+ All Categories
Home > Documents > Hypoxia and Gene Expression in Eukaryotic Microbes

Hypoxia and Gene Expression in Eukaryotic Microbes

Date post: 11-Dec-2016
Category:
Upload: geraldine
View: 212 times
Download: 0 times
Share this document with a friend
25
Hypoxia and Gene Expression in Eukaryotic Microbes Geraldine Butler School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland; email: [email protected] Annu. Rev. Microbiol. 2013. 67:291–312 First published online as a Review in Advance on June 26, 2013 The Annual Review of Microbiology is online at micro.annualreviews.org This article’s doi: 10.1146/annurev-micro-092412-155658 Copyright c 2013 by Annual Reviews. All rights reserved Keywords SREBP, fungi, evolution, biofilm, UPC2, ROX1 Abstract The response of eukaryotic microbes to low-oxygen (hypoxic) conditions is strongly regulated at the level of transcription. Comparative analysis shows that some of the transcriptional regulators (such as the sterol reg- ulatory element-binding proteins, or SREBPs) are of ancient origin and probably regulate sterol synthesis in most eukaryotic microbes. However, in some fungi SREBPs have been replaced by a zinc-finger transcription factor (Upc2). Nuclear localization of fungal SREBPs is determined by regulated proteolysis, either by site-specific proteases or by an E3 ligase complex and the proteasome. The exact mechanisms of oxygen sensing are not fully char- acterized but involve responding to low levels of heme and/or sterols and possibly to levels of nitric oxide and reactive oxygen species. Changes in central carbon metabolism (glycolysis and respiration) are a core hypoxic response in some, but not all, fungal species. Adaptation to hypoxia is an important virulence characteristic of pathogenic fungi. 291 Annu. Rev. Microbiol. 2013.67:291-312. Downloaded from www.annualreviews.org by University of Leeds on 09/16/13. For personal use only.
Transcript

MI67CH14-Butler ARI 5 August 2013 17:3

Hypoxia and Gene Expressionin Eukaryotic MicrobesGeraldine ButlerSchool of Biomolecular and Biomedical Science, Conway Institute, University College Dublin,Belfield, Dublin 4, Ireland; email: [email protected]

Annu. Rev. Microbiol. 2013. 67:291–312

First published online as a Review in Advance onJune 26, 2013

The Annual Review of Microbiology is online atmicro.annualreviews.org

This article’s doi:10.1146/annurev-micro-092412-155658

Copyright c© 2013 by Annual Reviews.All rights reserved

Keywords

SREBP, fungi, evolution, biofilm, UPC2, ROX1

Abstract

The response of eukaryotic microbes to low-oxygen (hypoxic) conditionsis strongly regulated at the level of transcription. Comparative analysisshows that some of the transcriptional regulators (such as the sterol reg-ulatory element-binding proteins, or SREBPs) are of ancient origin andprobably regulate sterol synthesis in most eukaryotic microbes. However, insome fungi SREBPs have been replaced by a zinc-finger transcription factor(Upc2). Nuclear localization of fungal SREBPs is determined by regulatedproteolysis, either by site-specific proteases or by an E3 ligase complex andthe proteasome. The exact mechanisms of oxygen sensing are not fully char-acterized but involve responding to low levels of heme and/or sterols andpossibly to levels of nitric oxide and reactive oxygen species. Changes incentral carbon metabolism (glycolysis and respiration) are a core hypoxicresponse in some, but not all, fungal species. Adaptation to hypoxia is animportant virulence characteristic of pathogenic fungi.

291

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

Metazoa:clade comprisingmulticellular animals

Hypoxia: reduction inoxygen concentration

Contents

INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292CONSERVATION AND DISTRIBUTION OF THE MAJOR

HYPOXIC REGULATORS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294Hypoxia-Inducible Factor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294Distribution of SREBP and Upc2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 294

SENSING OF OXYGEN LEVELS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296SREBP Pathway . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296Heme-Activated Proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 299Other Pathways . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300

REGULATION OF STEROL, LIPID, AND IRON METABOLISM GENES . . . . . 301REGULATION OF GLYCOLYTIC GENE EXPRESSION . . . . . . . . . . . . . . . . . . . . . . . 302EXPRESSION OF CELL WALL GENES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 302HYPOXIA AND PATHOGENESIS. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303

Biofilms and Hypoxia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303Hypoxic Regulators and Virulence. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 303

CONCLUSIONS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 304

INTRODUCTION

The level of oxygen in the atmosphere was very low until the evolution of photosynthetic or-ganisms (cyanobacteria), approximately 2.5–3 billion years ago (117). Oxygen concentrations in-creased to significant levels about 1.5 billion years ago, which coincided with the appearanceof the first eukaryotes. These organisms could generate energy by oxidative phosphorylationin internal organelles, the mitochondria. The first fungi probably appeared at the same time(51, 52). Oxygen levels continued to fluctuate, particularly during the expansion of the metazoans(past 500 million years). Oxygen is a toxic molecule; it is therefore not surprising that organismsevolved sophisticated mechanisms for responding to altered levels (130).

Many eukaryotes are also exposed to vastly different levels of oxygen in environmental niches.Some eukaryotic microbes are obligate anaerobes, such as the flagellated Neocallimastigales (re-lated to chytrids; 55), which were first isolated from highly anaerobic sheep rumen (42, 133).Other facultative anaerobes are found in stagnant waters (42). However, even obligate aer-obes and facultative aerobes can be exposed to long-lasting or transient hypoxic conditions.For example, many fungi grow in soil where oxygen levels are low. Others (such as Aspergillusfumigatus) colonize compost heaps where the concentration of O2 approaches 1.5% (136). Fun-gal pathogens are also exposed to variations in oxygen concentrations in the mammalian host(35).

Most organisms can sense oxygen concentrations and respond rapidly to changes. The tran-scriptional response of many fungal species during the early stages of adaptation to low-oxygenconditions has been well characterized (9, 11, 35, 46, 62, 75, 77, 119, 127). Genes that are differen-tially expressed in response to oxygen levels can be divided into two general groups: aerobic genesthat are expressed in normoxic conditions, and hypoxic genes that are expressed when oxygenis low or completely absent (104). A core transcriptional response is shared by most eukaryotes,including increased expression of genes encoding enzymes in oxygen-dependent pathways, such

292 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

Table 1 Summary of hypoxic regulation in fungi

SignalaRegulatory protein

(species)b Function Target pathways/genesc

Sterol levels SREBP (Cryptococcusneoformans,Schizosaccharomyces pombe,Aspergillus fumigatus)

Activator Sterol biosynthesis, ironhomeostasisd

Sterol levels/hemelevels

Upc2 (Saccharomycescerevisiae, Candida glabrata,Candida albicans, Candidaparapsilosis)

Ecm22 (Saccharomycescerevisiae, Candida glabrata)

Activator Sterol biosynthesis, steroltransportd

DAN/TIR cell wall genesd

Unknown Mga2 (Saccharomycescerevisiae, Kluyveromyceslactis)

Activator Lipid synthesis

Heme levels Hap1 (Saccharomycescerevisiae, Kluyveromyceslactis)

Activator Respiratory genes

Heme levels Rox1 (Saccharomyces cerevisiae) Repressor Respiratory genesUnknown Hog1 (Saccharomyces

cerevisiae)Activator PAU cell wall genes

O2 Ofd2 (Schizosaccharomycespombe)

Preventsrepression

Respiratory genes

Unknown Tye7 (Candida albicans) Activator GlycolysisUnknown Efg1 (Candida albicans) Activator/repressor Glycolysis

Biofilm genesNO Unknown (Saccharomyces

cerevisiae)– Cytochrome c

aSignal of oxygen levels, where known.bSpecies with experimental evidence are listed. Orthologs in other species are likely to have similar functions.cMajor target pathways are listed. Not all target genes in all species have been identified.dTarget genes that may be restricted to one species or a group of species.Abbreviations: NO, nitric oxide; SREBP, sterol-regulatory element binding protein.

as synthesis of sterols, lipids, and heme (14, 35) (Table 1). Other changes seen in some but notall species include an increase in glycolysis and a decrease in respiration, as well as changes in cellwall gene expression (5, 46, 75, 77, 127) (Table 1).

How oxygen is sensed by fungi is not completely understood. Sterol biosynthesis is an oxygen-dependent process, and in several species lowered sterols are apparently used to sense oxygenlevels (24, 27, 29, 62, 134). Similarly, heme (which contains an iron group that binds oxygen)can act as a secondary signal for oxygen, best characterized in Saccharomyces cerevisiae (11, 29,145).

In this review I examine how the transcriptional response of eukaryotic microbes to hypoxicconditions is regulated, concentrating predominantly on the fungi, which are the best character-ized. I also investigate the evolution of core regulatory molecules—some ancient and some veryrecent (summarized in Table 1).

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 293

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

HIF:hypoxia-induciblefactor

ARNT: arylhydrocarbon receptornuclear translocator

Basichelix-loop-helix(bHLH): a DNAbinding domain

SREBP:sterol regulatoryelement-bindingprotein

CONSERVATION AND DISTRIBUTION OF THE MAJORHYPOXIC REGULATORS

Hypoxia-Inducible Factor

Hypoxia-inducible factor (HIF) is probably the best-studied regulator of the oxygen responsein mammals (45). HIFs are dimeric transcription factors, consisting of a regulated subunit (oneof multiple HIFα components) and a constitutively expressed subunit [from multiple HIFβ orthe aryl hydrocarbon receptor nuclear translocator (ARNT) components]. When O2 levels arehigh, HIFα is modified by proline hydroxylases and subsequently degraded via the ubiquitin–proteasome pathway. As oxygen levels drop, hydroxylase activity is inhibited; HIFα enters thenucleus, dimerizes with HIFβ, and regulates the expression of hundreds of genes. Expression ofglucose transporters, glycolytic enzymes, and lactate dehydrogenase A is induced, which increasesglycolysis (129). The efficiency of mitochondrial respiration is increased (117). Genes associatedwith erythropoiesis, angiogenesis, and vasodilation are also regulated (47).

HIF subunits are well conserved throughout the Metazoa, from mammals to arthropods tocnidaria (50). The function is also well conserved; for example, HIFα is also regulated by hydrox-ylation in worms and flies (4, 34). However, HIF orthologs in fungi have never been reported. Todetermine the origin of HIF proteins, we took advantage of recent genome-sequencing projectsaimed at characterizing the evolution of multicellular eukaryotes (112). As suggested previously(88), there are no obvious HIFα orthologs outside the Metazoa (Figure 1). Surprisingly however,there are two HIFβ-like proteins in the genome of Capsaspora owczarzaki, a symbiotic amoeba(54). The proteins have basic helix-loop-helix (bHLH) and PAS (Per/Arnt/Sim) domains, whichcan act as sensors for oxygen, light, and redox potential (128). C. owczarzaki is a member of theFilasterea, a sister group to the choanoflagellates and the Metazoa (Figure 1). We could notidentify other HIFβ orthologs in the choanoflagellates or in older diverging groups, includingFungi; however, very little sequence information is available (Figure 1). It is likely that HIFβ

arose in the common ancestor of the Filasterea, the choanoflagellates, and the Metazoa, but theoxygen-dependent HIFα subunits did not appear until later, in the ancestor of the Metazoa. It isalso possible that C. owczarzaki obtained the HIFβ-like proteins by horizontal gene transfer froman unknown source (e.g., 121); this will be resolved as more sequenced genomes become available.It is not clear what role the HIFβ-like proteins play in C. owczarzaki. However, in mammals,HIF-1β is also involved in the response to dioxins and forms dimers with the aryl hydrocarbonreceptor as well as with HIFα (139).

Distribution of SREBP and Upc2

Sterol regulatory element-binding proteins (SREBPs) are an ancient family of regulators that areassociated with the hypoxic response. Mammals have two SREBP genes (SREBP-1 and SREBP-2)that regulate the expression of fatty acid and cholesterol synthesis and cholesterol uptake genes (60).Invertebrates such as Caenorhabditis elegans and Drosophila melanogaster have only one SREBP or-tholog and tend to be cholesterol auxotrophs (101). Some fungi such as A. fumigatus and Schizosac-charomyces pombe also have SREBP orthologs (101). The localization and activation of SREBPsare described in the next section.

Again, interrogation of recent genome sequences (112) provides insights into the evolutionof the SREBP pathway. Like HIF, SREBPs have bHLH domains but are unique in that a tyro-sine residue replaces a well-conserved arginine (71). SREBP homologs are conserved in Metazoa,Choanoflagellata, Filasterea, Ichthyosporea, and Fungi (Figure 1). The SREBP cleavage acti-vating proteins (SCAPs), which interact with SREBPs, are less widely distributed but have been

294 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

Homo sapiens

Drosophila melanogaster

Caenorhabditis elegans

Monosiga brevicollis

Salpingoeca rosetta

Capsaspora owczarzaki

+HIFα

+HIFβ –HIFβ

SREBP

Sphaeroforma arctica

+UPC2

Mortierella verticillata

Cryptococcus neoformans

Schizosaccharomyces pombe

Aspergillus fumigatus

Saccharomyces cerevisiae Candida albicans Candida parapsilosis

Fonticula alba

Thecamonas trahens

Spizellomyces punctatus

Coccidioides immitis Histoplasma capsulatum

Neurospora crassa Podospora anserina Magnaporthe grisea

Sclerotinia sclerotiorum Botryotinia fuckeliana

–SCAP

+ + + + –

HIFα HIFβ SREBP SCAP UPC2

+ + + + –+ + + + –

– – + – –

– – + + –

– + + – –

– – + + –

– – + + –

– – + + –

– – + + –

– – + + –

– – + – –– – + – –– – + – –

– – + + –– – + + –– – + + –

– – + + –– – + + –

– – + – +– – + – +– – + – +

– – – – –

Nuclearia simplex ? ????

Laccaria bicolor Ustilago maydis

– – + + –– – + + –

FUNGI

OPISTHOKONTS

SCAP

+

Zygomycota

Basidiomycota

Apusozoa/Amoebozoa

Chytridiomycota

Metazoa

Filasterea

Ichthyosporea

Eurotiomycetes

Choanoflagellata

Sordariomycetes

Leotiomycetes

Saccharomycotina

Nucleariidae

Taphrinomycotina

Figure 1Conservation of hypoxic regulators in eukaryotic microbes. Phylogenetic relations are shown indiagrammatic form and are adapted from Reference 112. Plus signs and minus signs indicate presence andapparent absence of genes, respectively. No sequence information is available from the Nucleariidae. Themost likely timing of gene gains and losses is shown on the left-hand side.

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 295

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

SCAP: SREBPcleavage activatingprotein

Opisthokonts:members of the cladethat comprisesMetazoa, Fungi, andtheir close unicellularlineages

Saccharomycotina:a subphylum of theAscomycota thatincludes Saccharomycesand Candida clades

Whole genomeduplication (WGD)event: an event thattook place in theancestor ofSaccharomyces cerevisiaeand closely relatedspecies

identified in some choanoflagellates and Ichthyosporea, in ancient fungal lineages (Spizellomycespunctatus and Mortierella verticillata), and throughout fungi (Figure 1). There are several speciesin which SCAP has been lost, including the entire Eurotiomycetes lineage (e.g., A. fumigatus). Inaddition, there are no obvious homologs in the Amoebozoa. The simplest interpretation is thatthe SREBP signaling pathway arose in the ancestor of the opisthokonts, following the divergencefrom the Amoebozoa, and that components were subsequently lost in some lineages.

SREBPs act as hypoxic regulators in fungi, but it is not clear if they also respond to oxygensignals in the Metazoa (101). In the ascomycetes S. pombe and A. fumigatus, SREBP homologscontrol the expression of genes involved in biosynthesis of lipids, ergosterol, and heme (131, 141).SREBP function is also conserved in the basidiomycete Cryptococcus neoformans (24, 27).

Surprisingly, in some ascomycete species (the Saccharomycotina; Figure 1) there has been anevolutionary rewiring of the sterol synthesis pathway. In these species, the role of SREBP in sterolregulation has been replaced by a different transcription factor, Upc2. Upc2 shares no sequencesimilarity with SREBPs and has a GAL4-like Zn2Cys6 binuclear cluster DNA-binding domain.Several fungal proteins have the same domain, but Upc2 appears to be unique to, or highly derivedin, the Saccharomycotina (Figure 1). Upc2 regulates the expression of sterol synthesis and otherhypoxic genes in S. cerevisiae, Candida albicans, and Candida parapsilosis (46, 58, 134).

The most likely homologs of SREBPs in the Saccharomycotina are represented by Cph2 (inC. albicans) (14) and Hms1 (in S. cerevisiae); these have bHLH domains with the characteristictyrosine (14). However, the transmembrane domains associated with SREBP function are poorlyconserved, and the proteins are shorter. In addition, Cph2 regulates hyphal growth (80), andHMS1 is a regulator of pseudohyphal growth (89).

One hypothesis is that Upc2 took over the role of major hypoxic regulator of the sterol pathwayin the Saccharomycotina, allowing the SREBP homologs to degenerate and acquire new functionsas regulators of filamentation. It is also possible that the ancestral fungal SREBP had roles in bothhypoxic regulation and in determination of morphology. This is supported by the observation thatSrbA in A. fumigatus is required both for hypoxic regulation and for hyphal branching (141).

The function of Upc2 in S. cerevisiae and closely related yeasts is complicated because therewas a whole genome duplication (WGD) event in a recent ancestor (142). Although the additionalcopies of most genes were subsequently lost, two Upc2 paralogs (Upc2 and Ecm22) were retainedin S. cerevisiae. Both paralogs play roles in regulating the expression of sterol synthesis genes (134),and the promoters of almost one-third of genes whose expression is induced in hypoxic conditionsin S. cerevisiae contain potential Upc2-binding sites (77). Upc2 probably senses lowered sterollevels and also lowered heme levels as a means of sensing oxygen. Treatment with sterol-depletingdrugs has an effect on transcription similar to that of lowering oxygen, and requires Upc2 inS. cerevisiae, C. albicans, and C. parapsilosis (29, 46, 127).

Although Upc2 and SREBPs are not structurally related, there is an intriguing functionalsimilarity. Both Upc2 and Ecm22 are localized to intracellular membranes and may be traffickedto the nucleus following treatment with sterol-depleting drugs (92). This is similar to the changein location observed following processing of SREBP in mammals and several fungi.

SENSING OF OXYGEN LEVELS

SREBP Pathway

Sensing of sterol levels (and therefore, indirectly, of oxygen) by SREBPs is by far the best-studiedand the best-understood hypoxic mechanism in fungal systems (14, 36, 101). In mammals and insome fungi, SREBPs are synthesized as inactive precursors that are inserted into the endoplasmic

296 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

INSIG:insulin-induced gene

HMG-CoA:5-hydroxy-3-methylglutaryl–coenzyme A

reticulum (ER) membrane (Figure 2). The presence of two transmembrane domains ensures thatboth the C terminus and the N terminus containing the DNA-binding domain face the cytoplasm.The SREBPs associate with SCAP in the ER membrane. SCAPs bind sterols, keeping them ininactive conformations and retaining SREBP in the membrane. As sterol levels drop, the N-terminal region of SREBP is cleaved off and enters the nucleus, where it activates the expressionof target genes.

Although the basic mechanism of SREBP activation is conserved between mammals and fungi,there are also substantial differences. Mammalian SREBP/SCAP complexes are retained in the ERmembrane by interaction with INSIGs (insulin-induced genes). Cholesterol binds to SCAP, in-ducing a conformational change that causes SCAP to interact with INSIGs (2), whereas oxysterolsbind directly to INSIG (106). As sterol levels drop, the interaction between SCAP and INSIG isdisrupted, and the SREBP-SCAP complex is transported to the Golgi apparatus via COPII-coatedvesicles (85). SREBP is cleaved in the Golgi apparatus, and the N terminus is released and entersthe nucleus (36). INSIGs also regulate proteolysis of HMG-CoA (5-hydroxy-3-methylglutaryl-coenzyme A; required for the production of mevalonate, a rate-controlling step in cholesterolbiosynthesis) reductases in mammalian cells (120).

Homologs of INSIG are well conserved across the fungal kingdom [except for C. neoformans(14)]. However, they have no apparent role in regulating transport of the SREBP-SCAP complex.In S. pombe, the INSIG ortholog Ins1 regulates sterol synthesis, but only by inhibiting activityof HMG-CoA reductases (21). Ngs1, the S. cerevisiae ortholog of INSIG, controls HMG-CoAreductase through regulation of the protein levels of one isoform encoded by Hmg2. However,Ngs1 stabilizes rather than inhibits Hmg2 activity (20, 40).

Mammalian SREBP is cleaved by two site-directed proteases. The first (site-1 protease, orSP1) cleaves within the loop in the Golgi lumen, and the second (site-2 protease, or SP2) cleaveswithin the transmembrane domain (32, 33). Site-1 proteases belong to the Kexin 2 family; they arewell conserved throughout fungi and have many functions (114). Site-2 proteases are conservedin C. neoformans and in many basidiomycetes but appear to have been lost from the ascomycetes(13, 14, 25). In C. neoformans, deleting the SP-2 ortholog STP1 results in increased sensitivity tohypoxia (25, 27). Stp1 is required for processing the SREBP Sre1; when residues in the catalyticsite are mutated, processed forms of Sre1 fail to accumulate (13). Stp1 is also required for Sre1processing when sterols are lowered by treatment with azole drugs (13).

Processing SREBPs in the ascomycetes is very different from that in the basidiomyceteC. neoformans. A. fumigatus and related species, for example, have no SCAP homolog (Figures 1and 2), whereas S. pombe contains two SREBPs (Sre1 and Sre2). Sre2 is much shorter than Sre1,it does not bind SCAP, and it is constitutively cleaved irrespective of the levels of sterols (62).Stewart et al. (123, 124) showed that the Dsc E3 ligase complex, which transfers ubiquitin froman E2 ubiquitin-conjugating enzyme to a target, is required for processing both Sre1 and Sre2(Figure 2). Sre1 cleavage also requires the E2-conjugating enzyme Ubc4 and components of theproteasome. Stewart et al. (123, 124) suggest that Scp1 transports SREBPs to the Golgi apparatus,where they are ubiquitinated by Ubc4-Dsc and targeted to the proteasome for cleavage. Thismechanism may be a common hypoxic response in many ascomycete species, compensating forthe loss of SP2, and is supported by the recent demonstration that the complex is required alsofor cleavage of SrbA in A. fumigatus (140).

S. pombe Sre1 is regulated independently of sterol levels, by 2-oxogluturate Fe(II) dioxygenase(Ofd1) (14). Ofd1 is a member of the prolyl hydroxylase (PHD) family, of which some membersregulate degradation of HIFα in mammals. When oxygen levels are high, Ofd1 accelerates thedegradation of Sre1N via the proteasome. However, the transcription factor is stabilized in lowoxygen levels (63). Ofd1 acts as an oxygen sensor and an effector via two distinct domains; the

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 297

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

– O2

Nro1

O2 , sterols

O2 , sterols

? O2 , sterols

C

Sre1N

C

Sre1N

C

SrbA

CSre1

Scp1 Scp1SRE

Ofd1

-Sterol synthesis-Lipid synthesis

Stp1 protease?

Aspergillus fumigatus

Schizosaccharomyces pombe

Cryptococcus neoformans

Mammals

CSre1N

Sre1N

Scp1 Scp1

Dsc E3 ligase

Sre1N

SRE

Ofd1

Sre1N

+ O2

SRE

Ofd1

Dsc E3 ligase

? ?

Ofd1 ?

SRE

SrbA

Proteasome

Proteasome

?

?

?

Sterols

C

SREBP

CSre1

SCAP

SCAPSRE

-Sterol synthesis-Sterol import

S1 protease

S2 protease

SREBP

ENDOPLASMICRETICULUM GOLGI APPARATUS NUCLEUS

-Sterol synthesis-Lipid and heme synthesis

-Sterol synthesis-Iron homeostasis

INSIG

298 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

dioxygenase domain in the N terminus, which senses oxygen, and the C-terminal domain, whichaccelerates degradation.

Subsequent analysis showed that the C terminus of Ofd1 interacts with Nro1, a nuclear protein(82). The interaction is regulated by oxygen; when levels are low, Nro1 binds to Ofd1 and inhibitsdegradation of Sre1N. Nro1 also regulates the nuclear localization of Ofd1 (144). Surprisingly,Ofd1 and Nro1 regulate binding of Sre1N to DNA, as well as proteolysis (83). When oxygenlevels are high, Ofd1 binds (via its C-terminal domain) to Sre1N, preventing binding of the tran-scription factor to regulatory regions on DNA. As oxygen levels drop, Nro1 binds the C-terminaldomain of Ofd1, thus releasing Sre1N and allowing transcription of hypoxic genes (83).

Using a sophisticated mathematical model in combination with experimental measurements,Porter et al. (103) suggest that the inhibitory effect of Ofd1 on DNA binding of Sre1N in highoxygen levels is the most important step in hypoxic regulation. Oxygen-dependent degradation ofSre1N may speed up the response, but it is not absolutely necessary. Hypoxic regulation by Sre1Nis a feed-forward mechanism, in which changes in oxygen levels result in compensatory alterationsin metabolism (such as increased sterol synthesis) (103). This mechanism encompasses positivefeedback through self-regulation of Sre1N (64) and negative feedback through increased produc-tion of Ofd1 in low oxygen. Changing the concentration of Nro1, which competes with Ofd1 forbinding to Sre1N, also allows the cell to fine-tune the response to changing oxygen conditions.

The prolyl hydroxylase activity of Ofd1 does not appear to be important for the hypoxicresponse, which is very different from the regulation of HIF stability by PHD proteins (83). How-ever, Ofd1 is generally well conserved in fungal species (14). The S. cerevisiae ortholog (Tpa1) isrequired for translation termination (53, 67) and binds to Nro1 (72). Tpa1 represses the expressionof respiratory genes regulated by Hap1 (53). It is therefore possible that Ofd1 has been recruitedfor hypoxic regulation in several fungal species but acts through different mechanisms.

Heme-Activated Proteins

Heme biosynthesis requires oxygen, and the expression of many biosynthetic genes is increasedwhen oxygen levels are low. Reducing heme levels is utilized by S. cerevisiae (and probably otherfungi) to monitor oxygen levels by way of the transcription factor Hap1 and, in some cases, Upc2

←−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−−Figure 2The SREBP pathway in fungi and mammals. The SREBP/SCAP complex in mammals is maintained in theendoplasmic reticulum by interacting with INSIGs. A drop in sterol levels causes a change in theconformation of SCAP, release from INSIG, and transport to the Golgi apparatus. The N-terminal region isreleased by cleavage by two site-specific proteases, enters the nucleus, and activates target gene expression.The transport pathway in fungi, and the role of the Golgi apparatus, is not completely understood. TheSREBP Sre1 is membrane bound in a complex with the SCAP Scp1 in Cryptococcus neoformans andSchizosaccharomyces pombe; there is no SCAP in Aspergillus fumigatus, and the mechanism of retention is notknown. Cleavage of the N-terminal region of SREBP (by one Site-1 protease in C. neoformans, and theproteasome in S. pombe and A. fumigatus) is inhibited by oxygen and high sterol levels. The N-terminaltranscription factor binds to the sterol regulatory element (SRE) in the nucleus and activates the expressionof sterol metabolism and other genes. In S. pombe, when oxygen levels are high, Ofd1 binds to Sre1N,inhibiting DNA binding and targeting the transcription factor for degradation. When oxygen levels are low,Ofd1 is sequestered by the nuclear transporter Nro1. The role of Ofd1 in regulating hypoxia in other fungiis not known. Several putative and/or currently uncharacterized steps in the pathways are indicated withquestion marks. The figure is based on Reference 14, with additional information. Abbreviations: INSIG,insulin-induced gene; SCAP, SREBP cleavage activating protein; SREBP, sterol regulatory element-bindingprotein.

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 299

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

(29). In the absence of heme, Hap1 forms a high-molecular-weight complex that is incapable ofbinding DNA and is gradually reduced to a dimer as heme binds (145). Hap1 directly regulatesthe expression of many genes associated with respiration, such as the cytochrome c protein CYC1and the cytochrome c oxidase COX5A. However, one of the most important roles of Hap1 isto regulate the expression of ROX1, which encodes an HMG domain protein that represses theexpression of a large number of hypoxic genes (11).

In S. cerevisiae, many duplicated genes retained after the WGD event are associated withrespiration. For several of the gene pairs, the expression of one member is induced in responseto high oxygen, and the expression of the other is induced in hypoxic conditions (146). Thesegene pairs include CYC1 (heme activated) and CYC7 (heme repressed), COX5A (heme activated)and COX5B (heme repressed), and the translation initiation factors TIF51A (heme activated) andANB1 (heme repressed). The expression of the gene pairs is regulated by HAP1 and/or ROX1 (57,90, 146).

Despite the evidence from S. cerevisiae, the breadth of heme sensing in hypoxia is not clear.HAP1 is conserved throughout the ascomycetes but has not been characterized in many species.HAP1 from Kluyveromyces lactis, a close relative of S. cerevisiae, can complement the defect ofa hap1 deletion in S. cerevisiae and also regulate the expression of cytochrome c subunits (8).The expression of HAP1 is induced in hypoxia in both species (8). However, there are significantdifferences in the hypoxic responses. Unlike in S. cerevisiae, Hap1 is not required for the expressionof genes involved in cholesterol biosynthesis or oxidative stress in K. lactis (78).

Many of the differences in the hypoxic response between S. cerevisiae and K. lactis are likely tobe related to differences in Rox1 function. An apparent homolog of ROX1 in K. lactis plays no rolein the hypoxic response (37). Similarly, a homolog of ROX1 in C. albicans (called RFG1) does notregulate hypoxic expression but instead controls filamentous growth (66, 69). The repressive roleof Rox1 is probably confined to S. cerevisiae and some very closely related species, and it is unlikelythat Rox1 orthologs act as major fungal regulators of gene expression in hypoxia.

It is likely that heme sensing is important for assessing oxygen concentrations in manyspecies (19). Other heme-activated proteins have been characterized in S. cerevisiae, includingthe Hap2/3/4/5 complex, which binds to a CCAAT box upstream of genes required for growthon nonfermentable carbon sources (100). Hap2, Hap3, and Hap5 are required for DNA bind-ing, and Hap4 is a regulatory subunit (75). Hap2 and Hap3 are functionally interchangeable withthe CP1-A/B subunits in human cells (26). It is not known how heme activates the Hap2/3/4/5complex, but it is unlikely to be through DNA binding (75). The activity of CCAAT-box-bindingproteins in mammals also is modulated by heme treatment (95), suggesting that the involvementof heme in oxygen sensing may represent an ancient conserved mechanism.

Other Pathways

In S. pombe, Ofd2, a 2-OG-Fe(II) dioxygenase, acts on histone H2A, a major component ofchromatin (79). Ofd2 is similar in structure to Ofd1, which regulates activity of the SREBP Sre1.Lando et al. (79) found that the expression of genes involved in ATP synthesis and mitochondrialelectron transport, which are repressed in hypoxia, are further repressed when ofd2 is deleted.Ofd2 is targeted to the promoters of the oxidative phosphorylation genes. It is not clear exactlyhow Ofd2 modifies histone H2A; possibilities include hydroxylation or demethylation. However,it appears that Ofd2 senses oxygen levels and inhibits repression of respiratory genes.

Considerable evidence suggests that mitochondria and the respiratory chain are important forthe hypoxic response in microbial eukaryotes. In S. cerevisiae, about 70% of the genes that aredifferentially expressed in hypoxia require a functioning mitochondrion (104). It is not known

300 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

how the changes in gene expression are regulated. However, yeast mitochondria produce nitricoxide (NO) from nitrite in low-oxygen conditions, through the activity of cytochrome c oxidase(23). NO is required for induction of CYC7, the hypoxic isoform of cytochrome c (23, 76). NOdirectly affects the level of nitration of tyrosine residues and also causes an increase in the overalllevel of protein carbonylation (23).

Mitochondrial NO also plays an important role in the hypoxic response of mammalian cells (23,104). Mitochondrial function, and particularly cytochrome c, is required for stabilization of HIFα

in hypoxic conditions (91). Recent evidence suggests that stabilization requires NO producedby cytochrome c oxidase in the mitochondrion (7). Other mitochondrial reactive oxygen species(ROS) are also likely to be important for hypoxic signaling in mammalian cells and in yeast cells(48, 49).

Because HIFα is not found outside the Metazoa (Figure 1), the role of the mitochondrionin the hypoxic response for mammalian cells cannot be the same as that for fungi. However, itis likely that NO regulates the stability or activity of other proteins required for the response tohypoxia in S. cerevisiae. Deletion of cytochrome c significantly attenuates growth of A. fumigatus inhypoxic conditions, suggesting that the electron transport chain is also important for the hypoxicresponse in this species (43).

The mitochondrion is also important for the hypoxic response of C. neoformans, where mutantssensitive to CoCl2 and to hypoxia are defective in mitochondrial function (65). CoCl2 is commonlyused as a mimic of hypoxia in mammalian cells, where it stabilizes HIFα by regulating the activityof prolyl hydroxylases (116). In C. neoformans and S. pombe, CoCl2 targets the sterol synthesispathway, probably by replacing iron in Erg25 and Erg23 enzymes (84). Some of the C. neoformansmitochondrial mutants produce increased levels of ROS, supporting the hypothesis that ROS maybe used as an oxygen-sensing mechanism in many fungi.

REGULATION OF STEROL, LIPID, AND IRON METABOLISM GENES

Sterols and lipids are essential for membrane fluidity, and levels are therefore carefully controlled.Induction of sterol synthesis gene expression is one of the most conserved responses to hypoxiain fungi and has been reported in every species studied (e.g., 9, 16, 22, 27, 29, 62, 141). Genesrequired for the oxygen-dependent steps are particularly upregulated (9, 127). Regulation isassociated most closely with activation of SREBPs (in basidiomycetes and many ascomycetes)or with members of the Upc2 family, as described above. However, other mechanisms mayalso be used. For example, in A. fumigatus, the expression of genes required for the early stepsin ergosterol synthesis (terpenoid synthesis) is reduced in hypoxia, leading to a decrease insubstrates such as mevalonate. Barker et al. (9) have suggested that there may be a direct linkbetween hypoxic regulation of terpenoid biosynthesis and ergosterol synthesis that bypassesdirect regulation by SREBP, possibly by acting through HMG-CoA reductase (108).

The expression of many other lipid biosynthesis genes is increased in low-oxygen conditions.In S. cerevisiae and K. lactis, hypoxic induction of fatty acid synthesis genes requires the Mga2transcription factor (94, 96). The expression of lipid synthesis may also be directly regulated bySREBP/Upc2 [e.g., in C. neoformans (27) and in S. pombe (62)] or other transcription factors [e.g.,Efg1 in C. albicans (119)]. However, it is likely that other mechanisms, such as a general stressresponse, are also involved (108).

The fungal response to hypoxia overlaps the response to low iron; many genes involved inheme biosynthesis and/or iron acquisition are upregulated in both conditions (9, 15, 25, 27, 119,127, 131). In A. fumigatus, iron acquisition genes are probably regulated directly by the SREBPSrbA (15). However, because the reductive transport system in species such as C. albicans requires

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 301

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

oxidation of Fe2+, it is hard to separate the direct effects of low oxygen from those of low iron(reviewed in 3). It is possible that iron is used as an oxygen sensor, distinct from its previouslydescribed role in heme-activated proteins, but this remains to be characterized (22).

REGULATION OF GLYCOLYTIC GENE EXPRESSION

Glycolysis is one of the oldest metabolic pathways, and glycolytic enzymes predate the appearanceof oxygen-requiring species by at least 2 billion years (137). In multicellular eukaryotes (includingmammalian cells), the expression of glycolytic enzymes is strongly and coordinately induced inhypoxic conditions, and it has been hypothesized that coregulation of glycolytic genes may dateback to the origins of life (137). However, the regulatory mechanisms are not conserved. Inmammalian cells, glycolytic gene expression is regulated by HIF; in fungi, it is not yet clear if there isa shared core mechanism; and in bacteria, expression is regulated by a two-component redox sensor(represented by ArcBA in Escherichia coli ) and by anaerobic activation of the FNR pathway (137).

Hypoxic induction of glycolytic genes is well characterized in S. cerevisiae (77). Similar changesin expression are observed in other ascomycetes such as C. albicans, C. parapsilosis, Pichia pastoris,S. pombe, and the aquatic Blastocladiella emersonii (5, 10, 22, 46, 111, 119, 127, 131), suggest-ing that changes in central carbon metabolism may be a shared response to hypoxia among theopisthokonts. The expression of glycolytic enzymes is also increased in A. fumigatus, at least atthe protein level (9). There are some exceptions, however, particularly among obligate aerobicspecies. For example, in Trichoderma reesei, glycolytic gene expression is repressed when oxygenlevels are drastically lowered (16), and in C. neoformans, respiratory gene expression is inducedrather than repressed when oxygen levels are low (27).

Even species with a shared hypoxic response may regulate expression in a different manner. InS. cerevisiae, glycolytic gene expression is controlled by two unrelated proteins, Gcr1 and Gcr2,in response to glucose levels (6). Both proteins are well conserved in the Saccharomyces clade andregulate glycolytic gene expression in K. lactis (98). However, Gcr1 and Gcr2 are likely to haveevolved relatively recently as they are not present in sister clades to Saccharomyces, such as theCandida clade.

The expression of glycolytic genes in S. cerevisiae also requires Tye7, which was originallyidentified through genetic interactions with Gcr1 and Gcr2 (99, 113). Tye7 is well conservedthroughout the Saccharomycotina and binds DNA through a bHLH domain similar to that inSREBPs (14). In C. albicans, Tye7 binds promoters of glycolytic genes (5). Tye7 is required forregulation of the glucose-6-phosphate flux and is a determining factor of glycolysis or glycogenmetabolism (5). Importantly, Tye7 is necessary for the hypoxic induction of glycolysis (5, 17).The prevalence of Tye7 as a hypoxic regulator is not clear, because few species have been tested.In C. albicans, for example, Gal4 acts with Tye7 to regulate glycolytic gene expression (5, 93). InS. cerevisiae, however, Gal4 controls galactose metabolic gene expression, not glycolysis (132).

Other potential hypoxic regulators of glycolysis in C. albicans have been identified. Efg1 facil-itates upregulation of glycolytic genes in hypoxia, which is particularly important during biofilmdevelopment (17, 119, 125). In C. albicans cells lacking the transcription factor ACE2, glycolyticgene expression is reduced and respiratory gene expression is increased. Ace2 is required for in-duction of filamentous growth in hypoxic conditions; however, the connection with glycolyticgene expression is unknown (97).

EXPRESSION OF CELL WALL GENES

Growth in hypoxia has dramatic effects on fungal cell walls. In S. cerevisiae, expression of theDAN/TIR family of mannoproteins is highly induced in hypoxic and anoxic conditions (28). The

302 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

Common in FungalExtracellularMembranes(CFEM): a proteinfamily

GPI: glycophos-phatidylinositol

expression of a related family of secreted seripauperin (PAU) genes is also induced (105). InC. albicans, hypoxia affects transcription and protein levels of GPI-anchored proteins and membersof the CFEM (Common in Fungal Extracellular Membranes) family (122, 127), and increasedexpression of cell wall genes is observed in A. fumigatus during hypoxic growth (9).

The regulation of cell wall gene expression is best studied in S. cerevisiae. The expression of theDAN/TIR family requires Upc2 (1) and is blocked by the addition of heme (118). Repression ofPAU family expression is mediated by heme, sterols, and Upc2 (56, 77, 105). Recently, Hickmanet al. (56) showed that Hog1 and other members of the osmotic stress pathway are required forhypoxic induction of the PAU genes. Changes in membrane fluidity (influenced by both steroland heme levels) may activate Hog1 in hypoxia, which then controls the expression of the PAUgenes via Upc2. The hypoxic role of Hog1 is distinct from its role in the stress response.

Hypoxic induction of the CFEM family of cell wall genes in C. albicans is mimicked by low-ering sterol levels with azole drugs, and it requires Upc2 and the transcription factor Bcr1 (127).However, expression of the CFEM family is apparently unaffected by hypoxia in C. parapsilosis(111). Therefore, changes in oxygen levels likely play a general role in modifying fungal cell wallstructure, but with major species-specific effects.

HYPOXIA AND PATHOGENESIS

Fungal pathogens of mammalian hosts are routinely exposed to varying oxygen levels (14, 35,44). Oxygen on the skin is at atmospheric concentrations (21% or 160 mm Hg partial pressure),whereas organs and tissues are generally hypoxic (with levels ranging from 1 to 110 mm Hg;reviewed in 35). Most infection sites are assumed to be hypoxic, even those in the lungs (43, 44).

Biofilms and Hypoxia

Communities of cells forming biofilms on indwelling medical devices are hypoxic environments(17, 111, 115, 125). Rossignol et al. (111) showed that the transcriptional response of biofilmcommunities of C. parapsilosis overlaps the response of this organism to hypoxia, with increasedexpression of lipid, ergosterol, glycolysis, and alcohol metabolism in both conditions. A similarexpression pattern was observed in cells detaching from C. albicans biofilms (115). The availabilityof oxygen does not regulate the detachment process, suggesting that biofilms respond to hypoxicenvironments and are not regulated by them (115). However, subsequent analysis (17, 125) showedthat hypoxic adaptation is important for biofilm development.

Whereas several transcriptional regulators are required for biofilm development in normoxicconditions only, others such as Efg1 and Flo8 also regulate biofilms in hypoxia (125). Efg1 isparticularly involved in the upregulation of glycolytic genes and is required for hypoxic adaptationduring biofilm development. Disrupting other regulators of glycolytic gene expression [such asTYE7 (17)] results in the formation of biofilms with reduced cohesiveness. Increased glycolyticflux is therefore likely to be important for biofilm development in Candida species. It is not clear,however, how conserved this response is in biofilms formed by other fungi (such as Cryptococcus)that do not increase glycolytic gene expression during hypoxia.

Hypoxic Regulators and Virulence

The importance of Efg1 in the hypoxic response extends beyond its role in biofilm formation.Efg1 is a member of the APSES (Asm1, Phd1, Sok2, Efg1, and StuA) family of transcriptionalregulators, which have important roles in regulating morphology in many fungal species (35).

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 303

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

C. albicans efg1 deletions are more invasive in mice transiently exposed to hypoxic conditions (70).In addition, cell-to-cell variability in EFG1 expression affects colonization of the gastrointestinaltract, and expression is influenced by the host immune system (102). Low oxygen levels in thegastrointestinal tract may influence the activity of EFG1.

In C. albicans, EFG1 is also required for the switch from yeast to hyphal growth when oxygenlevels are low (31); hyphal growth is generally associated with infection (87). Deleting EFG1 resultsin hyperfilamentous growth in hypoxic conditions (31). The transcription factor CZF1 is requiredfor filamentous growth when cells are embedded in agar where oxygen levels are low (18). Theexpression of CZF1 is regulated by Efg1 (135), and filamentation in embedded media appears toresult from modulation of the repressive role of Efg1 by Czf1 (41). Czf1 and Efg1 are componentsof a transcriptional network that regulates white-opaque switching in C. albicans (147). This switchis required for efficient mating in C. albicans, but it is also associated with pathogenesis; white cellsare more virulent in intravenous models of infection, whereas opaque cells are more virulent inskin infections (73, 74). Conversion of white to opaque cells is induced by low oxygen and highcarbon dioxide (61, 107).

Other regulators, such as Rho G protein Rac1 and its guanine exchange factor Dck1, havebeen associated with hyphal growth in hypoxic conditions (59). Hypoxic induction of filamen-tous growth requires the G protein Ras1 and the adenylate cyclase Cdc35 (127), similar to otherfilamentation pathways (81). Deleting UPC2, the major regulator of ergosterol synthesis, also abol-ishes filamentation in hypoxia, whereas strains carrying deletions of the BCR1 transcription factorare hyperfilamentous (127). Changes in the cell wall, such as alterations in sterol levels via Upc2 orcell wall proteins by Bcr1 (30, 39), may therefore be important for the hypoxic response. In addi-tion, it is difficult to separate the effect of reduced oxygen levels from effects of increases in carbondioxide, both of which are likely to occur within tissues. For example, the kinase Sch9 repressesfilamentation in hypoxic conditions but only when carbon dioxide levels are also high (126).

Although fungal pathogens likely share a common transcriptional response to hypoxia withnonpathogenic organisms, the effect of gene deletions on virulence suggests that hypoxic adap-tation is especially important for pathogenesis. For example, deleting components of the signaltransduction pathway that regulate filamentation of C. albicans in response to hypoxia (e.g., RAS1,CDC35, SCH9) and some of the transcription factors (TYE7, ACE2) greatly attenuates virulencein animal models (5, 68, 81, 86, 110). The role of regulators of sterol metabolism in pathogenesisof Candida species has not been investigated, although it is known that at least some sterol genesare required for survival and virulence (12).

The correlation between the hypoxic response and virulence is clearer in other species. Delet-ing components of the SREBP pathway reduces virulence in both C. neoformans and A. fumigatus(13, 24, 27, 140, 141). The Tco1 histidine kinase in C. neoformans is also necessary for both thehypoxic response and for virulence (27). The unfolded protein response, important for hypoxic reg-ulation in mammals (143), might also play a role in fungi. For example, Feng et al. (38) showed thatA. fumigatus strains deleted for the endoplasmic reticulum stress sensor IreA are avirulent and havereduced growth in hypoxic conditions. Finally, Grahl et al. (44) suggest that hypoxia may influencethe host response to infection with fungal pathogens, particularly because HIFα is important forcombating bacterial infection. Interestingly, C. albicans, like many bacterial pathogens, increasesthe stability of HIFα in the presence of oxygen (138).

CONCLUSIONS

Although much is known about the regulation of the hypoxic response in fungi, even more re-mains to be investigated. One fertile area is the identification of the signal that controls the

304 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

expression of glycolytic genes in hypoxia. In Candida species, for example, expression is not in-duced by exposure to sterol-lowering drugs (46, 127), suggesting that oxygen-induced changes incentral carbon metabolism are not sensed through lowered sterol levels. The role of heme proteinsand the potentially important roles of the mitochondria and the respiratory chain remain to befully explored. Other areas likely to be of future interest include the regulation of translation inhypoxia (109) and the nearly unstudied modulation of the host by fungal pathogens in hypoxicconditions.

SUMMARY POINTS

1. HIFα has evolved recently as a regulator of the hypoxic response in multicellular animals,but its partner, HIF-1β, may be conserved in some unicellular organisms (e.g., Capsasporaowczarzaki ), excluding the fungi.

2. The SREBPs are ancient regulators of sterol synthesis that were present in the ancestorof all the opisthokonts.

3. SREBPs are highly degenerate in the Saccharomyces and Candida clades, where their rolein hypoxic regulation has been replaced by the transcription factor Upc2.

4. Fungi sense low oxygen via reduced heme and sterol levels.

5. Fungi have a shared transcriptional response to reduced oxygen, which includes increasedexpression of lipid, sterol, and heme biosynthesis. Changes in central carbon metabolism(e.g., increased glycolysis, reduced respiration) are observed in some species.

6. Reduced sterol levels control cleavage and activation of SREBPs in some fungi, but bydramatically different mechanisms.

7. Adaptation to hypoxic conditions is important for biofilm formation and for virulence inhuman fungal pathogens.

FUTURE ISSUES

1. How important are the respiratory chain and the mitochondrion for regulating the hy-poxic response?

2. How are oxygen levels correlated with the expression of glycolytic genes and changes incentral carbon metabolism?

3. How important is protein hydroxylation for sensing oxygen in fungi?

4. Does hypoxia influence the immune response of the host to fungal pathogens?

DISCLOSURE STATEMENT

The author is not aware of any affiliations, memberships, funding, or financial holdings that mightbe perceived as affecting the objectivity of this review.

LITERATURE CITED

1. Abramova N, Sertil O, Mehta S, Lowry CV. 2001. Reciprocal regulation of anaerobic and aerobic cellwall mannoprotein gene expression in Saccharomyces cerevisiae. J. Bacteriol. 183:2881–87

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 305

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

2. Adams CM, Reitz J, De Brabander JK, Feramisco JD, Li L, et al. 2004. Cholesterol and 25-hydroxycholesterol inhibit activation of SREBPs by different mechanisms, both involving SCAP andINSIGs. J. Biol. Chem. 279:52772–80

3. Almeida RS, Wilson D, Hube B. 2009. Candida albicans iron acquisition within the host. FEMS Yeast Res.9:1000–12

4. Arquier N, Vigne P, Duplan E, Hsu T, Therond PP, et al. 2006. Analysis of the hypoxia-sensing pathwayin Drosophila melanogaster. Biochem. J. 393:471–80

5. Askew C, Sellam A, Epp E, Hogues H, Mullick A, et al. 2009. Transcriptional regulation of carbohydratemetabolism in the human pathogen Candida albicans. PLoS Pathog. 5:e1000612

6. Baker HV. 1991. GCR1 of Saccharomyces cerevisiae encodes a DNA binding protein whose binding isabolished by mutations in the CTTCC sequence motif. Proc. Natl. Acad. Sci. USA 88:9443–47

7. Ball KA, Nelson AW, Foster DG, Poyton RO. 2012. Nitric oxide produced by cytochrome c oxidasehelps stabilize HIF-1α in hypoxic mammalian cells. Biochem. Biophys. Res. Commun. 420:727–32

8. Bao WG, Guiard B, Fang ZA, Donnini C, Gervais M, et al. 2008. Oxygen-dependent transcriptionalregulator Hap1p limits glucose uptake by repressing the expression of the major glucose transportergene RAG1 in Kluyveromyces lactis. Eukaryot. Cell 7:1895–905

9. Barker BM, Kroll K, Vodisch M, Mazurie A, Kniemeyer O, Cramer RA. 2012. Transcriptomic andproteomic analyses of the Aspergillus fumigatus hypoxia response using an oxygen-controlled fermenter.BMC Genomics 13:62

10. Baumann K, Carnicer M, Dragosits M, Graf AB, Stadlmann J, et al. 2010. A multi-level study of recom-binant Pichia pastoris in different oxygen conditions. BMC Syst. Biol. 4:141

11. Becerra M, Lombardia-Ferreira LJ, Hauser NC, Hoheisel JD, Tizon B, Cerdan ME. 2002. The yeasttranscriptome in aerobic and hypoxic conditions: effects of hap1, rox1, rox3 and srb10 deletions. Mol.Microbiol. 43:545–55

12. Becker JM, Kauffman SJ, Hauser M, Huang L, Lin M, et al. 2010. Pathway analysis of Candida albicanssurvival and virulence determinants in a murine infection model. Proc. Natl. Acad. Sci. USA 107:22044–49

13. Bien CM, Chang YC, Nes WD, Kwon-Chung KJ, Espenshade PJ. 2009. Cryptococcus neoformans Site-2protease is required for virulence and survival in the presence of azole drugs. Mol. Microbiol. 74:672–90

14. Bien CM, Espenshade PJ. 2010. Sterol regulatory element binding proteins in fungi: hypoxic transcrip-tion factors linked to pathogenesis. Eukaryot. Cell 9:352–59

15. Blatzer M, Barker BM, Willger SD, Beckmann N, Blosser SJ, et al. 2011. SREBP coordinates iron andergosterol homeostasis to mediate triazole drug and hypoxia responses in the human fungal pathogenAspergillus fumigatus. PLoS Genet. e1002374

16. Bonaccorsi ED, Ferreira AJ, Chambergo FS, Ramos AS, Mantovani MC, et al. 2006. Transcriptionalresponse of the obligatory aerobe Trichoderma reesei to hypoxia and transient anoxia: implications forenergy production and survival in the absence of oxygen. Biochemistry 45:3912–24

17. Bonhomme J, Chauvel M, Goyard S, Roux P, Rossignol T, d’Enfert C. 2011. Contribution of theglycolytic flux and hypoxia adaptation to efficient biofilm formation by Candida albicans. Mol. Microbiol.80:995–1013

18. Brown DH Jr, Giusani AD, Chen X, Kumamoto CA. 1999. Filamentous growth of Candida albicans inresponse to physical environmental cues and its regulation by the unique CZF1 gene. Mol. Microbiol.34:651–62

19. Bunn HF, Poyton RO. 1996. Oxygen sensing and molecular adaptation to hypoxia. Physiol. Rev. 76:839–85

20. Burg JS, Espenshade PJ. 2011. Regulation of HMG-CoA reductase in mammals and yeast. Prog. LipidRes. 50:403–10

21. Burg JS, Powell DW, Chai R, Hughes AL, Link AJ, Espenshade PJ. 2008. Insig regulates HMG-CoAreductase by controlling enzyme phosphorylation in fission yeast. Cell. Metab. 8:522–31

22. Camilo CM, Gomes SL. 2010. Transcriptional response to hypoxia in the aquatic fungus Blastocladiellaemersonii. Eukaryot. Cell 9:915–25

23. Castello PR, David PS, McClure T, Crook Z, Poyton RO. 2006. Mitochondrial cytochrome oxidaseproduces nitric oxide under hypoxic conditions: implications for oxygen sensing and hypoxic signalingin eukaryotes. Cell. Metab. 3:277–87

306 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

24. Chang YC, Bien CM, Lee H, Espenshade PJ, Kwon-Chung KJ. 2007. Sre1p, a regulator of oxygen sensingand sterol homeostasis, is required for virulence in Cryptococcus neoformans. Mol. Microbiol. 64:614–29

25. Chang YC, Ingavale SS, Bien C, Espenshade P, Kwon-Chung KJ. 2009. Conservation of the sterol reg-ulatory element-binding protein pathway and its pathobiological importance in Cryptococcus neoformans.Eukaryot. Cell 8:1770–79

26. Chodosh LA, Olesen J, Hahn S, Baldwin AS, Guarente L, Sharp PA. 1988. A yeast and a human CCAAT-binding protein have heterologous subunits that are functionally interchangeable. Cell 53:25–35

27. Chun CD, Liu OW, Madhani HD. 2007. A link between virulence and homeostatic responses to hypoxiaduring infection by the human fungal pathogen Cryptococcus neoformans. PLoS Pathog. 3:e22

28. Cohen BD, Sertil O, Abramova NE, Davies KJ, Lowry CV. 2001. Induction and repression of DAN1and the family of anaerobic mannoprotein genes in Saccharomyces cerevisiae occurs through a complexarray of regulatory sites. Nucleic Acids Res. 29:799–808

29. Davies BS, Rine J. 2006. A role for sterol levels in oxygen sensing in Saccharomyces cerevisiae. Genetics174:191–201

30. Ding C, Vidanes GM, Maguire SL, Guida A, Synnott JM, et al. 2011. Conserved and divergent roles ofBcr1 and CFEM proteins in Candida parapsilosis and Candida albicans. PLoS One 6:e28151

31. Doedt T, Krishnamurthy S, Bockmuhl DP, Tebarth B, Stempel C, et al. 2004. APSES proteins regulatemorphogenesis and metabolism in Candida albicans. Mol. Biol. Cell 15:3167–80

32. Duncan EA, Brown MS, Goldstein JL, Sakai J. 1997. Cleavage site for sterol-regulated protease localizedto a Leu-Ser bond in the lumenal loop of sterol regulatory element-binding protein-2. J. Biol. Chem.272:12778–85

33. Duncan EA, Dave UP, Sakai J, Goldstein JL, Brown MS. 1998. Second-site cleavage in sterol regulatoryelement-binding protein occurs at transmembrane junction as determined by cysteine panning. J. Biol.Chem. 273:17801–9

34. Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O’Rourke J, et al. 2001. C. elegans EGL-9 andmammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell107:43–54

35. Ernst JF, Tielker D. 2008. Responses to hypoxia in fungal pathogens. Cell. Microbiol. 11:183–9036. Espenshade PJ, Hughes AL. 2007. Regulation of sterol synthesis in eukaryotes. Annu. Rev. Genet. 41:401–

2737. Fang ZA, Wang GH, Chen AL, Li YF, Liu JP, et al. 2009. Gene responses to oxygen availability in

Kluyveromyces lactis: an insight on the evolution of the oxygen-responding system in yeast. PLoS One4:e7561

38. Feng X, Krishnan K, Richie DL, Aimanianda V, Hartl L, et al. 2011. HacA-independent functions ofthe ER stress sensor IreA synergize with the canonical UPR to influence virulence traits in Aspergillusfumigatus. PLoS Pathog. 7:e1002330

39. Finkel JS, Xu W, Huang D, Hill EM, Desai JV, et al. 2012. Portrait of Candida albicans adherenceregulators. PLoS Pathog. 8:e1002525

40. Flury I, Garza R, Shearer A, Rosen J, Cronin S, Hampton RY. 2005. INSIG: a broadly conservedtransmembrane chaperone for sterol-sensing domain proteins. EMBO J. 24:3917–26

41. Giusani AD, Vinces M, Kumamoto CA. 2002. Invasive filamentous growth of Candida albicans is promotedby Czf1p-dependent relief of Efg1p-mediated repression. Genetics 160:1749–53

42. Gleason FH, Schmidt SK, Marano AV. 2010. Can zoosporic true fungi grow or survive in extreme orstressful environments? Extremophiles 14:417–25

43. Grahl N, Dinamarco TM, Willger SD, Goldman GH, Cramer RA. 2012. Aspergillus fumigatus mito-chondrial electron transport chain mediates oxidative stress homeostasis, hypoxia responses and fungalpathogenesis. Mol. Microbiol. 84:383–99

44. Grahl N, Shepardson KM, Chung D, Cramer RA. 2012. Hypoxia and fungal pathogenesis: to air or notto air? Eukaryot. Cell 11:560–70

45. Greer SN, Metcalf JL, Wang Y, Ohh M. 2012. The updated biology of hypoxia-inducible factor. EMBOJ. 31:2448–60

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 307

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

46. Guida A, Lindstadt C, Maguire SL, Ding C, Higgins DG, et al. 2011. Using RNA-seq to determinethe transcriptional landscape and the hypoxic response of the pathogenic yeast Candida parapsilosis. BMCGenomics 12:628

47. Guillemin K, Krasnow MA. 1997. The hypoxic response: huffing and HIFing. Cell 89:9–1248. Guzy RD, Mack MM, Schumacker PT. 2007. Mitochondrial complex III is required for hypoxia-induced

ROS production and gene transcription in yeast. Antioxid. Redox Signal. 9:1317–2849. Hamanaka RB, Chandel NS. 2009. Mitochondrial reactive oxygen species regulate hypoxic signaling.

Curr. Opin. Cell Biol. 21:894–9950. Hampton-Smith RJ, Peet DJ. 2009. From polyps to people: a highly familiar response to hypoxia. Ann.

N. Y. Acad. Sci. 1177:19–2951. Heckman DS, Geiser DM, Eidell BR, Stauffer RL, Kardos NL, Hedges SB. 2001. Molecular evidence

for the early colonization of land by fungi and plants. Science 293:1129–3352. Hedges SB, Blair JE, Venturi ML, Shoe JL. 2004. A molecular timescale of eukaryote evolution and the

rise of complex multicellular life. BMC Evol. Biol. 4:253. Henri J, Rispal D, Bayart E, van Tilbeurgh H, Seraphin B, Graille M. 2010. Structural and functional in-

sights into Saccharomyces cerevisiae Tpa1, a putative prolylhydroxylase influencing translation terminationand transcription. J. Biol. Chem. 285:30767–78

54. Hertel LA, Bayne CJ, Loker ES. 2002. The symbiont Capsaspora owczarzaki, nov. gen. nov. sp., isolatedfrom three strains of the pulmonate snail Biomphalaria glabrata is related to members of the Mesomyce-tozoea. Int. J. Parasitol. 32:1183–91

55. Hibbett DS, Binder M, Bischoff JF, Blackwell M, Cannon PF, et al. 2007. A higher-level phylogeneticclassification of the Fungi. Mycol. Res. 111:509–47

56. Hickman MJ, Spatt D, Winston F. 2011. The Hog1 mitogen-activated protein kinase mediates a hypoxicresponse in Saccharomyces cerevisiae. Genetics 188:325–38

57. Hodge MR, Kim G, Singh K, Cumsky MG. 1989. Inverse regulation of the yeast COX5 genes by oxygenand heme. Mol. Cell. Biol. 9:1958–64

58. Hoot SJ, Oliver BG, White TC. 2008. Candida albicans UPC2 is transcriptionally induced in response toantifungal drugs and anaerobicity through Upc2p-dependent and -independent mechanisms. Microbiology154:2748–56

59. Hope H, Bogliolo S, Arkowitz RA, Bassilana M. 2008. Activation of Rac1 by the guanine nucleotideexchange factor Dck1 is required for invasive filamentous growth in the pathogen Candida albicans. Mol.Biol. Cell 19:3638–51

60. Horton JD, Goldstein JL, Brown MS. 2002. SREBPs: activators of the complete program of cholesteroland fatty acid synthesis in the liver. J. Clin. Invest. 109:1125–31

61. Huang G, Srikantha T, Sahni N, Yi S, Soll DR. 2009. CO2 regulates white-to-opaque switching inCandida albicans. Curr. Biol. 19:330–34

62. First demonstrationthat SREBPs areimportant regulators ofthe hypoxic response infungi.

62. Hughes AL, Todd BL, Espenshade PJ. 2005. SREBP pathway responds to sterols and functionsas an oxygen sensor in fission yeast. Cell 120:831–42

63. Hughes BT, Espenshade PJ. 2008. Oxygen-regulated degradation of fission yeast SREBP by Ofd1, aprolyl hydroxylase family member. EMBO J. 27:1491–501

64. Hughes BT, Nwosu CC, Espenshade PJ. 2009. Degradation of sterol regulatory element-binding proteinprecursor requires the endoplasmic reticulum-associated degradation components Ubc7 and Hrd1 infission yeast. J. Biol. Chem. 284:20512–21

65. Implicatesmitochondria in thehypoxic response ofC. neoformans.

65. Ingavale SS, Chang YC, Lee H, McClelland CM, Leong ML, Kwon-Chung KJ. 2008. Importanceof mitochondria in survival of Cryptococcus neoformans under low oxygen conditions and toleranceto cobalt chloride. PLoS Pathog. 4:e1000155

66. Kadosh D, Johnson AD. 2005. Induction of the Candida albicans filamentous growth program by reliefof transcriptional repression: a genome-wide analysis. Mol. Biol. Cell 16:2903–12

67. Keeling KM, Salas-Marco J, Osherovich LZ, Bedwell DM. 2006. Tpa1p is part of an mRNP complexthat influences translation termination, mRNA deadenylation, and mRNA turnover in Saccharomycescerevisiae. Mol. Cell. Biol. 26:5237–48

68. Kelly MT, MacCallum DM, Clancy SD, Odds FC, Brown AJ, Butler G. 2004. The Candida albicansCaACE2 gene affects morphogenesis, adherence and virulence. Mol. Microbiol. 53:969–83

308 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

69. Khalaf RA, Zitomer RS. 2001. The DNA binding protein Rfg1 is a repressor of filamentation in Candidaalbicans. Genetics 157:1503–12

70. Kim AS, Garni RM, Henry-Stanley MJ, Bendel CM, Erlandsen SL, Wells CL. 2003. Hypoxia andextraintestinal dissemination of Candida albicans yeast forms. Shock 19:257–62

71. Kim JB, Spotts GD, Halvorsen YD, Shih HM, Ellenberger T, et al. 1995. Dual DNA binding specificityof ADD1/SREBP1 controlled by a single amino acid in the basic helix-loop-helix domain. Mol. Cell. Biol.15:2582–88

72. Krogan NJ, Cagney G, Yu H, Zhong G, Guo X, et al. 2006. Global landscape of protein complexes inthe yeast Saccharomyces cerevisiae. Nature 440:637–43

73. Kvaal C, Lachke SA, Srikantha T, Daniels K, McCoy J, Soll DR. 1999. Misexpression of the opaque-phase-specific gene PEP1 (SAP1) in the white phase of Candida albicans confers increased virulence in amouse model of cutaneous infection. Infect. Immun. 67:6652–62

74. Kvaal CA, Srikantha T, Soll DR. 1997. Misexpression of the white-phase-specific gene WH11 in theopaque phase of Candida albicans affects switching and virulence. Infect. Immun. 65:4468–75

75. Kwast KE, Burke PV, Poyton RO. 1998. Oxygen sensing and the transcriptional regulation of oxygen-responsive genes in yeast. J. Exp. Biol. 201:1177–95

76. First demonstrationthat the respiratorychain is important forthe hypoxic response infungi.

76. Kwast KE, Burke PV, Staahl BT, Poyton RO. 1999. Oxygen sensing in yeast: evidence for theinvolvement of the respiratory chain in regulating the transcription of a subset of hypoxic genes.Proc. Natl. Acad. Sci. USA 96:5446–51

77. Kwast KE, Lai LC, Menda N, James DT 3rd, Aref S, Burke PV. 2002. Genomic analyses of anaerobicallyinduced genes in Saccharomyces cerevisiae: functional roles of Rox1 and other factors in mediating theanoxic response. J. Bacteriol. 184:250–65

78. Lamas-Maceiras M, Nunez L, Rodriguez-Belmonte E, Gonzalez-Siso MI, Cerdan ME. 2007. Functionalcharacterization of KlHAP1: a model to foresee different mechanisms of transcriptional regulation byHap1p in yeasts. Gene 405:96–107

79. Lando D, Balmer J, Laue ED, Kouzarides T. 2012. The S. pombe histone H2A dioxygenase Ofd2 regulatesgene expression during hypoxia. PLoS One 7:e29765

80. Lane S, Zhou S, Pan T, Dai Q, Liu H. 2001. The basic helix-loop-helix transcription factor Cph2regulates hyphal development in Candida albicans partly via TEC1. Mol. Cell. Biol. 21:6418–28

81. Leberer E, Harcus D, Dignard D, Johnson L, Ushinsky S, et al. 2001. Ras links cellular morphogenesisto virulence by regulation of the MAP kinase and cAMP signalling pathways in the pathogenic fungusCandida albicans. Mol. Microbiol. 42:673–87

82. Lee CY, Stewart EV, Hughes BT, Espenshade PJ. 2009. Oxygen-dependent binding of Nro1 to theprolyl hydroxylase Ofd1 regulates SREBP degradation in yeast. EMBO J. 28:135–43

83. First demonstrationof the role of oxygen inregulating DNA bindingof SREBPs.

83. Lee CY, Yeh TL, Hughes BT, Espenshade PJ. 2011. Regulation of the Sre1 hypoxic transcriptionfactor by oxygen-dependent control of DNA binding. Mol. Cell 44:225–34

84. Lee H, Bien CM, Hughes AL, Espenshade PJ, Kwon-Chung KJ, Chang YC. 2007. Cobalt chloride, ahypoxia-mimicking agent, targets sterol synthesis in the pathogenic fungus Cryptococcus neoformans. Mol.Microbiol. 65:1018–33

85. Lee MC, Miller EA, Goldberg J, Orci L, Schekman R. 2004. Bi-directional protein transport betweenthe ER and Golgi. Annu. Rev. Cell Dev. Biol. 20:87–123

86. Liu W, Zhao J, Li X, Li Y, Jiang L. 2010. The protein kinase CaSch9p is required for the cell growth,filamentation and virulence in the human fungal pathogen Candida albicans. FEMS Yeast Res. 10:462–70

87. Lo HJ, Kohler JR, DiDomenico B, Loebenberg D, Cacciapuoti A, Fink GR. 1997. NonfilamentousC. albicans mutants are avirulent. Cell 90:939–49

88. Loenarz C, Coleman ML, Boleininger A, Schierwater B, Holland PW, et al. 2011. The hypoxia-inducibletranscription factor pathway regulates oxygen sensing in the simplest animal, Trichoplax adhaerens. EMBORep. 12:63–70

89. Lorenz MC, Heitman J. 1998. Regulators of pseudohyphal differentiation in Saccharomyces cerevisiae iden-tified through multicopy suppressor analysis in ammonium permease mutant strains. Genetics 150:1443–57

90. Lowry CV, Zitomer RS. 1988. ROX1 encodes a heme-induced repression factor regulating ANB1 andCYC7 of Saccharomyces cerevisiae. Mol. Cell. Biol. 8:4651–58

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 309

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

91. Mansfield KD, Guzy RD, Pan Y, Young RM, Cash TP, et al. 2005. Mitochondrial dysfunction resultingfrom loss of cytochrome c impairs cellular oxygen sensing and hypoxic HIF-α activation. Cell. Metab.1:393–99

92. Marie C, Leyde S, White TC. 2008. Cytoplasmic localization of sterol transcription factors Upc2p andEcm22p in S. cerevisiae. Fungal Genet. Biol. 45:1430–38

93. Martchenko M, Levitin A, Hogues H, Nantel A, Whiteway M. 2007. Transcriptional rewiring of fungalgalactose-metabolism circuitry. Curr. Biol. 17:1007–13

94. Martin CE, Oh CS, Jiang Y. 2006. Regulation of long chain unsaturated fatty acid synthesis in yeast.Biochim. Biophys. Acta 1771:271–85

95. Marziali G, Perrotti E, Ilari R, Testa U, Coccia EM, Battistini A. 1997. Transcriptional regulation of theferritin heavy-chain gene: the activity of the CCAAT binding factor NF-Y is modulated in heme-treatedFriend leukemia cells and during monocyte-to-macrophage differentiation. Mol. Cell. Biol. 17:1387–95

96. Micolonghi C, Ottaviano, Di Silvio E, Damata G, Heipieper HJ, Bianchi MM. 2012. A dual signallingpathway for the hypoxic expression of lipid genes, dependent on the glucose sensor Rag4, is revealed bythe analysis of the KlMGA2 gene in Kluyveromyces lactis. Microbiology 158:1734–44

97. Mulhern SM, Logue ME, Butler G. 2006. The Candida albicans transcription factor Ace2 regulatesmetabolism and is required for filamentation in hypoxic conditions. Eukaryot. Cell 5:2001–13

98. Neil H, Lemaire M, Wesolowski-Louvel M. 2004. Regulation of glycolysis in Kluyveromyces lactis: roleof KlGCR1 and KlGCR2 in glucose uptake and catabolism. Curr. Genet. 45:129–39

99. Nishi K, Park CS, Pepper AE, Eichinger G, Innis MA, Holland MJ. 1995. The GCR1 requirement foryeast glycolytic gene expression is suppressed by dominant mutations in the SGC1 gene, which encodesa novel basic-helix-loop-helix protein. Mol. Cell. Biol. 15:2646–53

100. Olesen JT, Guarente L. 1990. The HAP2 subunit of yeast CCAAT transcriptional activator containsadjacent domains for subunit association and DNA recognition: model for the HAP2/3/4 complex. GenesDev. 4:1714–29

101. Osborne TF, Espenshade PJ. 2009. Evolutionary conservation and adaptation in the mechanism thatregulates SREBP action: What a long, strange tRIP it’s been. Genes Dev. 23:2578–91

102. Pierce JV, Kumamoto CA. 2012. Variation in Candida albicans EFG1 expression enables host-dependentchanges in colonizing fungal populations. MBio 3:e00117–12

103. Elegant dissectionof the contributions ofvarious mechanisms ofregulating SREBPs inS. pombe

103. Porter JR, Lee CY, Espenshade PJ, Iglesias PA. 2012. Regulation of SREBP during hypoxiarequires Ofd1-mediated control of both DNA binding and degradation. Mol. Biol. Cell 23:3764–74

104. Poyton RO, Castello PR, Ball KA, Woo DK, Pan N. 2009. Mitochondria and hypoxic signaling: a newview. Ann. N. Y. Acad. Sci. 1177:48–56

105. Rachidi N, Martinez MJ, Barre P, Blondin B. 2000. Saccharomyces cerevisiae PAU genes are induced byanaerobiosis. Mol. Microbiol. 35:1421–30

106. Radhakrishnan A, Ikeda Y, Kwon HJ, Brown MS, Goldstein JL. 2007. Sterol-regulated transport ofSREBPs from endoplasmic reticulum to Golgi: oxysterols block transport by binding to Insig. Proc. Natl.Acad. Sci. USA 104:6511–18

107. Ramirez-Zavala B, Reuss O, Park YN, Ohlsen K, Morschhauser J. 2008. Environmental induction ofwhite-opaque switching in Candida albicans. PLoS Pathog. 4:e1000089

108. Raychaudhuri S, Young BP, Espenshade PJ, Loewen CJR. 2012. Regulation of lipid metabolism: a taleof two yeasts. Curr. Opin. Cell Biol. 24:502–8

109. Rispal D, Henri J, van Tilbeurgh H, Graille M, Seraphin B. 2011. Structural and functional analysis ofNro1/Ett1: a protein involved in translation termination in S. cerevisiae and in O2-mediated gene controlin S. pombe. RNA 17:1213–24

110. Rocha CR, Schroppel K, Harcus D, Marcil A, Dignard D, et al. 2001. Signaling through adenylylcyclase is essential for hyphal growth and virulence in the pathogenic fungus Candida albicans. Mol. Biol.Cell 12:3631–43

111. Rossignol T, Ding C, Guida A, d’Enfert C, Higgins DG, Butler G. 2009. Correlation between biofilmformation and the hypoxic response in Candida parapsilosis. Eukaryot. Cell 8:550–59

112. Ruiz-Trillo I, Burger G, Holland PW, King N, Lang BF, et al. 2007. The origins of multicellularity: amulti-taxon genome initiative. Trends Genet. 23:113–18

310 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

113. Sato T, Lopez MC, Sugioka S, Jigami Y, Baker HV, Uemura H. 1999. The E-box DNA binding proteinSgc1p suppresses the gcr2 mutation, which is involved in transcriptional activation of glycolytic genes inSaccharomyces cerevisiae. FEBS Lett. 463:307–11

114. Seidah NG, Chretien M, Day R. 1994. The family of subtilisin/kexin like pro-protein and pro-hormoneconvertases: divergent or shared functions. Biochimie 76:197–209

115. Characterization ofbiofilm detachmentover time.

115. Sellam A, Al-Niemi T, McInnerney K, Brumfield S, Nantel A, Suci PA. 2009. A Candida albicansearly stage biofilm detachment event in rich medium. BMC Microbiol. 9:25

116. Semenza GL. 2004. Hydroxylation of HIF-1: oxygen sensing at the molecular level. Physiology 19:176–82117. Semenza GL. 2007. Life with oxygen. Science 318:62–64118. Sertil O, Cohen BD, Davies KJ, Lowry CV. 1997. The DAN1 gene of S. cerevisiae is regulated in parallel

with the hypoxic genes, but by a different mechanism. Gene 192:199–205119. Setiadi ER, Doedt T, Cottier F, Noffz C, Ernst JF. 2006. Transcriptional response of Candida albicans

to hypoxia: linkage of oxygen sensing and Efg1p-regulatory networks. J. Mol. Biol. 361:399–411120. Sever N, Yang T, Brown MS, Goldstein JL, DeBose-Boyd RA. 2003. Accelerated degradation of HMG

CoA reductase mediated by binding of Insig-1 to its sterol-sensing domain. Mol. Cell 11:2–33121. Slot JC, Rokas A. 2010. Multiple GAL pathway gene clusters evolved independently and by different

mechanisms in fungi. Proc. Natl. Acad. Sci. USA 107:10136–41122. Sosinska GJ, de Groot PW, Teixeira de Mattos MJ, Dekker HL, de Koster CG, et al. 2008. Hypoxic

conditions and iron restriction affect the cell-wall proteome of Candida albicans grown under vagina-simulative conditions. Microbiology 154:510–20

123. Stewart EV, Lloyd SJ, Burg JS, Nwosu CC, Lintner RE, et al. 2012. Yeast sterol regulatory element-binding protein (SREBP) cleavage requires Cdc48 and Dsc5, a ubiquitin regulatory X domain-containingsubunit of the Golgi Dsc E3 ligase. J. Biol. Chem. 287:672–81

124. Characterization ofthe role of theproteasome inregulating cleavage ofSREBPs in S. pombe(see also 123).

124. Stewart EV, Nwosu CC, Tong Z, Roguev A, Cummins TD, et al. 2011. Yeast SREBP cleavageactivation requires the Golgi Dsc E3 ligase complex. Mol. Cell 42:160–71

125. Stichternoth C, Ernst JF. 2009. Hypoxic adaptation by Efg1 regulates biofilm formation of Candidaalbicans. Appl. Environ. Microbiol. 75:3663–72

126. Stichternoth C, Fraund A, Setiadi E, Giasson L, Vecchiarelli A, Ernst JF. 2011. Sch9 kinase integrateshypoxia and CO2 sensing to suppress hyphal morphogenesis in Candida albicans. Eukaryot. Cell 10:502–11

127. Synnott JM, Guida A, Mulhern-Haughey S, Higgins DG, Butler G. 2010. Regulation of the hypoxicresponse in Candida albicans. Eukaryot. Cell 9:1734–46

128. Taylor BL, Zhulin IB. 1999. PAS domains: internal sensors of oxygen, redox potential, and light.Microbiol. Mol. Biol. Rev. 63:479–506

129. Taylor CT. 2008. Mitochondria and cellular oxygen sensing in the HIF pathway. Biochem. J. 409:19–26130. Taylor CT, McElwain JC. 2010. Ancient atmospheres and the evolution of oxygen sensing via the

hypoxia-inducible factor in metazoans. Physiology 25:272–79131. Todd BL, Stewart EV, Burg JS, Hughes AL, Espenshade PJ. 2006. Sterol regulatory element binding

protein is a principal regulator of anaerobic gene expression in fission yeast. Mol. Cell. Biol. 26:2817–31132. Traven A, Jelicic B, Sopta M. 2006. Yeast Gal4: a transcriptional paradigm revisited. EMBO Rep. 7:496–99133. Trinci APJ, Davies DR, Gull K, Lawrence M, Nielsen BB, et al. 1994. Anaerobic fungi in herbivorous

animals. Mycol. Res. 98:129–52134. Vik A, Rine J. 2001. Upc2p and Ecm22p, dual regulators of sterol biosynthesis in Saccharomyces cerevisiae.

Mol. Cell. Biol. 21:6395–405135. Vinces MD, Haas C, Kumamoto CA. 2006. Expression of the Candida albicans morphogenesis regulator

gene CZF1 and its regulation by Efg1p and Czf1p. Eukaryot. Cell 5:825–35136. Wang W, Wang X, Liu J, Ishii M, Igarashi Y, Cui Z. 2007. Effect of oxygen concentration on the

composting process and maturity. Compost Sci. Util. 15:184–90137. Webster KA. 2003. Evolution of the coordinate regulation of glycolytic enzyme genes by hypoxia.

J. Exp. Biol. 206:2911–22

138. Suggests that fungiand bacteria modulatestability of human HIFduring infection.138. Werth N, Beerlage C, Rosenberger C, Yazdi AS, Edelmann M, et al. 2010. Activation of hypoxia

inducible factor 1 is a general phenomenon in infections with human pathogens. PLoS One5:e11576

www.annualreviews.org • Hypoxic Expression in Eukaryotic Microbes 311

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67CH14-Butler ARI 5 August 2013 17:3

139. Whitelaw M, Pongratz I, Wilhelmsson A, Gustafsson JA, Poellinger L. 1993. Ligand-dependent re-cruitment of the Arnt coregulator determines DNA recognition by the dioxin receptor. Mol. Cell. Biol.13:2504–14

140. Confirms thatcleavage of SREBPs bythe proteasome isconserved in someAscomycota species.

140. Willger SD, Cornish EJ, Chung D, Fleming BA, Lehmann MM, et al. 2012. Dsc orthologsare required for hypoxia adaptation, triazole drug responses and fungal virulence in Aspergillusfumigatus. Eukaryot. Cell 11:1557–67

141. Demonstrates thatfungal SREBPs areimportant for virulence.

141. Willger SD, Puttikamonkul S, Kim KH, Burritt JB, Grahl N, et al. 2008. A sterol-regulatoryelement binding protein is required for cell polarity, hypoxia adaptation, azole drug resistance,and virulence in Aspergillus fumigatus. PLoS Pathog. 4:e1000200

142. Wolfe KH, Shields DC. 1997. Molecular evidence for an ancient duplication of the entire yeast genome.Nature 387:708–13

143. Wouters BG, Koritzinsky M. 2008. Hypoxia signalling through mTOR and the unfolded protein re-sponse in cancer. Nat. Rev. Cancer 8:851–64

144. Yeh TL, Lee CY, Amzel LM, Espenshade PJ, Bianchet MA. 2011. The hypoxic regulator of sterolsynthesis Nro1 is a nuclear import adaptor. Structure 19:503–14

145. Zhang L, Guarente L. 1994. HAP1 is nuclear but is bound to a cellular factor in the absence of heme.J. Biol. Chem. 269:14643–47

146. Zitomer RS, Lowry CV. 1992. Regulation of gene expression by oxygen in Saccharomyces cerevisiae.Microbiol. Rev. 56:1–11

147. Zordan RE, Miller MG, Galgoczy DJ, Tuch BB, Johnson AD. 2007. Interlocking transcriptional feedbackloops control white-opaque switching in Candida albicans. PLoS Biol. 5:e256

312 Butler

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67-FrontMatter ARI 12 August 2013 12:26

Annual Review ofMicrobiology

Volume 67, 2013 Contents

Fifty Years Fused to LacJonathan Beckwith � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 1

3′ Cap-Independent Translation Enhancers of Plant VirusesAnne E. Simon and W. Allen Miller � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �21

Acyl-Homoserine Lactone Quorum Sensing: From Evolutionto ApplicationMartin Schuster, D. Joseph Sexton, Stephen P. Diggle, and E. Peter Greenberg � � � � � � � � �43

Mechanisms of Acid Resistance in Escherichia coliUsheer Kanjee and Walid A. Houry � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �65

The Biology of the PmrA/PmrB Two-Component System: The MajorRegulator of Lipopolysaccharide ModificationsH. Deborah Chen and Eduardo A. Groisman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � �83

Transcription Regulation at the Core: Similarities Among Bacterial,Archaeal, and Eukaryotic RNA PolymerasesKimberly B. Decker and Deborah M. Hinton � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 113

Bacterial Responses to Reactive Chlorine SpeciesMichael J. Gray, Wei-Yun Wholey, and Ursula Jakob � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 141

It Takes a Village: Ecological and Fitness Impactsof Multipartite MutualismElizabeth A. Hussa and Heidi Goodrich-Blair � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 161

Electrophysiology of BacteriaAnne H. Delcour � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 179

Microbial Contributions to Phosphorus Cycling in Eutrophic Lakesand WastewaterKatherine D. McMahon and Emily K. Read � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 199

Structure and Operation of Bacterial Tripartite PumpsPhilip Hinchliffe, Martyn F. Symmons, Colin Hughes, and Vassilis Koronakis � � � � � � � � � 221

Plasmodium Nesting: Remaking the Erythrocyte from the Inside OutJustin A. Boddey and Alan F. Cowman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 243

vi

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67-FrontMatter ARI 12 August 2013 12:26

The Algal Past and Parasite Present of the ApicoplastGiel G. van Dooren and Boris Striepen � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 271

Hypoxia and Gene Expression in Eukaryotic MicrobesGeraldine Butler � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 291

Wall Teichoic Acids of Gram-Positive BacteriaStephanie Brown, John P. Santa Maria Jr., and Suzanne Walker � � � � � � � � � � � � � � � � � � � � � � 313

Archaeal Biofilms: The Great UnexploredAlvaro Orell, Sabrina Frols, and Sonja-Verena Albers � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 337

An Inquiry into the Molecular Basis of HSV Latency and ReactivationBernard Roizman and Richard J. Whitley � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 355

Molecular Bacteria-Fungi Interactions: Effects on Environment, Food,and MedicineKirstin Scherlach, Katharina Graupner, and Christian Hertweck � � � � � � � � � � � � � � � � � � � � � � � 375

Fusarium PathogenomicsLi-Jun Ma, David M. Geiser, Robert H. Proctor, Alejandro P. Rooney,

Kerry O’Donnell, Frances Trail, Donald M. Gardiner, John M. Manners,and Kemal Kazan � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 399

Biological Consequences and Advantages of AsymmetricBacterial GrowthDavid T. Kysela, Pamela J.B. Brown, Kerwyn Casey Huang, and Yves V. Brun � � � � � � � 417

Archaea in Biogeochemical CyclesPierre Offre, Anja Spang, and Christa Schleper � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 437

Experimental Approaches for Defining Functional Roles of Microbesin the Human GutGautam Dantas, Morten O.A. Sommer, Patrick H. Degnan,

and Andrew L. Goodman � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 459

Plant Cell Wall Deconstruction by Ascomycete FungiN. Louise Glass, Monika Schmoll, Jamie H.D. Cate, and Samuel Coradetti � � � � � � � � � � � � 477

Cnidarian-Microbe Interactions and the Origin of InnateImmunity in MetazoansThomas C.G. Bosch � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 499

On the Biological Success of VirusesBrian R. Wasik and Paul E. Turner � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 519

Prions and the Potential Transmissibility of ProteinMisfolding DiseasesAllison Kraus, Bradley R. Groveman, and Byron Caughey � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 543

Contents vii

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.

MI67-FrontMatter ARI 12 August 2013 12:26

The Wonderful World of Archaeal VirusesDavid Prangishvili � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 565

Tip Growth in Filamentous Fungi: A Road Trip to the ApexMeritxell Riquelme � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 587

A Paradigm for Endosymbiotic Life: Cell Differentiation of RhizobiumBacteria Provoked by Host Plant FactorsEva Kondorosi, Peter Mergaert, and Attila Kereszt � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 611

Neutrophils Versus Staphylococcus aureus: A Biological Tug of WarAndras N. Spaan, Bas G.J. Surewaard, Reindert Nijland,

and Jos A.G. van Strijp � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � � 629

Index

Cumulative Index of Contributing Authors, Volumes 63–67 � � � � � � � � � � � � � � � � � � � � � � � � � � � 651

Errata

An online log of corrections to Annual Review of Microbiology articles may be found athttp://micro.annualreviews.org/

viii Contents

Ann

u. R

ev. M

icro

biol

. 201

3.67

:291

-312

. Dow

nloa

ded

from

ww

w.a

nnua

lrev

iew

s.or

gby

Uni

vers

ity o

f L

eeds

on

09/1

6/13

. For

per

sona

l use

onl

y.


Recommended