitutive mutant derived by interchromosomal gene conver- Gene transcription by recruitment. Cold Spring Harbor Symp. Quant. Biol.
sion to glucose insensitivity. Genetics 142:51–63. 63:167–171.
407. Wang, L., G. Renault, H. Garreau, and M. Jacquet. 2004. Stress induces 435. Zeng, X., S. J. Deminoff, and G. M. Santangelo. 1997. Specialized Rap1p/
depletion of Cdc25p and decreases the cAMP producing capability in Sac- Gcr1p transcriptional activation through Gcr1p DNA contacts requires
charomyces cerevisiae. Microbiology 150:3383–3391. Gcr2p, as does hyperphosphorylation of Gcr1p. Genetics 147:493–505.
408. Wang, Y., M. Pierce, L. Schneper, C. G. Guldal, X. Zhang, S. Tavazoie, and 436. Zenke, F. T., R. Engles, V. Vollenbroich, J. Meyer, C. P. Hollenberg, and
J. R. Broach. 2004. Ras and Gpa2 mediate one branch of a redundant K. D. Breunig. 1996. Activation of Gal4p by galactose-dependent interac-
glucose signaling pathway in yeast. PLoS Biol. 2:0610–0622. tion of galactokinase and Gal80p. Science 272:1662–1665.
409. Warner, J. R. 1989. Synthesis of ribosomes in Saccharomyces cerevisiae. 437. Zhang, S., S. Guha, and F. C. Volkert. 1995. The Saccharomyces SHP1
Microbiol. Rev. 53:256–271. gene, which encodes a regulator of phosphoprotein phosphatase 1 with
410. Warner, J. R. 1999. The economics of ribosome biosynthesis in yeast. differential effects on glycogen metabolism, meiotic differentiation, and
Trends Biochem. Sci. 24:437–440. mitotic cell cycle progression. Mol. Cell. Biol. 15:2037–2050.
411. Weeks, G., and G. B. Spiegelman. 2003. Roles played by Ras subfamily 438. Zhou, H., and F. Winston. 2001. NRG1 is required for glucose repression of
proteins in the cell and developmental biology of microorganisms. Cell the SUC2 and GAL genes of Saccharomyces cerevisiae. BMC Genet. 2:5.
Signal. 15:901–909. 439. Zhu, X., N. Demolis, M. Jacquet, and T. Michaeli. 2000. MSI1 suppresses
412. Wek, R. C., J. F. Cannon, T. E. Dever, and A. G. Hinnebusch. 1992. hyperactive RAS via the cAMP-dependent protein kinase and indepen-
Truncated protein phosphatase GLC7 restores translational activation of dently of chromatin assembly factor-1. Curr. Genet. 38:60–70.
GCN4 expression in yeast mutants defective for the eIF-2 alpha kinase 440. Zimmermann, F. K., I. Kaufmann, H. Rasenberger, and P. Haubetamann.
GCN2. Mol. Cell. Biol. 12:5700–5710. 1977. Genetics of carbon catabolite repression in Saccharomycess cerevisiae:
413. Wieczorke, R., S. Krampe, T. Weierstall, K. Freidel, C. P. Hollenberg, and genes involved in the derepression process. Mol. Gen. Genet. 151:95–103.
E. Boles. 1999. Concurrent knock-out of at least 20 transporter genes is 441. Zurita-Martinez, S. A., and M. E. Cardenas. 2005. Tor and cyclic AMP-
required to block uptake of hexoses in Saccharomyces Cerevisiae. FEBS protein kinase A: two parallel pathways regulating expression of genes
Lett. 464:123–128. required for cell growth. Eukaryot. Cell 4:63–71.

You might also like