Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

International Journal of Medical Microbiology 308 (2018) 237–245

Contents lists available at ScienceDirect

International Journal of Medical Microbiology


journal homepage: www.elsevier.com/locate/ijmm

Regulation of innate immune functions by guanylate-binding proteins T


a,b,⁎
Gerrit J.K. Praefcke
a
Division of Haematology / Transfusion Medicine, Paul-Ehrlich-Institut, Langen, Germany
b
Institute for Genetics, University of Cologne, Cologne, Germany

A R T I C L E I N F O A B S T R A C T

Keywords: Guanylate-binding proteins (GBP) are a family of dynamin-related large GTPases which are expressed in re-
Guanylate-binding proteins sponse to interferons and other pro-inflammatory cytokines. GBPs mediate a broad spectrum of innate immune
Interferon-inducible GTPase functions against intracellular pathogens ranging from viruses to bacteria and protozoa. Several binding partners
Autophagy for individual GBPs have been identified and several different mechanisms of action have been proposed de-
Inflammasome
pending on the organisms, the cell type and the pathogen used. Many of these anti-pathogenic functions of GBPs
Immunity-related GTPase
involve the recruitment to and the subsequent destruction of pathogen containing vacuolar compartments, the
Intracellular pathogens
assembly of large oligomeric innate immune complexes such as the inflammasome, or the induction of autop-
hagy. Furthermore, GBPs often cooperate with immunity-related GTPases (IRGs), another family of dynamin-
related GTPases, to exert their anti-pathogenic function, but since most IRGs have been lost in the evolution of
higher primates, the anti-pathogenic function of human GBPs seems to be IRG-independent. GBPs and IRGs share
biochemical and structural properties with the other members of the dynamin superfamily such as low nu-
cleotide affinity and a high intrinsic GTPase activity which can be further enhanced by oligomerisation.
Furthermore, GBPs and IRGs can interact with lipid membranes. In the case of three human and murine GBP
isoforms this interaction is mediated by C-terminal isoprenylation. Based on cell biological studies, and in
analogy to the function of other dynamins in membrane scission events, it has been postulated that both GBPs
and IRGs might actively disrupt the outer membrane of pathogen-containing vacuole leading to the detection
and destruction of the pathogen by the cytosolic innate immune system of the host. Recent evidence, however,
indicates that GBPs might rather function by mediating membrane tethering events similar to the dynamin-
related atlastin and mitofusin proteins, which mediate fusion of the ER and mitochondria, respectively. The aim
of this review is to highlight the current knowledge on the function of GBPs in innate immunity and to combine it
with the recent progress in the biochemical characterisation of this protein family.

1. Introduction human and murine GBPs showed an unusual conservation of the ca-
nonical GTP-binding motifs. While humans express seven GBPs, the
Interferons are cytokines released upon viral or microbial infections murine genome codes for 11 genes (Kresse et al., 2008; Olszewski et al.,
to induce the expression of several hundred antivirally or anti- 2006). In both, humans and mice, three GBPs carry a C-terminal iso-
microbially active genes (MacMicking, 2012; Platanias, 2005; Boehm prenylation motif, the CaaX-box (Cheng et al., 1991). While the se-
et al., 1998). Of these, four families of GTPases are among the most quence homology between dynamin and the antiviral Mx GTPases had
strongly induced genes: the guanylate-binding proteins (GBP), the im- been identified around that same time (Obar et al., 1990), the GBPs
munity-related GTPases (IRG), the Mx-proteins and the very large in- were only included in the dynamin superfamily when the biochemical
ducible GTPases (VLIG) (Li et al., 2009; MacMicking, 2012; Meunier and structural similarities became apparent and when other dynamin-
and Broz, 2016; Pilla-Moffett et al., 2016). related protein families were identified (Ghosh et al., 2006; Praefcke
The guanylate-binding proteins were identified due to their strong et al., 1999; Praefcke and McMahon, 2004; Prakash et al., 2000a;
induction by type II interferons (IFN-γ) and their ability to interact with Schwemmle and Staeheli, 1994)(Fig. 1). The common feature of dy-
guanine nucleotides (Cheng et al., 1983). The characterisation of the namin GTPases include a high intrinsic GTPase activity, which is fur-
GBP promotor was crucial for the elucidation of interferon-gamma ther stimulated by oligomerisation, and the interaction with biological
signalling (Darnell et al., 1994; Lew et al., 1991; Platanias, 2005) long membranes. Activation of the GTPases on membranes induces a change
before the cellular functions of GBPs were identified. Cloning of the first in membrane conformation resulting in either fission or fusion (Daumke


Corresponding author at: Division of Haematology / Transfusion Medicine, Paul-Ehrlich-Institut, Langen, Germany.
E-mail address: gerrit.praefcke@pei.de.

https://doi.org/10.1016/j.ijmm.2017.10.013
Received 22 August 2017; Received in revised form 27 October 2017; Accepted 31 October 2017
1438-4221/ © 2017 The Author. Published by Elsevier GmbH. This is an open access article under the CC BY license (http://creativecommons.org/licenses/BY/4.0/).
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

Fig. 1. Structural comparison of mammalian dy-


namin superfamily proteins. A: Structural compar-
ison of hGBP1 and two conformations of atlastin1.
The large GTPase (LG)-domains are shown in orange,
the middle domains in green and the C-terminal
α12/13-domain in hGBP1 in blue. Note that the
structures of atlastin only contain the N-terminal
part of the protein up to the first transmembrane
region. B: Structural comparison of dynamin1, MxA
and Irga6. The LG-domains are shown in orange, the
bundle signal element in red and the stalk in green.
The membrane binding PH-domain of dynamin1 and
the L4-region of MxA are shown in viridian green
and the N-terminal helical region of Irga6 in blue-
grey, respectively. Note that the structure of dy-
namin1 does not contain the C-terminal proline-rich
domain.

and Praefcke, 2016). The closest homologues of the GBPs within the (Haldar et al., 2013; Khaminets et al., 2010; Zhao et al., 2009). The
dynamin superfamily are the atlastin proteins. Atlastins are conserved current model for the mechanism of IRGs is based on analogy to other
from yeast to man and regulate dynamics of the endoplasmic reticulum dynamin-related proteins and on the careful observation of the se-
(ER) by tethering and fusion of ER tubules (Hu and Rapoport, 2016; quential order of pathogen entry, IRG recruitment to the PV membrane
McNew et al., 2013). (PVM), shape change and finally permeabilisation of the PV membrane.
The first publication of an innate immune function of GBPs reported A direct disruption of the PVM by IRG proteins, however, has so far not
an antiviral effect against vesicular stomatitis virus (VSV) and en- been formally proven (Zhao et al., 2009). Over time it became clear that
cephaloymyocarditis virus (EMCV) (Anderson et al., 1999). The anti- the deletion of individual regulatory GMS proteins can disrupt this
viral effect was not very prominent compared to the resistance medi- mutual regulatory network. In the case of Irgm1-/- mice, the mis-
ated by e.g. human MxA protein against influenza virus, which may be localisation of GKS-proteins leads to a loss of lysosomal acidity and
due to the incomplete depletion of hGBP1 and −2 by the used antisense autophagic processing resulting in IFN-γ-induced lymphopenia (Feng
RNA. A similar effect was found for murine GBP2 in a later study et al., 2008; King et al., 2011; Maric-Biresev et al., 2016; Traver et al.,
(Carter et al., 2005). The first publications showing an anti-pathogenic 2011). For some pathogens this effect has been interpreted as loss of
activity of GBPs against protozoa (Degrandi et al., 2013; Yamamoto resistance (Feng et al., 2008), but against others, such as Chlamydia,
et al., 2012) and bacteria (Tietzel et al., 2009) appeared a decade late. Irgm1 does mediate cell autonomous resistance (Coers et al., 2011). In
Since then is has become clear that GBPs are important for the defence mice the recruitment of IRGs and GBPs to PVs are linked to proteins
against a broad array of intracellular pathogens (Meunier and Broz, from the autophagy pathway (Al-Zeer et al., 2009; Haldar et al., 2014;
2016; Pilla-Moffett et al., 2016). Khaminets et al., 2010) (Fig. 2). After relocalisation to the PV of T.
The immunity-related GTPases (IRGs), were initially identified in gondii and C. trachomatis, the GKS IRGs mediate recruitment of ubi-
mice due to their strong induction by IFN-γ starting with IRGM1 under quitin E3 ligases such as TRAF6 to the membrane. The ubiquitylation of
the name LRG-47 (Gilly and Wall, 1992) followed by five other isoforms proteins on the PV, probably including the IRGs (Traver et al., 2011),
(Boehm et al., 1998; Carlow et al., 1995; Lafuse et al., 1995; Sorace creates binding sites for the ubiquitin-binding protein p62/sequesto-
et al., 1995; Taylor et al., 1996). Later it became apparent that the some (SQSTM1), which is itself a binding partner of mGBP2 (Haldar
genes coding for this protein family, which was referred to as p47 et al., 2015) and mGBP1 (Kim et al., 2011). The recruitment of GBPs to
GTPases to distinguish them from the previously identified p65/67 the T. gondii PV in murine cells requires GTP-binding but is independent
GBPs, is evolving fast leading to more than 20 murine isoforms de- of GTP-hydrolysis and of isoprenylation (Kravets et al., 2012; Virreira
pending on the mouse strain (Lilue et al., 2013). In contrast, IRGs are Winter et al., 2011). Accordingly, pathogen strains which express
almost completely lost in humans (Bekpen et al., 2005; Bekpen et al., virulence factors that disable the IRG system (Behnke et al., 2012;
2009). Biochemically, the best studied IRG protein is Irga6 (Ghosh Fentress et al., 2010; Fleckenstein et al., 2012; Steinfeldt et al., 2010)
et al., 2004; Pawlowski et al., 2011; Schulte et al., 2016; Uthaiah et al., also indirectly inactivate the GBPs and genetic deletions of GMS IRGs
2003) which displays a GTP-dependent oligomerisation and a dimer- resulting in mislocalisation of GKS IRGs also affect GBPs (Traver et al.,
isation induced stimulation of the GTPase activity. In cells Irga6 can be 2011). In human cells, IRGM is the only IRG isoform with an immune
N-terminally myristoylated (Martens et al., 2004; Papic et al., 2008). function (Bekpen et al., 2009; Chauhan et al., 2016). The protein is
Despite their later identification, the biological function of IRGs was truncated compared to canonical IRG proteins and its expression if not
more intensively studies than the GBPs. The first report of an anti-pa- inducible by interferons. IRGM is involved in the regulation of autop-
thogenic activity of an IRG identified the apicomplexan parasite Tox- hagy and polymorphisms in the gene are linked to Crohn’s disease and
oplasma gondii as a target pathogen (Taylor et al., 2000). The study of the susceptibility to tuberculosis. So far, a connection to the function of
murine IRG knock-out strains strongly accelerated the identification of human GBPs has not been shown.
additional resistance phenotypes. Cell biological studies showed that a
subgroup of the IRGs, the so called GMS-proteins, serve as negative 2. Structural and biochemical framework of GBPs
regulators of the other IRG family members. (Hunn et al., 2008). In the
absence of pathogens these GMS-proteins reside on different en- The domain architecture of GBPs consists of three parts (Fig. 1). The
domembrane compartment and protect them from the attack by the N-terminal large GTPase (LG) domain harbours the GTP-binding site
other IRGs, the so-called GKS proteins. Certain pathogens (e.g. T. gondii, which can be identified by three GTP-binding motifs: the phosphate
Chlamydia and E. cuniculi), enter the host cell by non-phagocytic me- binding P-loop, the DxxG motif and the RD-motif, which deviates from
chanisms. During the active invasion by T. gondii, most transmembrane the N/TKxD motif in other GTP-binding proteins (Cheng et al., 1991;
proteins of the host are excluded from the membrane of the forming PV Praefcke et al., 1999). Upon GTP-binding the GBPs dimerise, which
(Mordue et al., 1999). The GKS proteins identify such PVs as non-self stimulates their enzymatic activity (Ghosh et al., 2006; Praefcke et al.,
structures by the absence of GMS-proteins and are recruited to them 2004; Prakash et al., 2000a; Prakash et al., 2000b; Wehner et al., 2012;
resulting in the disruption of the PV and the destruction of the parasite Wehner and Herrmann, 2010). The LG-domain is followed by a helical

238
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

Fig. 2. Model of the innate immunity function of GBPs. A: In human cells, cytoplasmic GBPs can either form large oligomers (1) or interact with membrane compartments such as the
plasma membrane, phagosomes (2) or the Golgi. GTP-hydrolysis maintains a dynamic exchange of monomers and may lead to membrane tethering (3). Certain viruses can be targeted by
GBPs (4). Chlamydia and Toxoplasma parasites residing in parasitophorous vacuoles (PV) are labelled with ubiquitin (5) and become surrounded by autophagosome-like structure positive
for LC3. The recruitment of GBPs to the PV does not occur in all cell types and seems not to be linked to the ubiquitylation of the PV. For intracellular gram-negative bacteria it is currently
unclear whether GBPs contribute to membrane destabilisation (6) or whether they are recruited later. In immune cells, GBPs activate inflammasomes in collaboration with LPS and other
bacterial patterns leading to pyroptosis (7). B: In murine cells, IRG proteins from the GKS group are recruited from their resting localisations, where they were in complex with GMS group
members (1). On the PV IRGs trigger ubiquitylation resulting in the binding of further E3 ligases (2). GBPs reside in large oligomeric structures (3) and on vesicles (4). From there they are
shuttled to the ubiquitinylated PV by the adaptor protein p62 (5). For the isoprenylated GBP this recruitment is counteracted by the interaction with RabGDIα. It has been suggested that
the formation of large complexes of IRGs and GBPs leads to the lysis of both the PV and the parasite membrane (6) resulting in death of the pathogen. The activation of the inflammasome
in murine immune cells by gram-negative bacteria seems to follow a similar mechanism as in human cells (7).

part which has been subdivided into the middle domain (MD) and the that these extended conformations of the dimers of both, hGBP1 and
C-terminal α12/13-domain. The middle domain consists of two three- hGBP5, with their increased hydrodynamic radius have falsely been
helix bundles which share one helix and extend away from the LG- identified as tetramers in earlier studies (Pandita et al., 2016; Praefcke
domain. The α12/13-domain runs along the middle domain and back to et al., 2004; Prakash et al., 2000a; Shenoy et al., 2012; Vöpel et al.,
the LG-domain, where these two domains make a few but mechan- 2010; Wehner and Herrmann, 2010). This underlines the importance of
istically important interactions. During dimerisation of the LG-domains, shape-independent methods to determine the molecular weight of ex-
a conserved arginine residue in the phosphate-binding P-loop is posi- tended protein complexes in solution.
tioned into the active site and stimulates the cleavage of the distal All biochemical studies described so far have been performed with
phosphate group by stabilizing the transition state of the reaction. The GBPs lacking the lipid modification. However, when expressed in
presence of a catalytic arginine in the P-loop and the RD-motif are also mammalian cells, three of the seven human GBPs and three of the
found in atlastins (Daumke and Praefcke, 2016; Hu and Rapoport, eleven murine GBPs, the isoforms GBP1, −2 and −5, are post-trans-
2016; McNew et al., 2013), but not in other dynamin-like proteins. A lationally modified by attachment of a C15 (farnesyl) or a C20 (ger-
peculiar feature of the GBPs, and hGBP1 in particular, is the ability to anylgeranyl) isoprene moiety (Nantais et al., 1996; Olszewski et al.,
cleave GTP to GDP and GMP (Abdullah et al., 2010; Ghosh et al., 2006; 2006; Stickney and Buss, 2000). HGBP1 is isoprenylated with a C15
Kravets et al., 2012; Kunzelmann et al., 2006; Neun et al., 1996; farnesyl lipid moiety and, in addition, possesses a polybasic region di-
Pandita et al., 2016; Schwemmle and Staeheli, 1994; Wehner and rectly preceding the CaaX-motif of the protein, which could enhance
Herrmann, 2010). The product ratio differs between 0% and > 85% membrane affinity (Britzen-Laurent et al., 2010). Using a bacterial co-
GMP for different isoforms and experimental conditions. So far, the expression system, we were able to produce farnesylated recombinant
physiological function of this activity is not known. Based on gelfil- hGBP1 in E. coli. In contrast to most other isoprenylated proteins, the
tration studies we have postulated that hGBP1 dimers can assemble into lipid modified form of hGBP1 did not stably interact with membranes in
tetramers during GTP-hydrolysis or when bound to the transition state cosedimentation assays except when bound to the GDP*AlFx transition
analogue complex of GDP and aluminium fluoride (AlFx) (Prakash state analogue complex (Fres et al., 2010). In contrast, when the
et al., 2000a). Inhibition or reduction of dimer formation by mutations, binding of farnesylated hGBP1 to giant unilamellar vesicles (GUVs) was
immobilisation or dilution to low concentrations leads to a strongly directly imaged, the protein was also recruited to the membranes in the
reduced GTP turn-over and lower GMP production (Kunzelmann et al., presence of GTP or non-hydrolysable GTP-analogues (Shydlovskyi
2006; Praefcke et al., 2004; Syguda et al., 2012b). During GTP-hydro- et al., 2017). The interaction with membranes in the presence of GTP
lysis the interaction between the LG- and the α12/13-domain is tran- was transient and hGBP1 dissociated from the membrane when the
siently released in hGBP1 and the LG-domains of two hGBP1 molecules substrate was consumed. Accordingly, a mutant of the catalytic arginine
enter into an interaction (Syguda et al., 2012a; Vöpel et al., 2009; Vöpel displayed a stable interaction with membranes. In contrast to most
et al., 2014). This conformational change leads to an increase of the other dynamin-related proteins, membrane binding did not accelerate
length of the hGBP1 complex from 120 Å to up to 240 Å (Vöpel et al., the rate of GTP-hydrolysis. In the absence of membranes, however,
2014). Furthermore, a very recent study (Ince et al., 2017) has revealed GTP-bound farnesylated hGBP1 formed ring-like polymers which could

239
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

further assemble into longer forms. Another mutant with a strongly 4. Antiviral action of GBPs
reduced GMP production showed no evidence for such a polymerisa-
tion. Stimulation of the GTP-hydrolysis resulted in the disassembly of As mentioned before, the antiviral activity of hGBP1 against VSV
the polymers, which is reminiscent of the behaviour of Irga6 in the and EMCV was the first evidence for a function of GBPs in pathogen
presence of GTP (Ghosh et al., 2004; Pawlowski et al., 2011; Uthaiah defence (Anderson et al., 1999). It should be noted that the reported
et al., 2003). While we found no evidence for a disruption of the antiviral activity of GBPs is weak compared to that of Mx proteins for
membrane of the GUVs, we observed a nucleotide dependent tethering their target viruses (Haller et al., 2015). So far, antiviral activity of
activity of farnesylated hGBP1 (Shydlovskyi et al., 2017). Although we several GBPs against different viruses has been found and although
did not observe evidence for membrane fusion, these findings imply a there is no general pattern, many of these viruses are positive sense
mechanistic similarity between GBPs and the atlastins and other dy- RNA (+RNA) viruses. This includes hepatitis C virus (HCV), a member
namin-related proteins involved in membrane effusion events and add a of the flaviviridae family which showed increased replication upon
new aspect to the search for the function of GBPs on cellular mem- knock-down of hGBP1 (Itsui et al., 2009). Overexpression of hGBP1
branes. suppressed both replication and virus production of HCV, which is
mediated by the interaction of the LG-domain of hGBP1 with the finger
domain of non-structural protein 5B (NS5B) of HCV. Interestingly, the
3. Cellular localisation of GBPs interaction reduced the GTPase activity of hGBP1 and was blocked by a
mutation of the catalytic arginine residue in the P-loop and other re-
In interferon-induced cells and in the absence of pathogens, human sidues in the LG-domain of hGBP1 important for the GTPase cycle (Itsui
GBP1 displays a granular or vesicular cytosolic distribution (Britzen- et al., 2009; Pandita et al., 2016). These studies suggest that HCV-NS5B
Laurent et al., 2010). In cell fractionations it was mainly found in the interacts specifically with hGBP1 in its activated form. A very similar
cytosolic fraction despite its lipid modification (Nantais et al., 1996). scenario was also seen for classical swine fever virus (CSFV) (Li et al.,
This can be explained by the transient membrane localisation and 2016). In the case of this flavivirus, the antiviral effect of porcine GBP1
polymerisation which we have described (Shydlovskyi et al., 2017). (sGBP1) was also dependent on a functional LG-domain and binding of,
Addition of AlFx induces the redistribution of IFN-γ induced hGBP1 to in this case, the NS5A protein to the LG-domain inhibited the GTPase
the Golgi complex in a farnesylation-dependent manner (Modiano activity. Since also dengue virus, another human flavivirus, is inhibited
et al., 2005). Tripal and colleagues studied the localisation of fluores- by hGBP1 (Pan et al., 2012) it seems likely that also other viruses from
cently labelled hGBP1 in IFN-γ treated human endothelial cells (Tripal that family might be targets of GBPs.
et al., 2007) confirming these results. Furthermore, the localisation of Several human GBPs have been found to interfere with influenza
hGBP2 followed the same pattern with additional nuclear staining virus (-ssRNA). Nordmann and colleagues reported that hGBP1, hGBP3
which was also seen for hGBP4, while hGBP3 was exclusively cyto- and especially a novel splice variant hGBP-3ΔC repressed the activity of
plasmic and hGBP5 constitutively Golgi-associated. Not surprisingly, the viral polymerase complex (Nordmann et al., 2012). For this func-
localisation of the human CaaX-GBPs is prenylation dependent (Britzen- tion, GTP-binding, but not hydrolysis was necessary. Binding of NS1
Laurent et al., 2010) and the three prenylated isoforms are able to re- from influenza A virus (IAV) to hGBP1 resulted in a reduction in both
cruit the non-prenylated isoforms hGBP3 and hGBP4 to their respective GTPase activity and the anti-IAV activity (Nordmann et al., 2012). For
localisation by heterodimerisation. Furthermore, a double mutation of mGBP5 an indirect effect on IAV was described by upregulating the
the contact between the LG-domain and the α12/13-domain (R227E/ expression of IFN and other proinflammatory cytokines (Feng et al.,
K228E) results in enhanced membrane binding of hGBP1 and its re- 2017). A direct interference with viral propagation was described for
cruitment to the plasma membrane in vivo, while a protein mutant hGBP5 and human immunodeficiency virus (HIV) by interfering with
impaired in nucleotide-binding is exclusively cytosolic (Britzen-Laurent processing and incorporation of the viral envelope glycoprotein (Krapp
et al., 2010; Vöpel et al., 2010). Localisation of hGBP1 to the plasma et al., 2016). For this process, the isoprenylation-dependent Golgi lo-
membrane has also been seen in intestinal epithelial cells, where it calisation of hGBP5 was needed, but mutations impairing GTP-binding
localised at tight junctions to regulate the barrier function (Schnoor and hydrolysis retained their activity against HIV. In pigs, poly-
et al., 2009). We have recently analysed the nature of the punctate morphisms in the GBP locus have been associated with the outcome of
cytoplasmic hGBP1 structures in HeLa cells and found a partially co- porcine reproduction and respiratory syndrome virus (PRRSV,
localisation with marker proteins of the endolysosomal pathway such as +ssRNA) (Abella et al., 2016; Boddicker et al., 2012; Koltes et al.,
Rab5, EEA1, late Rab7 and LAMP1 (Shydlovskyi et al., 2017). Fur- 2015, 2015; Kommadath et al., 2017; Niu et al., 2016; Schroyen et al.,
thermore, endogenous IFN-γ-induced as well as recombinant mCherry- 2016). Animals homozygous for the unfavourable genotype pre-
labelled hGBP1 was recruited to latex bead phagosomes where it co- dominantly produced a splice variant of hGBP5 coding for a C-terminal
localised with F-actin. Photobleaching experiments revealed that the truncation of 88 amino acids lacking the isoprenylation site (Koltes
localisation to latex bead phagosomes and to the other cytosolic vesicle- et al., 2015; Schroyen et al., 2016). A similar splice variant was also
like structures is very dynamic. seen in human cutaneous T-cell lymphoma cells (Fellenberg et al.,
In IFN-treated murine macrophages and fibroblasts, a similar 2004; Wehner and Herrmann, 2010). The only antiviral effect of a GBP
granular cytoplasmic distribution and partial localisation to vesicle-like against a DNA virus has recently been shown for Kaposi’s sarcoma as-
structures of heterogeneous sizes was found for the CaaX bearing iso- sociated herpesvirus (KSHV) (Zou et al., 2017). In this study the authors
form mGBP2 (Balasubramanian et al., 2011; Vestal et al., 2000), while show that hGBP1 mediates the inhibition of the nuclear delivery of
mGBP1 displayed a homogeneous cytoplasmic distribution. Also the KSHV virions by disrupting the formation of actin filaments. In this
other murine GBPs displayed a discrete vesicle-like localisation which context it is noteworthy to mention that upregulation of hGBP1 had
has so far not been further analysed (Degrandi et al., 2007). Expression already been identified several years ago in vivo in endothelial cells
of mutant versions of mGBP2 in IFN-γ-induced mGBP2-/- MEFs re- from Kaposi's sarcoma tissue (Guenzi et al., 2001). This study initiated
vealed that GTP-binding and hydrolysis are important for the proper the investigation into the role of hGBP1 in the regulation of cell pro-
localisation and also for the formation of multimers in the cellular liferation in endothelial cells (Britzen-Laurent et al., 2016) which led to
context (Kravets et al., 2012). These large complexes and also the ve- the recent identification of hGBP1 as an actin binding protein (Ostler
sicle-like structures contain also heteromultimers of mGBP2 with other et al., 2014).
murine GBPs and serve as cytoplasmic reservoir from where mGBPs are Only two studies of antiviral activity of murine GBPs have so far
recruited to parasitophorous vacuoles (Kravets et al., 2016). been published. Carter and colleagues found that mGBP2 inhibited the
replication of both VSV (-ssRNA) and EMCV (+ssRNA) (Carter et al.,

240
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

2005) but with different requirements. While a functional GTP binding for C. muridarum infecting human endothelial cells followed by the
motif was required for inhibition of EMCV, it was dispensable for in- recruitment of p62, LC3 and hGBP1 and finally elimination of the in-
hibition of VSV. Recently, the targeting of the viral replication centres clusions. The human-specific pathogen C. trachomatis, however, was
of murine norovirus (MNV, +ssRNA) by the microtubule-associated- resistant to ubiquitylation and elimination (Haldar et al., 2016). Fur-
protein-1-light-chain-3 (LC3) followed by the recruitment of both IRGs thermore, the mechanisms by which cells respond to bacterial infec-
and GBPs has been described (Biering et al., 2017). The fact that both tions is also cell-type specific. In human macrophages, GBPs are not
LC3 and the IFN-inducible GTPases were necessary to inhibit MNV re- recruited to C. muridarum inclusions (Finethy et al., 2015). Instead,
plication in mice and in human cells is highly reminiscent of the de- GBPs activate NLRP3- and AIM2-inflammasomes leading to the release
fence against T. gondii and C. trachomatis. of inflammatory cytokines and pyroptosis of the macrophage. Similar
observations have also been made for GBP-dependent induction of
5. Antibacterial function pyroptosis by infection of murine macrophages with Legionella pneu-
moniae (Pilla et al., 2014). This study also showed that the induction of
In recent years, the antimicrobial function of the murine GBPs has pyroptosis by the mGBPs on chromosome 3 depends on the presence of
been established by several groups. Specificity towards different pa- LPS in the cytoplasm. Another study, however, indicated that these
thogens and the underlying molecular mechanisms have remained GBPs are required upstream of LPS sensing (Meunier et al., 2014). In-
controversial. The direct comparison of the different observations is stead the authors found reduced levels of Salmonella in cells that are
impeded by the use of different pathogens, host cell types and the use of positive for galectin-8, a marker for lysed vacuoles (Thurston et al.,
murine or human model systems. 2012). Since galectin-8 is also a binding partner for the autophagy
In proteomic studies several murine GBPs were enriched on latex adaptor protein NDP52, lysed vacuoles could be detected by the au-
bead phagosomes (Jutras et al., 2008; Trost et al., 2009). Tietzel and tophagic machinery. The early role of GBPs in the activation of in-
colleagues provided the first evidence for an antibacterial activity of flammatory signals prompted Meunier and colleagues to propose that
human GBPs (Tietzel et al., 2009). They showed that ectopically ex- GBPs might promote the lysis of vacuoles thereby triggering the de-
pressed hGBP1 and hGBP2 are recruited to the inclusion membrane of tection of pathogen associated molecular patterns (PAMPs) (Meunier
C. trachomatis in HeLa cells and inhibit growth of the bacteria, while et al., 2014). A following study suggested that GBPs might not only be
down regulation of hGBP1 and −2 alleviated the inhibitory effect of involved in the attack on the PV but also on the pathogen itself
interferon-γ. Recruitment of GBP1 to Chlamydia was also observed in (Meunier and Broz, 2015) since the number of lysed cells of Francisella
murine dendritic cells (Fiegl et al., 2013) and a human macrophage cell novicida in murine macrophages lacking the GBPs on chromosome 3
line (Al-Zeer et al., 2013). The latter study also showed that the anti- was higher than in macrophages from wild-type mice. In this context,
chlamydial action of hGBP1 and −2 involved changes in the autophagy mGBP2 and mGBP5 also triggered pyroptosis but via the activation of
pathway leading to the rerouting of the typically non-fusogenic bac- the AIM2-inflammasome. Recent evidence points towards a cooperative
terial inclusions to lysosomal degradation. Recently, it has been shown function of GBPs and IRGs, however in a different order than previously
that also other murine GBPs are recruited to the PV of Chlamydia or T. described for infections with C. trachomatis since Irgb10 was recruited
gondii to variable degrees (Lindenberg et al., 2017). to F. novicida cells in a GBP-dependent manner (Man et al., 2016). The
Kim et al. have reported that murine GBPs are critical for control of subsequent lysis of the bacteria releases LPS and DNA which then ac-
Listeria monocytogenes and Mycobacteria bovis infections (Kim et al., tivate caspase-11, AIM2 and NLRP3. A very recent study indicates that
2011). They proposed that mGBP1 and mGBP7 enhance the formation the effect of GBP against F. novicida is not only mediated by the AIM2
of reactive oxygen by recruitment of NADPH oxidase to phagosomes inflammasome. In interferon primed macrophages expressing high le-
and induce autophagy via the interaction with p62/SQSTM1 and Atg4b. vels of several GBPs the authors found that GBPs control also several
The same group also reported that human and murine GBP5 promote other apoptotic pathways and control killing of intracellular bacteria in
the assembly and activation of the NLRP3 inflammasome to control an inflammasome-independent manner (Wallet et al., 2017). The link
Listeria infections (Shenoy et al., 2012). However, Yamamoto et al. between galectins, GBPs and activation of inflammasomes was further
found no effect on the proliferation of Listeria in mice lacking a region devised by several recent studies. In cells infected with Yersinia or Le-
of chromosome 3 containing mGBP1, mGBP2, mGBP3, mGBP5 and gionella, the presence of bacterial secretion systems recruited galectin-3
mGBP7 and also no effect on the recruitment of Atg8/LC3 and the to the PVs followed by delivery of GBP1 and GBP2 (Feeley et al., 2017).
formation of reactive oxygen species (Yamamoto et al., 2012). Using Recently, it has been reported that GBPs can also mediate caspase-11-
mice lacking mGBP2 Degrandi and colleagues also found no effect dependent pyroptosis in response to LPS which is delivered to cells by
against Listeria (Degrandi et al., 2013). outer membrane vesicles of gram-negative bacteria (Finethy et al.,
A series of studies has investigated the cooperation of GBPs and 2017). In zebrafish, GBP4 contains a C-terminal CARD domain. By di-
IRGs in murine cells at the vacuoles of both T. gondii and Chlamydia. For merisation with the CARD-domain of the inflammasome adaptor ASC it
the IRGs it had already previously been shown that the loading on PVs mediates the clearance of Salmonella thyphimurium infections (Tyrkalska
requires an intact autophagy system (Al-Zeer et al., 2009; Khaminets et al., 2016). In case of cytosolic Shigella flexneri, the recruitment of
et al., 2010; Zhao et al., 2008). Haldar and colleagues showed that the several human GBPs seems to be independent of galectin-8 (Wandel
localisation of murine GBPs depends on an intact IRG system (Haldar et al., 2017). Instead, hGBP1 is recruited first and depending on its
et al., 2013). Subsequently, the same group showed that the docking of catalytic activity. Other GBPs follow in an hierarchical order to form a
IRGs and mGBPs to the PV depends on the expression of the autophagy dense coat which reduces the actin-based motility and spreading of the
proteins ATG3 and ATG5, the E2 enzyme and a subunit of the E3 bacteria. This effect is counteracted by a bacterial E3 ubiquitin ligase
complex for the LC3 lipidation, respectively (Haldar et al., 2015). The which targets hGBP1 and other GBPs for degradation.
link between these events is the ubiquitylation of the PV triggered by
the recruitment of E3-ubiquitin ligases to the PV by IRG proteins 6. Defence against protozoa
(Haldar et al., 2015). The following decoration of the PV with ubiquitin
then creates binding sites for p62/SQSTM1 which enhances the ubi- The analysis of the function of GBPs in innate immunity against
quitylation of the PV and recruits GBPs. As a result, the IFN-mediated protozoan pathogens has been following the study of the IRG proteins
and cell-autonomous resistance to C. trachomatis requires both families and has almost exclusively been focused on the apicomplexan parasites
of IFN-induced GTPases, the GBPs and the IRGs, and a functional au- T. gondii and Neospora caninum. Initially, only colocalisation of GBPs
tophagy pathway. In human cells, which lack most IRG proteins, the with the PV of T. gondii was shown (Degrandi et al., 2007; Kravets et al.,
mechanism seems to be different. Ubiquitylation of PVs was observed 2012; Virreira Winter et al., 2011), which, in the case of the CaaX-GBPs,

241
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

is depended on the isoprenylation and a functional GTPase-domain. The knock-down of hGBP1 reduced the IFN-γ dependent resistance (Qin
proof for an activity of GBPs against Toxoplasma infections was made et al., 2017).
possible by the generation of mouse strains lacking either individual
GBPs (Degrandi et al., 2013; Selleck et al., 2013) or the GBP locus on 7. Outlook
chromosome 3 containing mGBP1, mGBP2, mGBP3, mGBP5 and
mGBP7 (Yamamoto et al., 2012). The availability of these model sys- Research on GBP and IRG function has developed from the begin-
tems has been a milestone in the study of GBP function. Since then, the ning by mutual stimulation where similar questions were asked and
hierarchy of events at the PV of T. gondii in murine cells was elaborated addressed with similar experimental approaches. Now it has become
in parallel to the investigation of the activity against Chlamydia de- clear that the two IFN-inducible GTPase families are much more closely
scribed above. The possible sequence of events begins when the PV of interconnected than their primary sequence has indicated. In both fa-
actively invading T. gondii cells is recognised by the GKS IRG proteins milies, subsets of several isoforms act at distinct cellular compartments
due to the missing GMS IRGs (Hunn et al., 2008; Khaminets et al., 2010) and are recruited to their points of action in a controlled and hier-
and also by proteins of the autophagy pathway leading to the deposi- archical manner. Still the question remains if they all share the same
tion of LC3 on the PV (Choi et al., 2014). Several ubiquitin ligases such fundamental mechanism to control intracellular pathogens. The bio-
as TRAF6 (Haldar et al., 2015) and TRIM21 (Foltz et al., 2017) are chemical and structural analysis of the GBPs and IRGs has provided a
recruited to the PV, which they decorate with ubiquitin, probably on deep understanding of the conformational changes during GTP-binding
the IRGs (Traver et al., 2011). This initial ubiquitylation creates binding and hydrolysis. However, we should remember that we do actually only
site for the ubiquitin-binding adaptor protein p62 which escorts mGBP2 have detailed information about some of the family members and re-
to the PV (Pilla-Moffett et al., 2016). Before their relocalisation to the cent results show that posttranslational modifications or the presence of
PV, GBPs reside in high molecular weight complexes and on vesicle-like another isoforms may have dramatic effects on the biochemical activ-
structures (Kravets et al., 2016). On the PV they from even larger ities. Furthermore, we should be cautious with mechanistic models
complexes and after disruption of the PV they relocalise further to the which are based on circumstantial evidence and not direct proof. On the
parasite membrane. Upon disruption of its membrane the parasite dies, positive side is the availability of novel tools and models in which we
followed by necrotic cell death of the host cell (Zhao et al., 2009). Also can test our hypotheses. The CRISPR/Cas technology could be used to
as in the case of Chlamydia, intracellular T. gondii parasites trigger the introduce site-specific mutations into the genes of individual GBPs to
activation of the inflammasome in macrophages (Ewald et al., 2014; address the physiological significance of GMP-production, the inter-
Gorfu et al., 2014). It should be noted, however, that there are data and intramolecular interactions and the heterooligomerisation without
supporting alternative scenarios. Lee and colleagues, for example, come interfering with their endogenous IFN-stimulated expression. It will
to the conclusion that ubiquitylation of the PV and recruitment of p62 also be interesting to find out whether the defence of viruses involves
occur after the disruption of the PV by IFN-γ-inducible GTPases and are similar pathways as the defence against bacteria and protozoa and
needed to activate the acquired immune response (Lee et al., 2015). The whether the functions of GBPs in cell proliferation, motility and tumour
recruitment of GBP to the PV of T. gondii can also be negatively regu- suppression (Britzen-Laurent et al., 2016) are independent from the role
lated. Besides the indirect effect of mistargeting or inactivation of IRGs in innate immunity. The recent report that the processing and secretion
by virulence factors (Alaganan et al., 2014; Behnke et al., 2012; of hGBP1 depends on inflammatory caspase activity is a hint that these
Etheridge et al., 2014; Fentress et al., 2010; Fleckenstein et al., 2012; “moonlighting” functions may actually be functionally linked
Hermanns et al., 2016; Howard et al., 2011; Niedelman et al., 2012; (Naschberger et al., 2017).
Steinfeldt et al., 2010), T. gondii have also evolved effectors which di-
rectly target GBPs (Kim et al., 2016). Furthermore, also the host cells Funding
control the membrane recruitment of mGBP2 by the interaction with
the isoprenyl-binding protein RabGDIα (Ohshima et al., 2015). This work was funded by the German Research Foundation, Priority
In human cells, it was believed that tryptophan-depletion by the Programme 1580, Intracellular Compartments as Places of Pathogen-
IFN-inducible indoleamine 2,3-dioxygenase (IDO) (Pfefferkorn, 1984; Host Interactions, grant Pr 1090/3-1.
Pfefferkorn et al., 1986) and the generation of nitric oxide by inducible
nitric oxide synthase (iNOS) (Fox et al., 2004; Scharton-Kersten et al., References
1997) would mediate the effect against T. gondii infections (Yarovinsky,
2014). Within the recent years it has become apparent that the de- Abdullah, N., Balakumari, M., Sau, A.K., 2010. Dimerization and its role in GMP for-
pendence of the innate immune response against T. gondii in human cell mation by human guanylate binding proteins. Biophys. J. 99 (7), 2235–2244.
Abella, G., Pena, R.N., Nogareda, C., Armengol, R., Vidal, A., Moradell, L., Tarancon, V.,
shows a great variability between different cell-types and pathogen Novell, E., Estany, J., Fraile, L., 2016. A WUR SNP is associated with European
strains. In IFN-stimulated fibroblasts T. gondii is regulated by cell death Porcine Reproductive and Respiratory Virus Syndrome resistance and growth per-
independently of hGBP1 and hGBP2 (Niedelman et al., 2013), whereas formance in pigs. Res. Vet. Sci. 104, 117–122.
Al-Zeer, M.A., Al-Younes, H.M., Braun, P.R., Zerrahn, J., Meyer, T.F., 2009. IFN-gamma-
hGBP1 restricted replication of T. gondii in epithelial cells but did not inducible Irga6 mediates host resistance against Chlamydia trachomatis via autop-
localise to the PV (Johnston et al., 2016). Nevertheless, vacuoles of hagy. PLoS One 4 (2), e4588.
certain T. gondii strains in HUVECs were targeted by Lys63-linked Al-Zeer, M.A., Al-Younes, H.M., Lauster, D., Abu Lubad, M., Meyer, T.F., 2013. Autophagy
restricts Chlamydia trachomatis growth in human macrophages via IFNG-inducible
ubiquitin chains and recruited both p62 and NDP52, but not the au- guanylate binding proteins. Autophagy 9 (1), 50–62.
tophagy proteins Atg16L1, GABARAP and LC3B. The ubiquitylated Alaganan, A., Fentress, S.J., Tang, K., Wang, Q., Sibley, L.D., 2014. Toxoplasma GRA7
vacuoles were then destroyed in LAMP1-positive endo-lysosomal com- effector increases turnover of immunity-related GTPases and contributes to acute
virulence in the mouse. Proc. Natl. Acad. Sci. U. S. A. 111 (3), 1126–1131.
partments. In contrast to that, HeLa cells, which are a human epithelial
Anderson, S.L., Carton, J.M., Lou, J., Xing, L., Rubin, B.Y., 1999. Interferon-induced
cancer cell line, restricted T. gondii by non-canonical autophagy guanylate binding protein-1 (GBP-1) mediates an antiviral effect against vesicular
(Clough and Frickel, 2017). These results from endothelial cells are in stomatitis virus and encephalomyocarditis virus. Virology 256 (1), 8–14.
contrast to results obtained with the near haploid HAP1 cells, which are Balasubramanian, S., Messmer-Blust, A.F., Jeyaratnam, J.A., Vestal, D.J., 2011. Role of
GTP binding, isoprenylation, and the C-terminal α-helices in the inhibition of cell
derived from a leukaemia cell line. In these cells, GBPs did localise to spreading by the interferon-induced GTPase, mouse guanylate-binding protein-2. J.
the PV of T. gondii but deletion of all seven human GBPs had no effect Interferon Cytokine Res. 31 (3), 291–298.
on the IFN-γ mediated suppression of T. gondii replication (Ohshima Behnke, M.S., Fentress, S.J., Mashayekhi, M., Li, L.X., Taylor, G.A., Sibley, L.D., 2012. The
polymorphic pseudokinase ROP5 controls virulence in Toxoplasma gondii by reg-
et al., 2014). Again a different setting and outcome was reported for of ulating the active kinase ROP18. PLoS Pathog. 8 (11), e1002992.
human mesenchymal stromal cells. Depending on the T. gondii strain Bekpen, C., Hunn, J.P., Rohde, C., Parvanova, I., Guethlein, L., Dunn, D.M., Glowalla, E.,
between 10% and 20% of the vacuoles were recruiting hGBP1 and Leptin, M., Howard, J.C., 2005. The interferon-inducible p47 (IRG) GTPases in

242
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

vertebrates: loss of the cell autonomous resistance mechanism in the human lineage. proteins enable rapid activation of canonical and noncanonical inflammasomes in
Genome Biol. 6 (11), R92. Chlamydia-infected macrophages. Infect. Immun. 83 (12), 4740–4749.
Bekpen, C., Marques-Bonet, T., Alkan, C., Antonacci, F., Leogrande, M.B., Ventura, M., Finethy, R., Luoma, S., Orench-Rivera, N., Feeley, E.M., Haldar, A.K., Yamamoto, M.,
Kidd, J.M., Siswara, P., Howard, J.C., Eichler, E.E., 2009. Death and resurrection of Kanneganti, T.-D., Kuehn, M.J., Coers, J., 2017. Inflammasome activation by bac-
the human IRGM gene. PLoS Genet. 5 (3), e1000403. terial outer membrane vesicles requires guanylate binding proteins. mBio 8 (5).
Biering, S.B., Choi, J., Halstrom, R.A., Brown, H.M., Beatty, W.L., Lee, S., McCune, B.T., Fleckenstein, M.C., Reese, M.L., Könen-Waisman, S., Boothroyd, J.C., Howard, J.C.,
Dominici, E., Williams, L.E., Orchard, R.C., Wilen, C.B., Yamamoto, M., Coers, J., Steinfeldt, T., 2012. A Toxoplasma gondii pseudokinase inhibits host IRG resistance
Taylor, G.A., Hwang, S., 2017. Viral replication complexes are targeted by LC3- proteins. PLoS Biol. 10 (7), e1001358.
Guided interferon-Inducible GTPases. Cell Host Microbe 22 (1), 74–85 (e7). Foltz, C., Napolitano, A., Khan, R., Clough, B., Hirst, E.M., Frickel, E.-M., 2017. TRIM21 is
Boddicker, N., Waide, E.H., Rowland, R.R.R., Lunney, J.K., Garrick, D.J., Reecy, J.M., critical for survival of Toxoplasma gondii infection and localises to GBP-positive
Dekkers, J.C.M., 2012. Evidence for a major QTL associated with host response to parasite vacuoles. Sci. Rep. 7 (1), 5209.
porcine reproductive and respiratory syndrome virus challenge. J. Anim. Sci. 90 (6), Fox, B.A., Gigley, J.P., Bzik, D.J., 2004. Toxoplasma gondii lacks the enzymes required for
1733–1746. de novo arginine biosynthesis and arginine starvation triggers cyst formation. Int. J.
Boehm, U., Guethlein, L., Klamp, T., Ozbek, K., Schaub, A., Futterer, A., Pfeffer, K., Parasitol. 34 (3), 323–331.
Howard, J.C., 1998. Two families of GTPases dominate the complex cellular response Fres, J.M., Müller, S., Praefcke, G.J.K., 2010. Purification of the CaaX-modified, dynamin-
to IFN-gamma. J. Immunol. 161 (12), 6715–6723. related large GTPase hGBP1 by coexpression with farnesyltransferase. J. Lipid Res. 51
Britzen-Laurent, N., Bauer, M., Berton, V., Fischer, N., Syguda, A., Reipschläger, S., (8), 2454–2459.
Naschberger, E., Herrmann, C., Stürzl, M., 2010. Intracellular trafficking of guany- Ghosh, A., Uthaiah, R., Howard, J., Herrmann, C., Wolf, E., 2004. Crystal structure of
late-binding proteins is regulated by heterodimerization in a hierarchical manner. IIGP1: a paradigm for interferon-inducible p47 resistance GTPases. Mol. Cell 15 (5),
PLoS One 5 (12), e14246. 727–739.
Britzen-Laurent, N., Herrmann, C., Naschberger, E., Croner, R.S., Stürzl, M., 2016. Ghosh, A., Praefcke, G.J.K., Renault, L., Wittinghofer, A., Herrmann, C., 2006. How
Pathophysiological role of guanylate-binding proteins in gastrointestinal diseases. guanylate-binding proteins achieve assembly-stimulated processive cleavage of GTP
World J. Gastroenterol. 22 (28), 6434–6443. to GMP. Nature 440 (7080), 101–104.
Carlow, D.A., Marth, J., Clark-Lewis, I., Teh, H.S., 1995. 1995 Isolation of a gene en- Gilly, M., Wall, R., 1992. 1992 The IRG-47 gene is IFN-gamma induced in B cells and
coding a developmentally regulated T cell-specific protein with a guanine nucleotide encodes a protein with GTP-binding motifs. J. Immunol. 148 (10), 3275–3281.
triphosphate-binding motif. J. Immunol. 154 (4), 1724–1734. Gorfu, G., Cirelli, K.M., Melo, M.B., Mayer-Barber, K., Crown, D., Koller, B.H., Masters, S.,
Carter, C.C., Gorbacheva, V.Y., Vestal, D.J., 2005. Inhibition of VSV and EMCV replica- Sher, A., Leppla, S.H., Moayeri, M., Saeij, J.P.J., Grigg, M.E., 2014. Dual role for
tion by the interferon-induced GTPase, mGBP-2: differential requirement for wild- inflammasome sensors NLRP1 and NLRP3 in murine resistance to Toxoplasma gondii.
type GTP binding domain. Arch. Virol. 150 (6), 1213–1220. mBio 5 (1).
Chauhan, S., Mandell, M.A., Deretic, V., 2016. Mechanism of action of the tuberculosis Guenzi, E., Topolt, K., Cornali, E., Lubeseder-Martellato, C., Jorg, A., Matzen, K., Zietz, C.,
and Crohn disease risk factor IRGM in autophagy. Autophagy 12 (2), 429–431. Kremmer, E., Nappi, F., Schwemmle, M., Hohenadl, C., Barillari, G., Tschachler, E.,
Cheng, Y.S., Colonno, R.J., Yin, F.H., 1983. Interferon induction of fibroblast proteins Monini, P., Ensoli, B., Sturzl, M., 2001. The helical domain of GBP-1 mediates the
with guanylate binding activity. J. Biol. Chem. 258 (12), 7746–7750. inhibition of endothelial cell proliferation by inflammatory cytokines. EMBO J. 20
Cheng, Y.S., Patterson, C.E., Staeheli, P., 1991. Interferon-induced guanylate-binding (20), 5568–5577.
proteins lack an N(T)KXD consensus motif and bind GMP in addition to GDP and Haldar, A.K., Saka, H.A., Piro, A.S., Dunn, J.D., Henry, S.C., Taylor, G.A., Frickel, E.M.,
GTP. Mol. Cell. Biol. 11 (9), 4717–4725. Valdivia, R.H., Coers, J., 2013. IRG and GBP host resistance factors target aberrant,
Choi, J., Park, S., Biering, S.B., Selleck, E., Liu, C.Y., Zhang, X., Fujita, N., Saitoh, T., non-self vacuoles characterized by the missing of self IRGM proteins. PLoS Pathog. 9
Akira, S., Yoshimori, T., Sibley, L.D., Hwang, S., Virgin, H.W., 2014. The para- (6), e1003414.
sitophorous vacuole membrane of Toxoplasma gondii is targeted for disruption by Haldar, A.K., Piro, A.S., Pilla, D.M., Yamamoto, M., Coers, J., 2014. The E2-like con-
ubiquitin-like conjugation systems of autophagy. Immunity 40 (6), 924–935. jugation enzyme Atg3 promotes binding of IRG and Gbp proteins to Chlamydia- and
Clough, B., Frickel, E.-M., 2017. The toxoplasma parasitophorous vacuole: an evolving Toxoplasma-containing vacuoles and host resistance. PLoS One 9 (1), e86684.
host-Parasite frontier. Trends Parasitol. 33 (6), 473–488. Haldar, A.K., Foltz, C., Finethy, R., Piro, A.S., Feeley, E.M., Pilla-Moffett, D.M., Komatsu,
Coers, J., Gondek, D.C., Olive, A.J., Rohlfing, A., Taylor, G.A., Starnbach, M.N., 2011. M., Frickel, E.-M., Coers, J., 2015. Ubiquitin systems mark pathogen-containing va-
Compensatory T cell responses in IRG-deficient mice prevent sustained Chlamydia cuoles as targets for host defense by guanylate binding proteins. Proc. Natl. Acad. Sci.
trachomatis infections. PLoS Pathog. 7 (6), e1001346. U. S. A. 112 (41), E5628–37.
Darnell, J.E., Kerr, I.M., Stark, G.R., 1994. Jak-STAT pathways and transcriptional acti- Haldar, A.K., Piro, A.S., Finethy, R., Espenschied, S.T., Brown, H.E., Giebel, A.M., Frickel,
vation in response to IFNs and other extracellular signaling proteins. Science 264 E.-M., Nelson, D.E., Coers, J., 2016. Chlamydia trachomatis is resistant to inclusion
(5164), 1415–1421. ubiquitination and associated host defense in gamma interferon-Primed human epi-
Daumke, O., Praefcke, G.J.K., 2016. Invited review: mechanisms of GTP hydrolysis and thelial cells. mBio 7 (6).
conformational transitions in the dynamin superfamily. Biopolymers 105 (8), Haller, O., Staeheli, P., Schwemmle, M., Kochs, G., 2015. Mx GTPases: dynamin-like
580–593. antiviral machines of innate immunity. Trends Microbiol. 23 (3), 154–163.
Degrandi, D., Konermann, C., Beuter-Gunia, C., Kresse, A., Würthner, J., Kurig, S., Beer, Hermanns, T., Müller, U.B., Könen-Waisman, S., Howard, J.C., Steinfeldt, T., 2016. The
S., Pfeffer, K., 2007. 2007 Extensive characterization of IFN-induced GTPases mGBP1 Toxoplasma gondii rhoptry protein ROP18 is an Irga6-specific kinase and regulated
to mGBP10 involved in host defense. J. Immunol. 179 (11), 7729–7740. by the dense granule protein GRA7. Cell. Microbiol. 18 (2), 244–259.
Degrandi, D., Kravets, E., Konermann, C., Beuter-Gunia, C., Klümpers, V., Lahme, S., Howard, J.C., Hunn, J.P., Steinfeldt, T., 2011. The IRG protein-based resistance me-
Wischmann, E., Mausberg, A.K., Beer-Hammer, S., Pfeffer, K., 2013. Murine guany- chanism in mice and its relation to virulence in Toxoplasma gondii. Curr. Opin.
late binding protein 2 (mGBP2) controls Toxoplasma gondii replication. Proc. Natl. Microbiol. 14 (4), 414–421.
Acad. Sci. U. S. A. 110 (1), 294–299. Hu, J., Rapoport, T.A., 2016. Fusion of the endoplasmic reticulum by membrane-bound
Etheridge, R.D., Alaganan, A., Tang, K., Lou, H.J., Turk, B.E., Sibley, L.D., 2014. The GTPases. Semin. Cell Dev. Biol. 60, 105–111.
Toxoplasma pseudokinase ROP5 forms complexes with ROP18 and ROP17 kinases Hunn, J.P., Koenen-Waisman, S., Papic, N., Schroeder, N., Pawlowski, N., Lange, R.,
that synergize to control acute virulence in mice. Cell Host Microbe 15 (5), 537–550. Kaiser, F., Zerrahn, J., Martens, S., Howard, J.C., 2008. Regulatory interactions be-
Ewald, S.E., Chavarria-Smith, J., Boothroyd, J.C., 2014. NLRP1 is an inflammasome tween IRG resistance GTPases in the cellular response to Toxoplasma gondii. EMBO J.
sensor for Toxoplasma gondii. Infect. Immun. 82 (1), 460–468. 27 (19), 2495–2509.
Feeley, E.M., Pilla-Moffett, D.M., Zwack, E.E., Piro, A.S., Finethy, R., Kolb, J.P., Martinez, Ince, S., Kutsch, M., Shydlovskyi, S., Herrmann, C., 2017. The human guanylate-binding
J., Brodsky, I.E., Coers, J., 2017. Galectin-3 directs antimicrobial guanylate binding proteins hGBP-1 and hGBP-5 cycle between monomers and dimers only. FEBS J. 284
proteins to vacuoles furnished with bacterial secretion systems. Proc. Natl. Acad. Sci. (14), 2284–2301.
U. S. A. 114 (9), E1698–E1706. Itsui, Y., Sakamoto, N., Kakinuma, S., Nakagawa, M., Sekine-Osajima, Y., Tasaka-Fujita,
Fellenberg, F., Hartmann, T.B., Dummer, R., Usener, D., Schadendorf, D., Eichmuller, S., M., Nishimura-Sakurai, Y., Suda, G., Karakama, Y., Mishima, K., Yamamoto, M.,
2004. GBP-5 splicing variants: new guanylate-binding proteins with tumor-associated Watanabe, T., Ueyama, M., Funaoka, Y., Azuma, S., Watanabe, M., 2009. Antiviral
expression and antigenicity. J. Invest. Dermatol. 122 (6), 1510–1517. effects of the interferon-induced protein guanylate binding protein 1 and its inter-
Feng, C.G., Weksberg, D.C., Taylor, G.A., Sher, A., Goodell, M.A., 2008. The p47 GTPase action with the hepatitis C virus NS5B protein. Hepatology 50 (6), 1727–1737.
Lrg-47(Irgm1) links host defense and hematopoietic stem cell proliferation. Cell Stem Johnston, A.C., Piro, A., Clough, B., Siew, M., Virreira Winter, S., Coers, J., Frickel, E.-M.,
Cell 2 (1), 83–89. 2016. Human GBP1 does not localize to pathogen vacuoles but restricts Toxoplasma
Feng, J., Cao, Z., Wang, L., Wan, Y., Peng, N., Wang, Q., Chen, X., Zhou, Y., Zhu, Y., 2017. gondii. Cell. Microbiol. 18 (8), 1056–1064.
Inducible GBP5 mediates the antiviral response via interferon-Related pathways Jutras, I., Houde, M., Currier, N., Boulais, J., Duclos, S., LaBoissiere, S., Bonneil, E.,
during influenza a virus infection. J. Innate Immun. 9 (4), 419–435. Kearney, P., Thibault, P., Paramithiotis, E., Hugo, P., Desjardins, M., 2008.
Fentress, S.J., Behnke, M.S., Dunay, I.R., Mashayekhi, M., Rommereim, L.M., Fox, B.A., Modulation of the phagosome proteome by interferon-gamma. Mol. Cell. Proteomics
Bzik, D.J., Taylor, G.A., Turk, B.E., Lichti, C.F., Townsend, R.R., Qiu, W., Hui, R., 7 (4), 697–715.
Beatty, W.L., Sibley, L.D., 2010. Phosphorylation of immunity-related GTPases by a Khaminets, A., Hunn, J.P., Konen-Waisman, S., Zhao, Y.O., Preukschat, D., Coers, J.,
Toxoplasma gondii-secreted kinase promotes macrophage survival and virulence. Boyle, J.P., Ong, Y.C., Boothroyd, J.C., Reichmann, G., Howard, J.C., 2010.
Cell Host Microbe 8 (6), 484–495. Coordinated loading of IRG resistance GTPases on to the Toxoplasma gondii para-
Fiegl, D., Kägebein, D., Liebler-Tenorio, E.M., Weisser, T., Sens, M., Gutjahr, M., Knittler, sitophorous vacuole. Cell. Microbiol. 12 (7), 939–961.
M.R., 2013. 2013 Amphisomal route of MHC class I cross-presentation in bacteria- Kim, B.-H., Shenoy, A.R., Kumar, P., Das, R., Tiwari, S., MacMicking, J.D., 2011. A family
infected dendritic cells. J. Immunol. 190 (6), 2791–2806. of IFN- −Inducible 65-kD GTPases protects against bacterial infection. Science 332
Finethy, R., Jorgensen, I., Haldar, A.K., Zoete de, M.R., Strowig, T., Flavell, R.A., (6030), 717–721.
Yamamoto, M., Nagarajan, U.M., Miao, E.A., Coers, J., 2015. Guanylate binding Kim, B.-H., Chee, J.D., Bradfield, C.J., Park, E.-S., Kumar, P., MacMicking, J.D., 2016.

243
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

Interferon-induced guanylate-binding proteins in inflammasome activation and host establishes a moving junction that selectively excludes host cell plasma membrane
defense. Nat. Immunol. 17 (5), 481–489. proteins on the basis of their membrane anchoring. J. Exp. Med. 190 (12),
King, K.Y., Baldridge, M.T., Weksberg, D.C., Chambers, S.M., Lukov, G.L., Wu, S., Boles, 1783–1792.
N.C., Jung, S.Y., Qin, J., Liu, D., Songyang, Z., Eissa, N.T., Taylor, G.A., Goodell, Nantais, D.E., Schwemmle, M., Stickney, J.T., Vestal, D.J., Buss, J.E., 1996. Prenylation of
M.A., 2011. Irgm1 protects hematopoietic stem cells by negative regulation of IFN an interferon-gamma-induced GTP-binding protein: the human guanylate binding
signaling. Blood 118 (6), 1525–1533. protein, huGBP1. J. Leukoc. Biol. 60 (3), 423–431.
Koltes, J.E., Fritz-Waters, E., Eisley, C.J., Choi, I., Bao, H., Kommadath, A., Serão, N.V.L., Naschberger, E., Geißdörfer, W., Bogdan, C., Tripal, P., Kremmer, E., Stürzl, M., Britzen-
Boddicker, N.J., Abrams, S.M., Schroyen, M., Loyd, H., Tuggle, C.K., Plastow, G.S., Laurent, N., 2017. Processing and secretion of guanylate binding protein-1 depend on
Guan, L., Stothard, P., Lunney, J.K., Liu, P., Carpenter, S., Rowland, R.R.R., Dekkers, inflammatory caspase activity. J. Cell. Mol. Med. 21 (9), 1954–1966.
J.C.M., Reecy, J.M., 2015. Identification of a putative quantitative trait nucleotide in Neun, R., Richter, M.F., Staeheli, P., Schwemmle, M., 1996. GTPase properties of the
guanylate binding protein 5 for host response to PRRS virus infection. BMC Genomics interferon-induced human guanylate-binding protein 2. FEBS Lett. 390 (1), 69–72.
16, 412. Niedelman, W., Gold, D.A., Rosowski, E.E., Sprokholt, J.K., Lim, D., Farid Arenas, A.,
Kommadath, A., Bao, H., Choi, I., Reecy, J.M., Koltes, J.E., Fritz-Waters, E., Eisley, C.J., Melo, M.B., Spooner, E., Yaffe, M.B., Saeij, J.P.J., 2012. The rhoptry proteins ROP18
Grant, J.R., Rowland, R.R.R., Tuggle, C.K., Dekkers, J.C.M., Lunney, J.K., Le Guan, L., and ROP5 mediate Toxoplasma gondii evasion of the murine, but not the human,
Stothard, P., Plastow, G.S., 2017. Genetic architecture of gene expression underlying interferon-gamma response. PLoS Pathog. 8 (6), e1002784.
variation in host response to porcine reproductive and respiratory syndrome virus Niedelman, W., Sprokholt, J.K., Clough, B., Frickel, E.-M., Saeij, J.P.J., 2013. Cell death of
infection. Sci. Rep. 7, 46203. gamma interferon-stimulated human fibroblasts upon Toxoplasma gondii infection
Krapp, C., Hotter, D., Gawanbacht, A., McLaren, P.J., Kluge, S.F., Stürzel, C.M., Mack, K., induces early parasite egress and limits parasite replication. Infect. Immun. 81 (12),
Reith, E., Engelhart, S., Ciuffi, A., Hornung, V., Sauter, D., Telenti, A., Kirchhoff, F., 4341–4349.
2016. Guanylate binding protein (GBP) 5 is an interferon-Inducible inhibitor of HIV-1 Niu, P., Shabir, N., Khatun, A., Seo, B.-J., Gu, S., Lee, S.-M., Lim, S.-K., Kim, K.-S., Kim,
infectivity. Cell Host Microbe 19 (4), 504–514. W.-I., 2016. Effect of polymorphisms in the GBP1, Mx1 and CD163 genes on host
Kravets, E., Degrandi, D., Weidtkamp-Peters, S., Ries, B., Konermann, C., Felekyan, S., responses to PRRSV infection in pigs. Vet. Microbiol. 182, 187–195.
Dargazanli, J.M., Praefcke, G.J.K., Seidel, C.A.M., Schmitt, L., Smits, S.H.J., Pfeffer, Nordmann, A., Wixler, L., Boergeling, Y., Wixler, V., Ludwig, S., 2012. A new splice
K., 2012. The GTPase activity of murine guanylate-binding protein 2 (mGBP2) con- variant of the human guanylate-binding protein 3 mediates anti-influenza activity
trols the intracellular localization and recruitment to the parasitophorous vacuole of through inhibition of viral transcription and replication. FASEB J. 26 (3), 1290–1300.
Toxoplasma gondii. J. Biol. Chem. 287 (33), 27452–27466. Obar, R.A., Collins, C.A., Hammarback, J.A., Shpetner, H.S., Vallee, R.B., 1990. Molecular
Kravets, E., Degrandi, D., Ma, Q., Peulen, T.-O., Klümpers, V., Felekyan, S., Kühnemuth, cloning of the microtubule-associated mechanochemical enzyme dynamin reveals
R., Weidtkamp-Peters, S., Am Seidel, C., Pfeffer, K., 2016. Guanylate binding proteins homology with a new family of GTP-binding proteins. Nature 347 (6290), 256–261.
directly attack Toxoplasma gondii via supramolecular complexes. eLife 5. Ohshima, J., Lee, Y., Sasai, M., Saitoh, T., Su Ma, J., Kamiyama, N., Matsuura, Y., Pann-
Kresse, A., Konermann, C., Degrandi, D., Beuter-Gunia, C., Wuerthner, J., Pfeffer, K., Beer, Ghill, S., Hayashi, M., Ebisu, S., Takeda, K., Akira, S., Yamamoto, M., 2014. Role of
S., 2008. Analyses of murine GBP homology clusters based on in silico, in vitro and in mouse and human autophagy proteins in IFN-γ-induced cell-autonomous responses
vivo studies. BMC Genomics 9, 158. against Toxoplasma gondii. J. Immunol. 192 (7), 3328–3335.
Kunzelmann, S., Praefcke, G.J.K., Herrmann, C., 2006. Transient kinetic investigation of Ohshima, J., Sasai, M., Liu, J., Yamashita, K., Ma, J.S., Lee, Y., Bando, H., Howard, J.C.,
GTP hydrolysis catalyzed by interferon-gamma-induced hGBP1 (human guanylate Ebisu, S., Hayashi, M., Takeda, K., Standley, D.M., Frickel, E.-M., Yamamoto, M.,
binding protein 1). J. Biol. Chem. 281 (39), 28627–28635. 2015. RabGDIα is a negative regulator of interferon-γ-inducible GTPase-dependent
Lafuse, W.P., Brown, D., Castle, L., Zwilling, B.S., 1995. Cloning and characterization of a cell-autonomous immunity to Toxoplasma gondii. Proc. Natl. Acad. Sci. U. S. A. 112
novel cDNA that is IFN-gamma-induced in mouse peritoneal macrophages and en- (33), E4581–90.
codes a putative GTP-binding protein. J. Leukoc. Biol. 57 (3), 477–483. Olszewski, M.A., Gray, J., Vestal, D.J., 2006. In silico genomic analysis of the human and
Lee, Y., Sasai, M., Ma, J.S., Sakaguchi, N., Ohshima, J., Bando, H., Saitoh, T., Akira, S., murine guanylate-binding protein (GBP) gene clusters. J. Interferon Cytokine Res. 26
Yamamoto, M., 2015. P62 plays a specific role in interferon-γ-Induced presentation of (5), 328–352.
a toxoplasma vacuolar antigen. Cell Rep. 13 (2), 223–233. Ostler, N., Britzen-Laurent, N., Liebl, A., Naschberger, E., Lochnit, G., Ostler, M., Forster,
Lew, D.J., Decker, T., Strehlow, I., Darnell, J.E., 1991. Overlapping elements in the F., Kunzelmann, P., Ince, S., Supper, V., Praefcke, G.J.K., Schubert, D.W., Stockinger,
guanylate-binding protein gene promoter mediate transcriptional induction by alpha H., Herrmann, C., Stürzl, M., 2014. Gamma interferon-induced guanylate binding
and gamma interferons. Mol. Cell. Biol. 11 (1), 182–191. protein 1 is a novel actin cytoskeleton remodeling factor. Mol. Cell. Biol. 34 (2),
Li, G., Zhang, J., Sun, Y., Wang, H., Wang, Y., 2009. The evolutionarily dynamic IFN- 196–209.
inducible GTPase proteins play conserved immune functions in vertebrates and ce- Pan, W., Zuo, X., Feng, T., Shi, X., Dai, J., 2012. Guanylate-binding protein 1 participates
phalochordates. Mol. Biol. Evol. 26 (7), 1619–1630. in cellular antiviral response to dengue virus. Virol. J. 9, 292.
Li, L.-F., Yu, J., Li, Y., Wang, J., Li, S., Zhang, L., Xia, S.-L., Yang, Q., Wang, X., Yu, S., Luo, Pandita, E., Rajan, S., Rahman, S., Mullick, R., Das, S., Sau, A.K., 2016. Tetrameric as-
Y., Sun, Y., Zhu, Y., Munir, M., Qiu, H.-J., 2016. Guanylate-Binding protein 1, an sembly of hGBP1 is crucial for both stimulated GMP formation and antiviral activity.
interferon-Induced GTPase, exerts an antiviral activity against classical swine fever Biochem. J. 473 (12), 1745–1757.
virus depending on its GTPase activity. J. Virol. 90 (9), 4412–4426. Papic, N., Hunn, J.P., Pawlowski, N., Zerrahn, J., Howard, J.C., 2008. Inactive and active
Lilue, J., Müller, U.B., Steinfeldt, T., Howard, J.C., 2013. Reciprocal virulence and re- states of the interferon-inducible resistance GTPase, Irga6, in vivo. J. Biol. Chem. 283
sistance polymorphism in the relationship between Toxoplasma gondii and the house (46), 32143–32151.
mouse. eLife 2, e01298. Pawlowski, N., Khaminets, A., Hunn, J.P., Papic, N., Schmidt, A., Uthaiah, R.C., Lange, R.,
Lindenberg, V., Mölleken, K., Kravets, E., Stallmann, S., Hegemann, J.H., Degrandi, D., Vopper, G., Martens, S., Wolf, E., Howard, J.C., 2011. The activation mechanism of
Pfeffer, K., 2017. Broad recruitment of mGBP family members to Chlamydia tra- Irga6, an interferon-inducible GTPase contributing to mouse resistance against
chomatis inclusions. PLoS One 12 (9), e0185273. Toxoplasma gondii. BMC Biol. 9, 7.
MacMicking, J.D., 2012. Interferon-inducible effector mechanisms in cell-autonomous Pfefferkorn, E.R., Rebhun, S., Eckel, M., 1986. Characterization of an indoleamine 2,3-
immunity. Nature reviews. Immunology 12 (5), 367–382. dioxygenase induced by gamma-interferon in cultured human fibroblasts. J.
Man, S.M., Karki, R., Sasai, M., Place, D.E., Kesavardhana, S., Temirov, J., Frase, S., Zhu, Interferon Res. 6 (3), 267–279.
Q., Malireddi, R.K.S., Kuriakose, T., Peters, J.L., Neale, G., Brown, S.A., Yamamoto, Pfefferkorn, E.R., 1984. Interferon gamma blocks the growth of Toxoplasma gondii in
M., Kanneganti, T.-D., 2016. IRGB10 liberates bacterial ligands for sensing by the human fibroblasts by inducing the host cells to degrade tryptophan. Proc. Natl. Acad.
AIM2 and caspase-11-NLRP3 inflammasomes. Cell 167 (2), 382–396 (e17). Sci. U. S. A. 81 (3), 908–912.
Maric-Biresev, J., Hunn, J.P., Krut, O., Helms, J.B., Martens, S., Howard, J.C., 2016. Loss Pilla, D.M., Hagar, J.A., Haldar, A.K., Mason, A.K., Degrandi, D., Pfeffer, K., Ernst, R.K.,
of the interferon-γ-inducible regulatory immunity-related GTPase (IRG), Irgm1, Yamamoto, M., Miao, E.A., Coers, J., 2014. Guanylate binding proteins promote
causes activation of effector IRG proteins on lysosomes, damaging lysosomal function caspase-11-dependent pyroptosis in response to cytoplasmic LPS. Proc. Natl. Acad.
and predicting the dramatic susceptibility of Irgm1-deficient mice to infection. BMC Sci. U. S. A. 111 (16), 6046–6051.
Biol. 14, 33. Pilla-Moffett, D., Barber, M.F., Taylor, G.A., Coers, J., 2016. Interferon-Inducible GTPases
Martens, S., Sabel, K., Lange, R., Uthaiah, R., Wolf, E., Howard, J.C., 2004. Mechanisms in host resistance, inflammation and disease. J. Mol. Biol. 428 (17), 3495–3513.
regulating the positioning of mouse p47 resistance GTPases LRG-47 and IIGP1 on Platanias, L.C., 2005. Mechanisms of type-I- and type-II-interferon-mediated signalling.
cellular membranes: retargeting to plasma membrane induced by phagocytosis. J. Nat. Rev. Immunol. 5 (5), 375–386.
Immunol. 173 (4), 2594–2606. Praefcke, G.J.K., McMahon, H.T., 2004. The dynamin superfamily: universal membrane
McNew, J.A., Sondermann, H., Lee, T., Stern, M., Brandizzi, F., 2013. GTP-dependent tubulation and fission molecules? Nat. Rev. Mol. Cell Biol. 5 (2), 133–147.
membrane fusion. Annu. Rev. Cell Dev. Biol. 29, 529–550. Praefcke, G.J.K., Geyer, M., Schwemmle, M., Robert Kalbitzer, H., Herrmann, C., 1999.
Meunier, E., Broz, P., 2015. Interferon-induced guanylate-binding proteins promote cy- Nucleotide-binding characteristics of human guanylate-binding protein 1 (hGBP1)
tosolic lipopolysaccharide detection by caspase-11. DNA Cell Biol. 34 (1), 1–5. and identification of the third GTP-binding motif. J. Mol. Biol. 292 (2), 321–332.
Meunier, E., Broz, P., 2016. Interferon-inducible GTPases in cell autonomous and innate Praefcke, G.J.K., Kloep, S., Benscheid, U., Lilie, H., Prakash, B., Herrmann, C., 2004.
immunity. Cell. Microbiol. 18 (2), 168–180. Identification of residues in the human guanylate-binding protein 1 critical for nu-
Meunier, E., Dick, M.S., Dreier, R.F., Schürmann, N., Kenzelmann Broz, D., Warming, S., cleotide binding and cooperative GTP hydrolysis. J. Mol. Biol. 344 (1), 257–269.
Roose-Girma, M., Bumann, D., Kayagaki, N., Takeda, K., Yamamoto, M., Broz, P., Prakash, B., Praefcke, G.J.K., Renault, L., Wittinghofer, A., Herrmann, C., 2000a.
2014. Caspase-11 activation requires lysis of pathogen-containing vacuoles by IFN- Structure of human guanylate-binding protein 1 representing a unique class of GTP-
induced GTPases. Nature 509 (7500), 366–370. binding proteins. Nature 403 (6769), 567–571.
Modiano, N., Lu, Y.E., Cresswell, P., 2005. Golgi targeting of human guanylate-binding Prakash, B., Renault, L., Praefcke, G.J.K., Herrmann, C., Wittinghofer, A., 2000b.
protein-1 requires nucleotide binding, isoprenylation, and an IFN-gamma-inducible Triphosphate structure of guanylate-binding protein 1 and implications for nucleo-
cofactor. Proc. Natl. Acad. Sci. U. S. A. 102 (24), 8680–8685. tide binding and GTPase mechanism. EMBO J. 19 (17), 4555–4564.
Mordue, D.G., Desai, N., Dustin, M., Sibley, L.D., 1999. Invasion by Toxoplasma gondii Qin, A., Lai, D.-H., Liu, Q., Huang, W., Wu, Y.-P., Chen, X., Yan, S., Xia, H., Hide, G., Lun,

244
G.J.K. Praefcke International Journal of Medical Microbiology 308 (2018) 237–245

Z.-R., Ayala, F.J., Xiang, A.P., 2017. Guanylate-binding protein 1 (GBP1) contributes Pfeffer, K., Coers, J., Taylor, G.A., 2011. Immunity-related GTPase M (IRGM) proteins
to the immunity of human mesenchymal stromal cells against Toxoplasma gondii. influence the localization of guanylate-binding protein 2 (GBP2) by modulating
Proc. Natl. Acad. Sci. U. S. A. 114 (6), 1365–1370. macroautophagy. J. Biol. Chem. 286 (35), 30471–30480.
Scharton-Kersten, T.M., Yap, G., Magram, J., Sher, A., 1997. Inducible nitric oxide is Tripal, P., Bauer, M., Naschberger, E., Mortinger, T., Hohenadl, C., Cornali, E., Thurau,
essential for host control of persistent but not acute infection with the intracellular M., Sturzl, M., 2007. Unique features of different members of the human guanylate-
pathogen Toxoplasma gondii. J. Exp. Med. 185 (7), 1261–1273. binding protein family. J. Interferon Cytokine Res. 27 (1), 44–52.
Schnoor, M., Betanzos, A., Weber, D.A., Parkos, C.A., 2009. Guanylate-binding protein-1 Trost, M., English, L., Lemieux, S., Courcelles, M., Desjardins, M., Thibault, P., 2009. The
is expressed at tight junctions of intestinal epithelial cells in response to interferon- phagosomal proteome in interferon-gamma-activated macrophages. Immunity 30
gamma and regulates barrier function through effects on apoptosis. Mucosal (1), 143–154.
Immunol. 2 (1), 33–42. Tyrkalska, S.D., Candel, S., Angosto, D., Gómez-Abellán, V., Martín-Sánchez, F., García-
Schroyen, M., Eisley, C., Koltes, J.E., Fritz-Waters, E., Choi, I., Plastow, G.S., Guan, L., Moreno, D., Zapata-Pérez, R., Sánchez-Ferrer, Á., Sepulcre, M.P., Pelegrín, P., Mulero,
Stothard, P., Bao, H., Kommadath, A., Reecy, J.M., Lunney, J.K., Rowland, R.R.R., V., 2016. Neutrophils mediate Salmonella Typhimurium clearance through the GBP4
Dekkers, J.C.M., Tuggle, C.K., 2016. Bioinformatic analyses in early host response to inflammasome-dependent production of prostaglandins. Nat. Commun. 7, 12077.
Porcine Reproductive and Respiratory Syndrome virus (PRRSV) reveals pathway Uthaiah, R.C., Praefcke, G.J.K., Howard, J.C., Herrmann, C., 2003. IIGP1, an interferon-
differences between pigs with alternate genotypes for a major host response QTL. gamma-inducible 47-kDa GTPase of the mouse, showing cooperative enzymatic ac-
BMC Genomics 17, 196. tivity and GTP-dependent multimerization. J. Biol. Chem. 278 (31), 29336–29343.
Schulte, K., Pawlowski, N., Faelber, K., Fröhlich, C., Howard, J., Daumke, O., 2016. The Vöpel, T., Kunzelmann, S., Herrmann, C., 2009. Nucleotide dependent cysteine reactivity
immunity-related GTPase Irga6 dimerizes in a parallel head-to-head fashion. BMC of hGBP1 uncovers a domain movement during GTP hydrolysis. FEBS Lett. 583 (12),
Biol. 14, 14. 1923–1927.
Schwemmle, M., Staeheli, P., 1994. The interferon-induced 67-kDa guanylate-binding Vöpel, T., Syguda, A., Britzen-Laurent, N., Kunzelmann, S., Lüdemann, M.-B.,
protein (hGBP1) is a GTPase that converts GTP to GMP. J. Biol. Chem. 269 (15), Dovengerds, C., Stürzl, M., Herrmann, C., 2010. Mechanism of GTPase-activity-in-
11299–11305. duced self-assembly of human guanylate binding protein 1. J. Mol. Biol. 400 (1),
Selleck, E.M., Fentress, S.J., Beatty, W.L., Degrandi, D., Pfeffer, K., Virgin, H.W., 63–70.
MacMicking, J.D., Sibley, L.D., 2013. Guanylate-binding protein 1 (Gbp1) contributes Vöpel, T., Hengstenberg, C.S., Peulen, T.-O., Ajaj, Y., Seidel, C.A.M., Herrmann, C., Klare,
to cell-autonomous immunity against Toxoplasma gondii. PLoS Pathog. 9 (4), J.P., 2014. Triphosphate induced dimerization of human guanylate binding protein 1
e1003320. involves association of the C-terminal helices: a joint double electron–electron re-
Shenoy, A.R., Wellington, D.A., Kumar, P., Kassa, H., Booth, C.J., Cresswell, P., sonance and FRET study. Biochemistry 53 (28), 4590–4600.
MacMicking, J.D., 2012. GBP5 promotes NLRP3 inflammasome assembly and im- Vestal, D.J., Gorbacheva, V.Y., Sen, G.C., 2000. Different subcellular localizations for the
munity in mammals. Science 336 (6080), 481–485. related interferon-induced GTPases, MuGBP-1 and MuGBP-2: implications for dif-
Shydlovskyi, S., Zienert, A.Y., Ince, S., Dovengerds, C., Hohendahl, A., Dargazanli, J.M., ferent functions? Journal Interferon Cytokine Res. 20 (11), 991–1000.
Blum, A., Günther, S.D., Kladt, N., Stürzl, M., Schauss, A.C., Kutsch, M., Roux, A., Virreira Winter, S., Niedelman, W., Jensen, K.D., Rosowski, E.E., Julien, L., Spooner, E.,
Praefcke, G.J.K., Herrmann, C., 2017. Nucleotide-dependent farnesyl switch orches- Caradonna, K., Burleigh, B.A., Saeij, J.P.J., Ploegh, H.L., Frickel, E.-M., 2011.
trates polymerization and membrane binding of human guanylate-binding protein 1. Determinants of GBP recruitment to Toxoplasma gondii vacuoles and the parasitic
Proc. Natl. Acad. Sci. U. S. A. 114 (28), E5559–E5568. factors that control it. PLoS One 6 (9), e24434.
Sorace, J.M., Johnson, R.J., Howard, D.L., Drysdale, B.E., 1995. Identification of an en- Wallet, P., Benaoudia, S., Mosnier, A., Lagrange, B., Martin, A., Lindgren, H., Golovliov,
dotoxin and IFN-inducible cDNA: possible identification of a novel protein family. J. I., Michal, F., Basso, P., Djebali, S., Provost, A., Allatif, O., Meunier, E., Broz, P.,
Leukoc. Biol. 58 (4), 477–484. Yamamoto, M., Py, B.F., Faudry, E., Sjöstedt, A., Henry, T., 2017. IFN-γ extends the
Steinfeldt, T., Könen-Waisman, S., Tong, L., Pawlowski, N., Lamkemeyer, T., Sibley, L.D., immune functions of Guanylate Binding Proteins to inflammasome-independent an-
Hunn, J.P., Howard, J.C., 2010. Phosphorylation of mouse immunity-related GTPase tibacterial activities during Francisella novicida infection. PLoS Pathog. 13 (10),
(IRG) resistance proteins is an evasion strategy for virulent Toxoplasma gondii. PLoS e1006630.
Biol. 8 (12), e1000576. Wandel, M.P., Pathe, C., Werner, E.I., Ellison, C.J., Boyle, K.B., Malsburg, A., von der
Stickney, J.T., Buss, J.E., 2000. Murine guanylate-binding protein: incomplete ger- Rohde, J., Randow, F., 2017. GBPs inhibit motility of shigella flexneri but are tar-
anylgeranyl isoprenoid modification of an interferon-gamma-inducible guanosine geted for degradation by the bacterial ubiquitin ligase IpaH9. 8. Cell Host Microbe 22
triphosphate-binding protein. Mol. Biol. Cell 11 (7), 2191–2200. (4), 507–518 (e5).
Syguda, A., Bauer, M., Benscheid, U., Ostler, N., Naschberger, E., Ince, S., Stürzl, M., Wehner, M., Herrmann, C., 2010. Biochemical properties of the human guanylate binding
Herrmann, C., 2012a. Tetramerization of human guanylate-binding protein 1 is protein 5 and a tumor-specific truncated splice variant. FEBS J. 277 (7), 1597–1605.
mediated by coiled-coil formation of the C-terminal α-helices. FEBS J. 279 (14), Wehner, M., Kunzelmann, S., Herrmann, C., 2012. The guanine cap of human guanylate-
2544–2554. binding protein 1 is responsible for dimerization and self-activation of GTP hydro-
Syguda, A., Kerstan, A., Ladnorg, T., Stüben, F., Wöll, C., Herrmann, C., 2012b. lysis. FEBS J. 279 (2), 203–210.
Immobilization of biotinylated hGBP1 in a defined orientation on surfaces is crucial Yamamoto, M., Okuyama, M., Ma, J.S., Kimura, T., Kamiyama, N., Saiga, H., Ohshima, J.,
for uniform interaction with analyte proteins and catalytic activity. Langmuir 28 Sasai, M., Kayama, H., Okamoto, T., Huang, D.C.S., Soldati-Favre, D., Horie, K.,
(15), 6411–6418. Takeda, J., Takeda, K., 2012. A cluster of interferon-γ-inducible p65 GTPases plays a
Taylor, G.A., Jeffers, M., Largaespada, D.A., Jenkins, N.A., Copeland, N.G., Vande Woude, critical role in host defense against Toxoplasma gondii. Immunity 37 (2), 302–313.
G.F., 1996. Identification of a novel GTPase, the inducibly expressed GTPase, that Yarovinsky, F., 2014. Innate immunity to Toxoplasma gondii infection. Nature reviews.
accumulates in response to interferon gamma. J. Biol. Chem. 271 (34), 20399–20405. Immunology 14 (2), 109–121.
Taylor, G.A., Collazo, C.M., Yap, G.S., Nguyen, K., Gregorio, T.A., Taylor, L.S., Eagleson, Zhao, Z., Fux, B., Goodwin, M., Dunay, I.R., Strong, D., Miller, B.C., Cadwell, K., Delgado,
B., Secrest, L., Southon, E.A., Reid, S.W., Tessarollo, L., Bray, M., McVicar, D.W., M.A., Ponpuak, M., Green, K.G., Schmidt, R.E., Mizushima, N., Deretic, V., Sibley,
Komschlies, K.L., Young, H.A., Biron, C.A., Sher, A., Vande Woude, G.F., 2000. L.D., Virgin, H.W., 2008. Autophagosome-independent essential function for the
Pathogen-specific loss of host resistance in mice lacking the IFN-gamma-inducible autophagy protein Atg5 in cellular immunity to intracellular pathogens. Cell Host
gene IGTP. Proc. Natl. Acad. Sci. U. S. A. 97 (2), 751–755. Microbe 4 (5), 458–469.
Thurston, T.L.M., Wandel, M.P., Muhlinen, N., von Foeglein, A., Randow, F., 2012. Zhao, Y.O., Khaminets, A., Hunn, J.P., Howard, J.C., 2009. Disruption of the Toxoplasma
Galectin 8 targets damaged vesicles for autophagy to defend cells against bacterial gondii parasitophorous vacuole by IFNgamma-inducible immunity-related GTPases
invasion. Nature 482 (7385), 414–418. (IRG proteins) triggers necrotic cell death. PLoS Pathog. 5 (2), e1000288.
Tietzel, I., El-Haibi, C., Carabeo, R.A., 2009. Human guanylate binding proteins po- Zou, Z., Meng, Z., Ma, C., Liang, D., Sun, R., Lan, K., 2017. Guanylate-Binding protein 1
tentiate the anti-chlamydia effects of interferon-gamma. PLoS One 4 (8), e6499. inhibits nuclear delivery of kaposi's sarcoma-associated herpesvirus virions by dis-
Traver, M.K., Henry, S.C., Cantillana, V., Oliver, T., Hunn, J.P., Howard, J.C., Beer, S., rupting formation of actin filament. J. Virol. 91 (16).

245

You might also like