ArticlePDF Available

The SKPO-1 Peroxidase Functions in the Hypodermis to Protect Caenorhabditis elegans From Bacterial Infection

Authors:

Abstract and Figures

In recent years, the synergistic relationship between NADPH Oxidases (NOX)/Dual Oxidases (DUOX) enzymes and peroxidases has received increased attention. Peroxidases utilize NOX/DUOX-generated H2O2 for a myriad of functions including, but not limited to thyroid hormone biosynthesis, cross-linking extracellular matrices (ECM) and immune defense. We postulated that one or more peroxidases produced by Caenorhabditis elegans would act in host defense, possibly in conjunction with BLI-3, the only NOX/DUOX enzyme encoded by the genome that is expressed. Animals exposed to RNAi of the putative peroxidase genes were screened for susceptibility to the human pathogen Enterococcus faecalis. One of three genes identified, skpo-1 (ShkT-containing peroxidase), was studied in-depth. Animals mutant for this gene were significantly more susceptible to E. faecalis, but not Pseudomonas aeruginosa. A slight decrease in longevity was also observed. The skpo-1 mutant animals had a dumpy phenotype of incomplete penetrance; half the animals displayed a dumpy phenotype ranging from slight to severe, and half were morphologically wild type. The SKPO-1 protein contains the critical catalytic residues necessary for peroxidase activity, and in a whole animal assay more H2O2 was detected from the mutant compared to the wild type, consistent with the loss of an H2O2 sink. By using tissue-specific skpo-1 RNAi and immunohistochemical localization with an anti-SKPO-1 antibody, it was determined that the peroxidase is functionally and physically present in the hypodermis. In conclusion, these results characterize a peroxidase that functions protectively in the hypodermis during exposure to E. faecalis.
Content may be subject to copyright.
GENETICS OF IMMUNITY
The SKPO-1 Peroxidase Functions in the Hypodermis
to Protect Caenorhabditis elegans From
Bacterial Infection
George R. Tiller and Danielle A. Garsin
1
Department of Microbiology and Molecular Genetics, University of Texas Health Science Center and Graduate School of Biomedical
Sciences, Houston, Texas 77030
ABSTRACT In recent years, the synergistic relationship between NADPH oxidase (NOX)/dual oxidase (DUOX) enzymes and peroxidases
has received increased attention. Peroxidases utilize NOX/DUOX-generated H
2
O
2
for a myriad of functions including, but not limited to,
thyroid hormone biosynthesis, cross-linking extracellular matrices (ECM), and immune defense. We postulated that one or more
peroxidases produced by Caenorhabditis elegans would act in host defense, possibly in conjunction with BLI-3, the only NOX/DUOX
enzyme encoded by the genome that is expressed. Animals exposed to RNA interference (RNAi) of the putative peroxidase genes were
screened for susceptibility to the human pathogen Enterococcus faecalis. One of three genes identied, skpo-1 (ShkT-containing
peroxidase), was studied in depth. Animals mutant for this gene were signicantly more susceptible to E. faecalis, but not Pseudo-
monas aeruginosa. A slight decrease in longevity was also observed. The skpo-1 mutant animals had a dumpy phenotype of in-
complete penetrance; half the animals displayed a dumpy phenotype ranging from slight to severe, and half were morphologically wild
type. The SKPO-1 protein contains the critical catalytic residues necessary for peroxidase activity, and in a whole animal assay, more
H
2
O
2
was detected from the mutant compared to the wild type, consistent with the loss of an H
2
O
2
sink. By using tissue-specicskpo-
1RNAi and immunohistochemical localization with an anti-SKPO-1 antibody, it was determined that the peroxidase is functionally and
physically present in the hypodermis. In conclusion, these results characterize a peroxidase that functions protectively in the hypodermis
during exposure to E. faecalis.
HEME-containing peroxidases play critical, wide-ranging
roles in biological systems. Once called the animal
heme peroxidases,members of the peroxidasecyclooxgenase
superfamily are actually found in all kingdoms of life (Zamocky
et al. 2008). The roles of most are poorly characterized, but
some are clearly involved in immune defense. For example,
the most famous and best-studied member of this group,
myeloperoxidase (MPO), is found in the granulocytes of
neutrophils where it catalyzes the formation of the potent
oxidant HOCl from H
2
O
2
and Cl
2
to kill invading microbes
(reviewed by Klebanoff 2005). Another is lactoperoxidase
(LPO), which is found on mucosal surfaces and generates
the protective oxidant hypothiocyanite (OSCN
2
)from
H
2
O
2
and thiocyanate (SCN
2
).Thisprocessisimpaired
in the lungs of patients with cystic brosis (CF), contributing
to the poor clearance of pathogens such as Pseudomonas
aeruginosa and Staphylococcus aureus, the most common
causes of lung infection in patients with CF (Conner et al.
2002; Geiszt et al. 2003; Forteza et al. 2005; Moskwa et al.
2007).
Many of these peroxidases are functionally associated
with members of the NADPH oxidase (NOX)/dual oxidase
(DUOX) family of proteins. These enzymes generate the
H
2
O
2
required as substrate for the peroxidases. For example,
Nox2 is the source of H
2
O
2
for MPO. DUOXs differ from
NOXs in that they encode a peroxidase domain in addition
to the oxidant-generating NADPH oxidase domain (reviewed
by Rada et al. 2008; Sumimoto 2008). Despite the fact that
they have a peroxidase domain, DUOXs appear to still asso-
ciate with separate peroxidases. For example, LPO utilizes
H
2
O
2
generated by Duox1 or Duox2 (Conner et al. 2002;
Geiszt et al. 2003; Forteza et al. 2005). In fact, the peroxidase
Copyright © 2014 by the Genetics Society of America
doi: 10.1534/genetics.113.160606
Manuscript received December 10, 2013; accepted for publication March 9, 2014;
published Early Online March 12, 2014.
Supporting information is available online at http://www.genetics.org/lookup/suppl/
doi:10.1534/genetics.113.160606/-/DC1.
1
Corresponding author: University of Texas Health Science Center at Houston,
Department of Microbiology and Molecular Genetics, 6431 Fannin St., MSB 1.168,
Houston, TX 77030. E-mail: Danielle.A.Garsin@uth.tmc.edu
Genetics, Vol. 197, 515526 June 2014 515
domains of human Duox1/2 are reported to lack peroxi-
dase activity and may have other functions, such as serving
as an interaction domain for separate, active peroxidases
(Meitzler and Ortiz de Montellano 2009, 2011; Meitzler
et al. 2013).
Caenorhabditis elegans has been used as a model host to
study various aspects of the innate immune response, in-
cluding the purposeful generation of reactive oxygen spe-
cies (ROS) as a defense mechanism (Chavez et al. 2007,
2009). The animal encodes only one functional NADPH
oxidase, a dual oxidase called BLI-3 (Edens et al. 2001).
When exposed to human pathogens such as the Gram-
positive, opportunistic bacterium, Enterococcus faecalis,H
2
O
2
is released (Chavez et al. 2007). The response appears pro-
tective, as its loss by reducing the expression of bli-3 by
RNA interference (RNAi) renders the animals more sensi-
tive to killing by the pathogen (Chavez et al. 2009). Using
indirect immunouorescence, BLI-3 was localized to the
hypodermis, which is essentially the skinof C. elegans
(Edens et al. 2001). There is also speculation that BLI-3
maybepresentintheintestinalcells(Chavezet al. 2007,
2009). In addition to playing a role in innate immunity,
BLI-3 is essential to the normal development of the worm
because it contributes to the generation of the tyrosine-
linked collagen necessary for proper biogenesis of the cu-
ticle, i.e., the exoskeleton of the animal. Its role appears
twofold. First, it generates the necessary substrate H
2
O
2
,
and then it oxidizes the tyrosines of the collagen proteins
that then combine to form di- and trityrosines, thereby
cross-linking the cuticle (Edens et al. 2001). Unlike human
Duox1/2, the peroxidase domain of BLI-3 has low levels of
peroxidase activity that is essential to this process, and
mutations in the peroxidase domain that disrupt this activ-
ity result in a blistered(bli) phenotype (Brenner 1974;
Simmer et al. 2003; Meitzler and Ortiz de Montellano
2009, 2010, 2011). However, more recent work has dem-
onstrated that the peroxidase domain of BLI-3 is not the
only peroxidase involved in this process. A separate perox-
idase, MLT-7,alsocontributestocuticlecross-linkingand
loss of this activity results in the same bli phenotype as loss
of the BLI-3 peroxidase domain. Additionally, loss of both
peroxidase activities greatly increases the severity of cuti-
cle blistering (Thein et al. 2009).
Because of the prevalent involvement of peroxidases in
immune responses, we hypothesized that C. elegans might
also utilize a peroxidase(s) in host defense, perhaps in con-
junction with its NADPH oxidase, BLI-3. The results of an
earlier investigation indicated that the peroxidase domain of
BLI-3 is not involved, as point mutants in this domain had
wild-type resistance to E. faecalis, despite their blistered phe-
notype (Chavez et al. 2009). In this study, we characterized
the effects of reducing the expression of putative peroxi-
dases encoded by the C. elegans genome on susceptibility
to E. faecalis. We identied three genes whose loss resulted
in susceptibility. One gene, named skpo-1,forShkT-containing
peroxidase, was investigated in depth.
Materials and Methods
Creation of complementary DNA pL4440 constructs for
double-stranded RNA production in Escherichia coli
A standard RNA isolation procedure was used to procure
total RNA from healthy N2 adults grown on Escherichia coli
OP50. We followed the rst-strand complementary DNA
(cDNA) synthesis protocol from the Life Technologies Super-
Script III Synthesis system to produce rst-strand total
cDNA. We then followed a standard PCR protocol using
primers specictoF32A52.A,F32A52.B, and pxn-1, respec-
tively (Supporting Information,Table S5). PCR products
were digested and ligated into pL4440 and transformed into
chemically competent E. coli HT115 to produce double-
stranded RNA (dsRNA) to the gene of interest, respectively.
C. elegans strains and growth conditions
C. elegans strains were grown and maintained as previously
described (Hope 1999). The strains used for the hypodermal
and intestinal RNAi studies are as follows: rde-1(ne219);
Is[wrt-2prom::RDE-1::unc54 39utr;myo2p::RFP3] and sid-
1(qt9); Is[vha-6::sid-1], respectively. The RB1437 [skpo-1
(ok1640) II] strain is a partial deletion mutant for skpo-1
and was veried via sequencing (www.wormbase.org). The
RB1437 [skpo1 (ok1640) II] strain was created by the
C. elegans Reverse Genetics Core Facility at the University of
British Columbia, part of the International C. elegans Gene
Knockout Consortium and obtained through the Caenorhab-
ditis Genetics Center. The independent lines of skpo-1
strains, GF89GF91, were obtained through backcrossing
the RB1437 [skpo-1 (ok1640) II] strain with the wild-type
N2 Bristol strain ve times (backcrossing, BC 35).
RNAi
RNAi was performed by exposing L1L4 stage larvae to
E. coli HT115-expressing dsRNA to target genes. RNAi clones
were obtained from the C. elegans library (Fraser et al. 2000;
Kamath et al. 2003). skpo-1 worms exposed to pathogens
were prone to a maternal bagging phenotype; therefore, cdc-
25.1 RNAi was used to induce sterility in some experiments
(van der Hoeven et al. 2011).
Survival and longevity assays
Unless otherwise indicated, the following bacterial strains
were used: OP50 (E. coli) (Brenner 1974), OG1RF (E. fae-
calis) (Dunny et al. 1978), and PA14 (P. aeruginosa) (Rahme
et al. 1995). Exposure to RNAi, survival assays, and longev-
ity assays were performed as previously described (Garsin
et al. 2001, 2003; Kim et al. 2002; van der Hoeven et al.
2011). Briey, for E. faecalis survival assays, E. faecalis
grown in brain heart infusion (BHI) medium for 5 hr was
seeded (10 ml) onto BHI plates (gentamycin 50 mg/ml) and
incubated at 37°for 24 hr, whereas for P. aeruginosa survival
assays, P. aeruginosa was cultured in Luria broth (LB) over-
night at 37°, seeded (10 ml) onto slow-killing plates, and
incubated rst for 24 hr at 37°and then for the duration of
516 G. R. Tiller and D. A. Garsin
the experiment at 25°. For the E. coli longevity assays, 203
E. coli was seeded (100 ml) onto nematode growth (NG)
medium plates supplemented with 5-Fluoro-29-deoxyuridine
(25 mg/ml) and streptomycin (25 mg/ml). Seeded NG plates
were incubated at 25°for 24 hr. In all assays, a total of 90 L4
larvae were transferred to three replica plates and the assay
was performed at 25°. Worms were scored as live and dead
at various time points.
Amplex Red assay for H
2
O
2
measurements
The Amplex Red hydrogen peroxide/peroxidase kit (Invi-
trogen Molecular Probes, Eugene, OR) was previously
adapted to C. elegans to measure pathogen-stimulated hy-
drogen peroxide release (Chavez et al. 2007, 2009). The
same protocol was followed with the following modica-
tions: L4 worms were exposed to a bacterial strain for 12
hr, and the uorescence of 30 worms per well was measured
after 30 min of incubation with Amplex Red (540/590 nm
excitation and emission, respectively). A total of 80 mM
diphenyleneiodinium chloride (DPI) (TCI, Tokyo) was
added to some wells and allowed to incubate for 15 min
prior to addition of Amplex Red, HRP, and Na
2
PO
4
(pH
7.4), and uorescence was measured as above.
Indirect immunouorescence
Immunouorescence for C. elegans was performed according
to Seydoux and Dunn (1997) on young adult animals. Cus-
tom peptide synthesis, rabbit immunization, and afnity pu-
rication of the SKPO-1 polyclonal antibody was performed
by LifeTein (South Plaineld, NJ). The CRVGRRAFDIENGSR
peptide corresponds to the C terminus of the C. elegans pro-
tein SKPO-1. Adult N2 and GF89 worms were imaged using
an Olympus IX81 automated inverted microscope and Slide-
book (version 5.0) software along with the SKPO-1 poly-
clonal primary antibody and Alexa Fluor 488 goat
antirabbit IgG secondary antibody to localize SKPO-1 in
C. elegans.
Immunogold labeling and transmission
electron microscopy
Approximately 1000 N2 and GF89 animals were raised un-
der standard conditions to the young adult stage. They were
rinsed off of the propagation plates with M9 and spun down
at 2000 rpm for 1 min. The worms were sequentially resus-
pended and pelleted three times in 1 ml M9 (Hope 1999).
After the nal wash, they were resuspended in 1 ml 3%
formalin, 0.15% glutaraldehyde in Millonigs buffer, pH
7.45, and xed for 3 days at 40°. Then the buffer was drawn
off and the samples were incubated in 0.1% fresh made
sodium borohydride in Millonigs buffer (room temperature,
RT, for 10 min). This was followed by incubations in Mill-
onigs buffer (RT, 10 min, repeated twice). The samples
were then dehydrated at room temperature by incubating
in 50% ethanol (10 min, twice) and 70% ethanol, (15 min,
twice). The samples were then permeated with 50% LR
White resin and 50% ethanol for 60 min, followed by an
overnight incubation at room temperature in 50% LR White
resin sealed on a rotator. The samples were then incubated
at room temperature for 4 hr in 70% LR White resin and
then for 5 hr in 100% LR White resin, while on a rotator. The
pellets were then sealed into BEEM capsules and allowed to
polymerize in a 53°oven overnight. Thin sections (120 nm)
of the LR White blocks were cut using a DiATOME diamond
knife and a Leica Ultracut R microtome. Sections were
oated onto 200 mesh nickel grids. The grids were then
oated in 50 mM glycine (RT, 15 min) and then washed
twice by oating in drops of PBS. The samples were blocked
by oating them for 30 min in Aurion Blocking Solution
followed by washes in incubation buffer (PBS with 0.1%
Aurion BSA-c) (5 min, three times). The SKPO-1 antibody
was diluted into the incubation buffer at 1:1000 and the
grids were incubated for 1.5 hr at RT. This was followed
by washes in incubation buffer (10 min, four times) and
then incubation with the secondary antibody (EMS goat
antirabbit IgG with 10 nm gold) diluted 1:20 in incubation
buffer. The samples were washed again in incubation buffer
(10 min, ve times) followed by PBS (10 min, three times).
A postx treatment of 3% glutaraldehyde was applied for
10 min followed by a nal wash with distilled water (5 min,
two times). The grids were dried in a 70°oven and then
imaged using a JEOL 1200 transmission electron microscope
at 60 kV and captured with a 2k 32k Gatan CCD camera.
Statistical analysis
Amplex Red, survival, and longevity assays were analyzed
using GraphPad Prism version 5.0 (GraphPad Software, San
Diego). Students paired t-test was used to determine the
statistical signicance of the Amplex Red data. P-values of
,0.05 were considered to be statistically signicant. Statis-
tically signicant differences are denoted in the gures with
asterisks. KaplanMeier log rank analysis was used to com-
pare survival and longevity curves pairwise and to calculate
the median survival. P-values of ,0.05 were considered to
be statistically signicant.
Results
Host resistance to E. faecalis is affected by skpo-1
To investigate if C. elegans possesses any peroxidases in-
volved in host defense, we utilized an RNAi-killing assay
to screen candidate peroxidase genes. The candidate genes
were found using the BLI-3 peroxidase domain as the BLAST
query on www.wormbase.org. Twelve putative peroxidase-
encoding genes were identied, 9 of which were available in
the RNAi library (Fraser et al. 2000; Kamath et al. 2003),
including the previously studied mlt-7 (Thein et al. 2009).
Standard molecular biology techniques were used to gener-
ate the three missing RNAi constructs (Materials and Meth-
ods). Using a background in which RNAi efciency is
increased (eri-1) (Kennedy et al. 2004), the expression of
these genes was reduced in C. elegans. The animals were
SKPO-1 Protects Against Infection 517
then exposed to E. faecalis and survival was scored over
time. By these means, the genes were screened for possible
roles in innate immune function. The reduced expression of
three genesF09F3.5,R08F11.7, and F49E12.1resulted
in a statistically signicant susceptibility phenotype (Table
1). We decided to focus on F49E12.1, as it possessed high
homology to human myeloperoxidase (hMPO, 34% se-
quence identity) and a deletion mutant was available
through the Caenorhabditis Genetics Center (CGC). As de-
scribed in more detail below, the predicted protein associ-
ated with this gene contains an N-terminal metridin Shk
toxin (ShkT)-like domain and a C-terminal peroxidase do-
main and was therefore named SKPO-1 for ShkT-containing
peroxidase.
An example of the survival of skpo-1 RNAi eri-1 mutant
animals on E. faecalis compared to vector control (VC) RNAi
eri-1 mutant animals is shown in Figure 1A. skpo-1 RNAi
animals displayed an enhanced susceptibility phenotype rel-
ative to VC RNAi (P= 0.0011). The experiment was re-
peated ve times and the data were tabulated in Table S1.
We also examined the lifespan of these animals to see if
skpo-1 RNAi caused a general tness defect. However, on
E. coli OP50, lifespan of the skpo-1 RNAi animals was not
signicantly different when compared to VC RNAi animals
(P= 0.3772) (Figure 1B and Table S2).
To further examine the phenotype resulting from the loss
of skpo-1, we obtained the partial deletion strain, RB1437
from the CGC. The deletion is between bases 827 and 3202
of the 5008 full-length transcript, which ablates a signicant
portion of the protein, including the critical amino acid res-
idues necessary for peroxidase activity in classical animal
heme peroxidases (reviewed by Ortiz de Montellano
2008). Strain RB1437 was backcrossed ve times into our
wild-type N2 strain and three lines were generated: GF89,
GF90, and GF91. As shown in Figure S1, these animals were
very susceptible to E. faecalis compared to wild-type N2
animals. Because signicant differences in susceptibility
were not observed between the strains, we continued our
studies using GF89, which is referred to as skpo-1.
We observed a signicant baggingphenotypehatching
of embryos inside the hermaphrodite that had failed to be
expelledwhen the skpo-1 mutant animals were exposed to
E. faecalis, unlike the skpo-1 RNAi animals. There was concern
that this was articially enhancing their sensitivity to E. faecalis,
thus cdc-25.1 RNAi was employed to induce sterility, as done in
previous C. elegans pathogenesis studies (Shapira et al. 2006;
Irazoqui et al. 2008; van der Hoeven et al. 2011) (Figure 1, CE).
skpo-1 mutant animals grown on cdc-25.1 dsRNA-expressing
E. coli HT115 did not display this maternal bagging phenotype
when they were subsequently exposed to E. faecalis.cdc-25.1
RNAi-exposed skpo-1 mutant worms retained a more pro-
nounced susceptibility phenotype to E. faecalis relative to simi-
larly exposed wild-type animals (P,0.0001) (Figure 1C and
Table S1), suggesting that the pathogen sensitivity is not com-
pletely explained by an increase in internal hatching of progeny.
Regardless of whether skpo-1 mutant worms were sterile,
i.e.,cdc-25.1-treated (Figure 1D and Table S2), or fecund
(Figure S2 and Table S2), they displayed a slight reduction
in lifespan, relative to wild type, when exposed to live E. coli
OP50 (P,0.0001, for both). Because live OP50 has been
Table 1 Susceptibility to E. faecalis following RNAi of putative peroxidase genes
Gene Susceptibility phenotype P-value Median survival (days)
mlt-7 control Wild type 0.3070 6
6
pxn-1 control Wild type 0.9218 4
4
pxn-2 control Wild type 0.8233 6
6
C16C8.2 control Wild type 0.1067 9
9
C46A5.4 control Wild type 0.7206 10
10
F09F3.5 control Susceptible 0.0058 6
7
F32A5.2a control Wild type 0.2499 5
5
F32A5.2b control Wild type 0.4115 6
6
F49E12.1 (skpo-1) control Susceptible 0.0053 5
9
R08F11.7 control Susceptible 0.0004 6
7
K10B4.1 control Wild type 0.7904 8
8
T06D8.10 control Wild type 0.6636 8
8
518 G. R. Tiller and D. A. Garsin
shown to have slightly pathogenic effects (Garigan et al.
2002), we also examined lifespan on heat-killed OP50 and
found that cdc-25.1 RNAi-exposed skpo-1 mutant worms still
displayed a reduction in lifespan (Figure S3 and Table S2).
To determine if the defect in lifespan completely accounted
for the pathogen sensitivity or not, the relative mortality of
skpo-1 mutant worms, compared to wild type, was calcu-
lated (Figure 1F). Relative mortality is the ratio of the lethal
time to kill 50% of the organisms (LT
50
) of pathogen-
infected animals to uninfected animals with the ratio of
wild-type animals normalized to one and has been used in
previous work to analyze susceptibility phenotypes (Tenor
et al. 2004; Chavez et al. 2007). The survival defect for the
skpo-1 mutant animals on E. faecalis was more severe than
on nonpathogenic E. coli compared to wild-type animals
(P= 0.0091) (Figure 1E and Table S3), arguing against
a general tness defect.
To further examine the extent of the susceptibility-to-
pathogen phenotype and determine if it was pathogen
specic, we used P. aeruginosa instead of E. faecalis in the
killing assay (P= 0.3783) (Figure 1F and Table S4). Inter-
estingly, we observed no signicant difference in survival
between cdc-25.1 RNAi skpo-1 mutants and cdc-25.1 RNAi
wild-type animals when exposed to P. aeruginosa. These
results further argue against a general tness defect and
suggest that the function of SKPO-1 may be benecial dur-
ing infection with some, but not all pathogens.
To determine if the susceptibility phenotype on E. faecalis
was a result of changes in pathogen burden, we assessed
how many colony-forming units (CFUs) were in the intes-
tines of the infected worms. Wild-type and skpo-1 mutant
animals were raised on E. coli OP50 until L4 and then ex-
posed to E. faecalis for 12 or 36 hr. The infected worms were
washed to remove surface bacteria, homogenized, and then
Figure 1 SKPO-1 contributes to
C. elegans resistance to E. faeca-
lis. (A) Survival of eri-1 mutant
worms on E. faecalis OG1RF fol-
lowing exposure to vector con-
trol (VC) RNAi and skpo-1 RNAi
(P= 0.0011). (B) Longevity of eri-1
mutant worms on E. coli OP50
following exposure to VC RNAi
and skpo-1 RNAi (P= 0.3772). (C)
Survival of wild-type and skpo-1
mutant worms on E. faecalis fol-
lowing exposure to cdc-25.1
RNAi (P,0.0001). (D) Longevity
of wild-type and skpo-1 mutant
worms on E. coli OP50 following
exposure to cdc-25.1 RNAi (P,
0.0001). (E) The relative mortal-
ity of skpo-1 mutant worms ex-
posed to E. faecalis (EF) is
expressedasaratioof(LT
50
of
wild-type cdc-25.1 RNAi on EF/
LT
50
of skpo-1 mutant cdc-25.1
RNAi on EF) over (LT
50
of wild-
type cdc-25.1 RNAi on E. coli/
LT
50
of skpo-1 mutant cdc-25.1
RNAi on E. coli). The average of
four independent experiments,
with 90 animals each, was used
to calculate the relative mortality. (F)
Survival of wild-type and skpo-1
mutant worms on P. aeruginosa
following exposure to cdc-25.1
RNAi (P= 0.3783). Error bars rep-
resent standard error of the mean
(SEM) and the asterisk indicates
asignicant difference between
wild-type and skpo-1 mutant
worms (P=0.0091). The P-values
were calculated using Students
paired t-test. The median survival
for survival and longevity assays
are listed in Table S1 and Table
S2 along with replicates of the
experiments.
SKPO-1 Protects Against Infection 519
serial dilutions of the homogenates were plated. Interest-
ingly, we observed no signicant difference in CFUs per
worm between the wild-type and skpo-1 mutant animals
at either time point (Figure S4). The result suggests that
the susceptibility phenotype of the skpo-1 mutant cannot
be explained by an increased load of bacteria in the
intestine.
To test for alterations in the immune response between
the skpo-1 and wild-type animals a small panel of genes
known to be upregulated in response to E. faecalis were
examined, clec-35,-42,-60, and -71. These genes encode
C-type lectins (clec), many of which are upregulated in re-
sponse to pathogens (Irazoqui et al. 2010; Engelmann et al.
2011). Sterile (by exposure to cdc-25 RNAi) wild-type and
skpo-1 mutant animals at the L4 stage were exposed to E.
faecalis or E. coli for 18 hr, at which point the animals were
lysed and RNA was extracted. By qRT-PCR, no signicant
difference in the expression of clec-35 and -71 was observed
between the wild type and skpo-1 mutants exposed to E.
faecalis. However, we observed clec-60s expression was sig-
nicantly higher in the skpo-1 mutant and there was a trend
toward increased expression of clec-42, which was not sta-
tistically signicant (Figure S5). These data suggest that the
immune response is altered in the skpo-1 mutant.
Morphological characterization of skpo-1
mutant animals
Animals decient in the peroxidase mlt-7 displayed altered
morphology, including a bli (blistered) phenotype, indicative
of incomplete cross-linking of the cuticle (Thein et al. 2009).
The phenotype strongly suggests that MLT-7s peroxidase
activity contributes to cuticle formation (Thein et al.
2009). Interestingly, despite this rather dramatic cuticle de-
fect, an increase in susceptibility to E. faecalis was not ob-
served (Table 1). To determine if SKPO-1 is also involved in
cuticle biogenesis, we observed the morphology of skpo-1
RNAi and mutant animals. RNAi of skpo-1 did not result in
any visible morphological change in the eri-1 mutant ani-
mals (Figure 2B). However, the skpo-1 mutant did display
some morphological phenotypes. We observed young adult
animals under the dissecting microscope (N= 300) and
found that they ranged from very dumpy (16.5%), to
slightly dumpy (32.9%), to wild type (50.6%) in appearance
(Figure 2, CE). No blistering of the cuticle was ever ob-
served. Because a dumpy phenotype can be associated with
cuticle defects (Page and Johnstone 2007), these results
suggest that SKPO-1 may have some role in cuticle biogene-
sis, but one that is different than MLT-7. Additionally, we
observed no signicant difference in susceptibility between
skpo-1 mutant morphotypes, i.e., all were equally hypersuscep-
tible to E. faecalis relative to wild-type animals (Figure S1).
SKPO-1 features and activity
As mentioned, SKPO-1 contains a ShkT-like domain at its
N terminus from residues 2156 (www.wormbase.org)in
addition to the predicted peroxidase domain (Figure 3A).
It shares this feature with MLT-7 (Thein et al. 2009), and
putative peroxidases C16C8.2 and F32A5.2 (Table 1). For this
reason we propose naming F49E12.1,SKPO-1, and C16C8.2
and F32A5.2,SKPO-2,andSKPO-3 for ShkT-containing
peroxidase. In addition to the ShkT-like domain, the very 59
end contains a predicted signal sequence for secretion, and
cleavage is predicted to occur at Ser19 (www.predisi.de).
To examine SKPO-1s peroxidase domain, we aligned it
with other well-characterized peroxidase domains of inter-
est, those contained in hMPO, BLI-3, and MLT-7. Upon
alignment, we observed SKPO-1 possesses the catalytic triad
(distal histidine, H
222
; arginine, R
332
; and proximal histi-
dine, H
428
) necessary for peroxidase activity (reviewed by
Ortiz de Montellano 2008). However, SKPO-1 lacks the
Figure 2 skpo-1 mutant displays multiple morphotypes.
(A) Wild-type young adult representing the approximate
average size and morphology of a typical C. elegans. (B)
skpo-1 RNAi young adult raised on skpo-1 dsRNA-expressing
E. coli HT115 from L1L4 stages. (CE) Young adult skpo-1
mutants ranged from very dumpy to wild type in size. The
310 microscopy images are representative of .100 wild-
type, skpo-1 RNAi, and skpo-1 worms observed, respectively.
520 G. R. Tiller and D. A. Garsin
residues necessary for covalent heme binding, characteristic
of the animal heme peroxidase family (reviewed by Ortiz de
Montellano 2008). Interestingly, human Duox1/2 (hDuox1/
2) also lack the conserved aspartic and glutamic residues
required for covalent linkage of the heme prosthetic group;
however, both of these recombinant peroxidase domains still
bind heme, albeit weakly (Meitzler and Ortiz de Montellano
2009, 2011). This noncovalent binding may explain MLT-7s
peroxidase activity even though it lacks these covalent
heme-binding residues (Thein et al. 2009).
By comparison, SKPO-1 possesses all of the highlighted
residues in MLT-7 (Figure 3A) and shares signicant identity
with hMPO (34% identity). Thus, we decided to indirectly
assay SKPO-1s potential peroxidase activity using an
Amplex Red assay, modied for whole animals, that detects
H
2
O
2
(Chavez et al. 2007, 2009). Previously, we observed
that exposure to E. faecalis causes a signicant release in
H
2
O
2
dependent on the NADPH oxidase, BLI-3 (Chavez
et al. 2007, 2009). We reasoned that loss of an important
peroxidase during this release might increase the amount of
H
2
O
2
detected. After infecting VC and skpo-1 RNAi eri-1
mutant animals with E. faecalis for 12 hr, released H
2
O
2
was measured (P= 0.0091) (Figure 3B). skpo-1 RNAi eri-1
mutant animals released signicantly greater amounts of
H
2
O
2
, relative to VC RNAi eri-1 mutants, consistent with
the loss of a predicted H
2
O
2
sink. The same result was ob-
served with the skpo-1 mutants compared to wild-type ani-
mals (P,0.0001) (Figure 3C). The difference required E.
faecalis, and the concomitant release of H
2
O
2
, as no signi-
cant differences were observed between wild-type and skpo-
1-decient animals on E. coli (Figure 3, B and C).
To investigate if the increased amount of H
2
O
2
produced
by the skpo-1 mutant was dependent on BLI-3 activity, we
added the NADPH oxidase inhibitor, DPI, to the assay. We
previously demonstrated that BLI-3 is the only expressed
NADPH oxidase encoded by the genome (Chavez et al.
2009), and DPI can be utilized to reduce its activity in
wild-type animals (Chavez et al. 2007). In Figure 3C, DPI
also abrogated the enhanced H
2
O
2
generation observed in
skpo-1 mutant animals. Taken together, Figure 3 supports
the hypothesis that SKPO-1 is a peroxidase that utilizes
H
2
O
2
produced by BLI-3 during infection. Unfortunately,
attempts to purify the protein to demonstrate denitive per-
oxidase activity in vitro were not successful.
SKPO-1 localizes to the hypodermis
C. elegans interfaces with E. faecalis at the cuticle, synthe-
sized by the underlying hypodermis, as it crawls through the
pathogen lawn, and in its intestine, due to ingestion of the
bacteria (Garsin et al. 2001). It has previously been demon-
strated that C. elegans mounts immune responses at these
hostpathogen boundaries, depending on the pathogen and
the nature of the infection (Wong et al. 2007; Pujol et al.
2008; Irazoqui et al. 2010). To address what tissue SKPO-1s
activity is required for normal levels of resistance to E. fae-
calis, a tissue-specic RNAi approach was employed. RNAi-
defective strains of C. elegans were used in which RNAi
activity was genetically restored to specic tissues through
intestinal or hypodermal-specic promoters (vha-6::SID-1
and wrt-2::RDE-1, respectively) (Melo and Ruvkun 2012).
In Figure 4, A and C (and Table S1), we tested the suscep-
tibility of the hypodermal and intestinal-specicRNAi
Figure 3 Evidence that skpo-1 is a potential peroxidase.
(A) Peroxidase domain sequences were aligned against
the putative peroxidase domain of SKPO-1. SKPO-1 pos-
sesses the distal histidine (H
222
), catalytic arginine (R
332
),
and proximal histidine (H
428
), which are necessary for
peroxidase activity. However, SKPO-1 lacks covalent
heme-binding residues (S
221
and L
335
) that are character-
istic of mammalian peroxidases (Ortiz de Montellano
2008). (B) eri-1 mutant worms were grown on VC RNAi
or skpo-1 RNAi prior to exposure with either E. coli or E.
faecalis for 12 hr at 25°. (C) Wild-type and skpo-1 mutant
worms were grown on cdc-25.1 RNAi prior to exposure
with either E. coli or E. faecalis for 12 hr at 25°. (B and C)
Following exposure to E. coli or E. faecalis, the amount of
H
2
O
2
produced per minute was determined using the
Amplex Red assay. Error bars represent the SEM and the
asterisks indicate signicant differences between eri-1
worms exposed to VC RNAi or skpo-1 RNAi that were
infected with E. faecalis as well as between wild-type and
skpo-1 mutant worms exposed to cdc-25.1 RNAi prior to
infection with E. faecalis [P=0.0091 (B) and P,0.0001
(C)]. Additionally, wild-type and skpo-1 mutant worms were
exposedto80mM diphenyleneiodinium chloride (DPI) and
H
2
O
2
levels were calculated for both E. coli-andE. faecalis-
exposed animals (EC, P= 0.0752; EF, P= 0.4161, respec-
tively) P-values were calculated via Studentspairedt-test.
Data in B and C are representative of at least two indepen-
dent replicates.
SKPO-1 Protects Against Infection 521
strains to E. faecalis.AsinFigure1,thesestrainswere
exposed to VC and skpo-1 RNAi prior to infection. In the
hypodermal RNAi strain, we observed an enhanced suscep-
tibility phenotype to E. faecalis in the skpo-1 RNAi animals
relative to VC RNAi (P= 0.0002); however, in the intesti-
nalRNAistrain,nosignicant difference between skpo-1
andVCRNAianimalswasobservedwhenonE. faecalis
(P= 0.9435). On E. coli,nosignicant difference was
observed between VC and skpo-1 RNAi for either the hypo-
dermal or intestinal RNAi line (P= 0.9997 and P=0.6379;
Figure 4, B and D, respectively, and Table S2). From these
experiments we conclude that SKPO-1s functional activity
during pathogen exposure is required in the C. elegans
hypodermis.
To determine in which tissue SKPO-1 is produced, we
used indirect immunouorescence to visualize SKPO-1 local-
ization in young adult animals raised under standard con-
ditions. Using rabbits, a polyclonal primary antibody was
raised against a chemically synthesized SKPO-1 peptide
(see Materials and Methods). A freeze-cracking methodology
was used to disrupt the cuticle of the animal and allow for
internal xation and staining (Seydoux and Dunn 1997).
Following xation, the samples were double stained with
the polyclonal primary antibody to SKPO-1 and an Alexa
Fluor 488 secondary antibody. Localization was clearly ob-
served in the hypodermis of wild-type animals, and an ex-
ample is shown in Figure 5, AC and DF (40 times and 10
times, respectively). Note the strong uorescent staining just
under the outermost layer of the animal. No internal organs
showed evidence of staining, including the gonad and the
intestine, which in this animal had become partially sepa-
rated from the rest of the body. In contrast to wild type,
skpo-1 mutant animals showed no evidence of staining in
any organ (Figure 5, GI). We also examined wild-type and
skpo-1 mutant animals that had been exposed to E. faecalis
for 24 hr prior to staining and did not observe any differ-
ences in the localization of SKPO-1 (data not shown).
By using a secondary antibody conjugated to 10 nm gold
particles for immunogold labeling, we examined localization
at higher resolution by transmission electron microscopy
(TEM). As seen in Figure 5J, the black dots, indicating the
gold particles, were located just under the cuticle layer in
association with the hypodermal cells of wild-type animals.
Hardly any particles were observed in the skpo-1 mutant
animals (Figure 5K). The few observed were randomly scat-
tered. In total, these results demonstrate SKPO-1 is both
physically present and functionally active in the C. elegans
hypodermis.
Discussion
In this work, we demonstrated that a previously unstudied
C. elegans protein with a peroxidase domain, F49E12.1,
plays a protective role during infection with E. faecalis.We
named this protein SKPO-1 for ShkT-containing peroxidase,
because it contains an N-terminal ShkT-like domain. The
ShkT domain family was originally dened as a potassium
channel blocker in the sea anemone (Metridium senile).
Figure 4 SKPO-1 is necessary in
the hypodermis for resistance to
E. faecalis infection. (A) Survival
of the hypodermal RNAi strain
on E. faecalis following exposure
to VC RNAi or skpo-1 RNAi (P=
0.0002). (B) Longevity assay on
E. coli OP50 of the hypodermal
RNAi strain following exposure
to VC RNAi or skpo-1 RNAi (P=
0.9997). (C) Survival of the intes-
tinal RNAi strain on E. faecalis fol-
lowing exposure to VC RNAi or
skpo-1 RNAi (P= 0.9435). (D)
Longevity assay of the intestinal
RNAi strain on E. coli OP50 fol-
lowing exposure to VC RNAi or
skpo-1 RNAi (P= 0.6379).
522 G. R. Tiller and D. A. Garsin
Binding to the potassium channel requires two conserved
residues that are not found in this particular ShkT-like do-
main of SKPO-1 or others from C. elegans (data not shown).
It is postulated that the more general function of ShkT-like
domains are as contact surfaces for protein interactions
(Tsang et al. 2007). For this reason, SKPO-1 and other ShkT-
containing peroxidases may be most closely related to the
peroxidasins, subfamily 2 of the peroxidase-cyclooxygenase
superfamily, and in fact, SKPO-1 was placed in this family
by phylogenetic analysis (Soudi et al. 2012). These peroxi-
dase domain-containing proteins also have protein interaction
domains, but they are typically type C-like immunoglobulin
domains, leucine-rich repeats, orvonWillebrandfactorCmod-
ules. This is in contrast to the mammalian peroxidases, MPO,
LPO, and eosinophil peroxidase that lack these extra domains
and belong to subfamily 1 (Zamocky et al. 2008). Additionally,
it is thought that subfamily 1 evolved from subfamily 2
(Zamocky et al. 2008). Other C. elegans peroxidase do-
main-containing proteins that have a ShkT-like domain in-
clude MLT-7 (Thein et al. 2009), C16C8.2,andF32A5.2,
but no study has yet addressed this domains function in
the context of a peroxidase.
An unexpected nding was the discovery that SKPO-1 is
located in the hypodermis and is functionally protective in
this tissue against E. faecalis infection. Infection of C. elegans
with E. faecalis results in colonization of the gut, leading to
distension of the intestinal lumen and clear signs of physical
damage, such as effacement of the microvilli (Garsin et al.
2001; Cruz et al. 2013). Though the worm is exposed to E.
faecalis on its outer surface as it moves through the patho-
gen lawn, there is no notable colonization or characterized
physical damage to the cuticle or hypodermis. In contrast,
Figure 5 SKPO-1 localizes to the
C. elegans hypodermis. (AF and
J) Wild-type and (GI and K)
skpo-1 mutant worms were immu-
nostained with anti-SKPO-1 poly-
clonal antibodies and imaged
using uorescence (AI) or trans-
mission electron microscopy (J and
K), respectively. (AC) The 340
magnication of a wild-type worm
shows hypodermal SKPO-1 lo-
calization. (DF) The 310 mag-
nication of a wild-type worm.
(GI) The 310 magnication of
askpo-1 worm shows loss of
SKPO-1 staining. (J) In wild-type
worms, the black dots, indica-
tive of immunogold labeling,
are localized beneath the cuticle
layer, but external to the outer
hypodermal cell surface. (K) In
skpo-1 mutant worms, very few
black dots are observed. White
arrowhead, apical hypodermal
surface; black arrowhead, 10 nm
gold-labeled secondary to SKPO-1).
Microscopy images are representa-
tive of .100 (uorescent) or .10
(TEM) wild-type and skpo-1 mu-
tant worms observed.
SKPO-1 Protects Against Infection 523
bacterial pathogens such as Microbacterium nematophilum,
Xenorhabdus nematophila, and Yersinia pestis adhere to and
colonize the cuticle surface (Hodgkin et al. 2000; Couillault
and Ewbank 2002; Darby et al. 2002). The natural fungal
pathogen Drechmeria coniospora initially adheres to the cu-
ticle and then penetrates the hypodermis, whereas the hu-
man fungal pathogen Candida albicans rst colonizes the
intestine and eventually penetrates the cuticle from the in-
side out (Jansson et al. 1985; Breger et al. 2007). We pre-
viously showed that tissue-specic loss of bli-3 in the
hypodermis also increased susceptibility of C. elegans to E.
faecalis (Chavez et al. 2009). Based on the protective effects
of SKPO-1 and BLI-3 in this tissue, we postulate that a hypo-
dermal immune response does play some role in protecting
C. elegans during infection with E. faecalis. The question is by
what mechanism?
Several models for how these proteins might exert their
protective effects can be imagined. It could be that loss of
skpo-1 results in a weaker cuticle barrier that increases sus-
ceptibility. Alternatively, SKPO-1 could use H
2
O
2
produced
by BLI-3 to form more potent antioxidants, analogous to the
human Duox/LPO system on mucosal surfaces (Conner et al.
2002; Geiszt et al. 2003; Forteza et al. 2005). Or the H
2
O
2
produced by BLI-3 could act as a signaling molecule in either
an autonomous or noncell-autonomous fashion. Unlike
many signaling molecules, H
2
O
2
is highly diffusible, and it
could directly move into other tissues, such as the intestine,
to signal responses. Interestingly, in zebrash, H
2
O
2
pro-
duced by a Duox in response to wounding acts as an attrac-
tant in a diffusion gradient to draw leukocytes to the area of
damage (Niethammer et al. 2009). Clearly, the amount of
H
2
O
2
is important. We previously showed that loss of BLI-3
and the resulting decrease in H
2
O
2
production caused an
increase in susceptibility to infection (Chavez et al. 2009),
whereas in this work we show that loss of a peroxidase and
a concurrent increase in H
2
O
2
also increases susceptibility. A
similar situation is apparent from studies of infection using
the model host Drosophila melanogaster (Ha et al. 2005a,b).
Loss of an intestinal DUOX enzyme or an intestinal catalase
both increase susceptibility to infection, but with opposite
effects on ROS levels in this tissue. It is possible that SKPO-1
is catalyzing the degradation of excess H
2
O
2
to prevent host
damage. We formerly demonstrated, by a variety of means,
that infection causes oxidative stress in C. elegans (Chavez
et al. 2007; Mohri-Shiomi and Garsin 2008; van der Hoeven
et al. 2011), much of it dependent on BLI-3 activity (van der
Hoeven et al. 2011).
Interestingly, there is evidence for a hypodermal response
to several pathogens that are thought to mainly cause
infection in the intestine. Microarray studies that examined
the transcriptional responses of C. elegans to E. faecalis,Ser-
ratia marcescens,Erwinia carotovora,Photorhabdus lumines-
cens,S. aureus, and P. aeruginosa all noted a dramatic
downregulation in the expression of genes related to cuticle
biosynthesis, such as those encoding collagens (Wong et al.
2007; Irazoqui et al. 2010). The response is not thought to
be due to a general reduction of gene transcription in this
tissue and may be indicative of several possibilities (Wong
et al. 2007). The changes in transcription of the genes could
be part of a protective response that is occurring in the
hypodermis to protect against pathogens. Or the changes
could be purposely caused by the pathogens as part of their
virulence programs to damage the host. Alternatively, the
changes in expression of the cuticle biosynthetic genes could
be a neutral side effect resulting from alterations in signal-
ing due to pathogen exposure. Another microarray study
observed an increase in the expression of a large number
of genes related to cuticle biosynthesis in an arr-1 mutant
that was found to have altered susceptibility to pathogens
(Singh and Aballay 2012). arr-1 encodes for arrestin-1,
a G-protein coupled receptor (GPCR) adaptor protein that
is necessary for GPCR signaling in several neurons of
C. elegans. Whether these changes in resistance had anything
to do with alterations in the amount of cuticle biosynthetic
proteins being produced was not investigated (Singh and
Aballay 2012). Overall, these studies suggest that pathogen
exposure, even to those pathogens not thought to directly
affect the cuticle, cause major changes in the expression of
the cuticle biosynthetic genes that might be indicative of a re-
sponse to the infection in the hypodermis.
In conclusion, we uncovered putative peroxidases that
affected susceptibility to E. faecalis in C. elegans. We focused
our characterization on F49E12.1, named SKPO-1 because it
contains a ShkT-like domain and a peroxidase domain. In
addition to the pathogen susceptibility phenotype, loss of
skpo-1 resulted in a dumpy phenotype of uneven pene-
trance, suggestive of some role in cuticle biosynthesis (Page
and Johnstone 2007). In support of a functional peroxidase
domain, we noted that SKPO-1 shares the same critical res-
idues as MLT-7 in its active site (Thein et al. 2009), and
animals mutant for skpo-1 produce signicantly more
H
2
O
2
during infection. The functional and physical location
for the protein was shown to be the hypodermis, which was
surprising since E. faecalis has been characterized as infect-
ing the intestine of C. elegans (Garsin et al. 2001; Cruz et al.
2013). This result, along with other evidence from the liter-
ature (Wong et al. 2007; Irazoqui et al. 2010), suggests that
the hypodermis plays an important role during exposure of
C. elegans to many human pathogens that do not obviously
colonize or damage the cuticle, warranting further
investigation.
Acknowledgments
For the TEM imaging, we thank S. Kolodziej and P. Navarro
(Department of Pathology and Laboratory Medicine EM
Laboratory, University of Texas Health Science Center at
Houston). We thank G. Ruvkun for the tissue-specic RNAi
C. elegans strains and K. McCallum, S. Arur, K. Park, T. Furata,
T. Leto, A. Page, M. Cruz, and W. Nauseef for helpful input on
this project. We acknowledge the Caenorhabditis Genetics
Center for providing us with many of the C. elegans strains
524 G. R. Tiller and D. A. Garsin
used in the study, J. Ahringer and Geneservice for the bacte-
rial strains for RNAi, and WormBase for their help and advice
in naming genes. This work was supported by Public Health
Service grants R01AI076406 to D.A.G. and T32AI55449 to
G.R.T. from the National Institute of Allergy and Infectious
Diseases.
Literature Cited
Breger, J., B. B. Fuchs, G. Aperis, T. I. Moy, F. M. Ausubel et al.,
2007 Ant ifun gal ch emical compounds identied using a
C. elegans pathogenicity assay. PLoS Pathog. 3: e18.
Brenner, S., 1974 The genetics of Caenorhabditis elegans. Genet-
ics 77: 7194.
Chavez, V., A. Mohri-Shiomi, A. Maadani, L. A. Vega, and D. A.
Garsin, 2007 Oxidative stress enzymes are required for DAF-
16-mediated immunity due to generation of reactive oxygen
species by Caenorhabditis elegans. Genetics 176: 15671577.
Chavez, V., A. Mohri-Shiomi, and D. A. Garsin, 2009 Ce-Duox1/
BLI-3 generates reactive oxygen species as a protective innate
immune mechanism in Caenorhabditis elegans. Infect. Immun.
77: 49834989.
Conner, G. E., M. Salathe, and R. Forteza, 2002 Lactoperoxidase
and hydrogen peroxide metabolism in the airway. Am. J. Respir.
Crit. Care Med. 166: S57S61.
Couillault, C., and J. J. Ewbank, 2002 Diverse bacteria are patho-
gens of Caenorhabditis elegans. Infect. Immun. 70: 47054707.
Cruz, M. R., C. E. Graham, B. C. Gagliano, M. C. Lorenz, and D. A.
Garsin, 2013 Enterococcus faecalis inhibits hyphal morpho-
genesis and virulence of Candida albicans. Infect. Immun. 81:
189200.
Darby, C., J. W. Hsu, N. Ghori, and S. Falkow, 2002 Caenorhabditis
elegans: plague bacteria biolm blocks food intake. Nature 417:
243244.
Dunny, G. M., B. L. Brown, and D. B. Clewell, 1978 Induced cell
aggregation and mating in Streptococcus faecalis: evidence for
a bacterial sex pheromone. Proc. Natl. Acad. Sci. USA 75: 3479
3483.
Edens, W. A., L. Sharling, G. Cheng, R. Shapira, J. M. Kinkade et al.,
2001 Tyrosine cross-linking of extracellular matrix is catalyzed
by Duox, a multidomain oxidase/peroxidase with homology to
the phagocyte oxidase subunit gp91phox. J. Cell Biol. 154: 879
891.
Engelmann, I., A. Griffon, L. Tichit, F. Montanana-Sanchis, G. Wang
et al., 2011 A comprehensive analysis of gene expression
changes provoked by bacterial and fungal infection in C. ele-
gans. PLoS ONE 6: e19055.
Forteza, R., M. Salathe, F. Miot, R. Forteza, and G. E. Conner,
2005 Regulated hydrogen peroxide production by Duox in human
airway epithelial cells. Am. J. Respir. Cell Mol. Biol. 32: 462469.
Fraser, A. G., R. S. Kamath, P. Zipperlen, M. Martinez-Campos, M.
Sohrmann et al., 2000 Functional genomic analysis of C. ele-
gans chromosome I by systematic RNA interference. Nature 408:
325330.
Garigan, D., A. L. Hsu, A. G. Fraser, R. S. Kamath, J. Ahringer et al.,
2002 Genetic analysis of tissue aging in Caenorhabditis ele-
gans: a role for heat-shock factor and bacterial proliferation.
Genetics 161: 11011112.
Garsin, D. A., C. D. Sifri, E. Mylonakis, X. Qin, K. V. Singh et al.,
2001 A simple model host for identifying Gram-positive viru-
lence factors. Proc. Natl. Acad. Sci. USA 98: 1089210897.
Garsin, D. A., J. M. Villanueva, J. Begun, D. H. Kim, C. D. Sifri et al.,
2003 Long-lived C. elegans daf-2 mutants are resistant to bac-
terial pathogens. Science 300: 1921.
Geiszt, M., K. Lekstrom, S. Brenner, S. M. Hewitt, R. Dana et al.,
2003 NAD(P)H oxidase 1, a product of differentiated colon
epithelial cells, can partially replace glycoprotein 91phox in
the regulated production of superoxide by phagocytes. J. Immu-
nol. 171: 299306.
Ha, E. M., C. T. Oh, Y. S. Bae, and W. J. Lee, 2005a A direct role
for dual oxidase in Drosophila gut immunity. Science 310: 847
850.
Ha, E. M., C. T. Oh, J. H. Ryu, Y. S. Bae, S. W. Kang et al.,
2005b An antioxidant system required for host protection
against gut infection in Drosophila. Dev. Cell 8: 125132.
Hodgkin, J., P. E. Kuwabara, and B. Corneliussen, 2000 A novel
bacterial pathogen, Microbacterium nematophilum, induces
morphological change in the nematode C. elegans. Curr. Biol.
10: 16151618.
Hope, I. A., 1999 C. elegans: A Practical Approach, Oxford Univer-
sity Press, Oxford.
Irazoqui, J. E., A. Ng, R. J. Xavier, and F. M. Ausubel, 2008 Role
for beta-catenin and HOX transcription factors in Caenorhabdi-
tis elegans and mammalian host epithelial-pathogen interac-
tions. Proc. Natl. Acad. Sci. USA 105: 1746917474.
Irazoqui, J. E., E. R. Troemel, R. L. Feinbaum, L. G. Luhachack,
B. O. Cezairliyan et al., 2010 Distinct pathogenesis and host re-
sponses during infection of C. elegans by P. aeruginosa and S.
aureus. PLoS Pathog. 6: e1000982.
Jansson, H. B., A. Jeyaprakash, and B. M. Zuckerman,
1985 Differential adhesion and infection of nematodes by
the endoparasitic fungus Meria coniospora (Deuteromycetes).
Appl. Environ. Microbiol. 49: 552555.
Kamath, R. S., A. G. Fraser, Y. Dong, G. Poulin, R. Durbin et al.,
2003 Systematic functional analysis of the Caenorhabditis ele-
gans genome using RNAi. Nature 421: 231237.
Kennedy, S., D. Wang, and G. Ruvkun, 2004 A conserved siRNA-
degrading RNase negatively regulates RNA interference in C.
elegans. Nature 427: 645649.
Kim, D. H., R. Feinbaum, G. Alloing, F. E. Emerson, D. A. Garsin
et al., 2002 A conserved p38 MAP kinase pathway in Caeno-
rhabditis elegans innate immunity. Science 297: 623626.
Klebanoff, S. J., 2005 Myeloperoxidase: friend and foe. J. Leukoc.
Biol. 77: 598625.
Meitzler, J. L., and P. R. Ortiz de Montellano, 2009 Caenorhabditis
elegans and human dual oxidase 1 (DUOX1) peroxidasedo-
mains: insights into heme binding and catalytic activity. J. Biol.
Chem. 284: 1863418643.
Meitzler, J. L., and P. R. Ortiz de Montellano, 2011 Structural
stability and heme binding potential of the truncated human
dual oxidase 2 (DUOX2) peroxidase domain. Arch. Biochem.
Biophys. 512: 197203.
Meitzler, J. L., R. Brandman, and P. R. Ortiz de Montellano,
2010 Perturbed heme binding is responsible for the blistering
phenotype associated with mutations in the Caenorhabditis el-
egans dual oxidase 1 (DUOX1) peroxidase domain. J. Biol.
Chem. 285: 4099141000.
Meitzler, J. L., S. Hinde, B. Ban, W. M. Nauseef, and P. R. Ortiz de
Montellano, 2013 Conserved cysteine residues provide a pro-
tein-protein interaction surface in dual oxidase (DUOX) pro-
teins. J. Biol. Chem. 288: 71477157.
Melo, J. A., and G. Ruvkun, 2012 Inactivation of conserved C.
elegans genes engages pathogen- and xenobiotic-associated de-
fenses. Cell 149: 452466.
Mohri-Shiomi, A., and D. A. Garsin, 2008 Insulin signaling and
the heat shock response modulate protein homeostasis in the
Caenorhabditis elegans intestine during infection. J. Biol. Chem.
283: 194201.
Moskwa, P., D. Lorentzen, K. J. Excoffon, J. Zabner, P. B. McCray, Jr.
et al., 2007 A novel host defense system of airways is defective
in cystic brosis. Am. J. Respir. Crit. Care Med. 175: 174183.
SKPO-1 Protects Against Infection 525
Niethammer, P., C. Grabher, A. T. Look, and T. J. Mitchison,
2009 A tissue-scale gradient of hydrogen peroxide mediates
rapid wound detection in zebrash. Nature 459: 996999.
Ortiz de Montellano, P. R., 2008 Mechanism and role of covalent
heme binding in the CYP4 family of P450 enzymes and the
mammalian peroxidases. Drug Metab. Rev. 40: 405426.
Page, A. P., and I. L. Johnstone, 2007 The cuticle (March 19,
2007), WormBook, ed. The C. elegans Research Community,
WormBook, doi/10.1895/wormbook.1.138.1, http://www.
wormbook.org.
Pujol, N., O. Zugasti, D. Wong, C. Couillault, C. L. Kurz et al.,
2008 Anti-fungal innate immunity in C. elegans is enhanced
by evolutionary diversication of antimicrobial peptides. PLoS
Pathog. 4: e1000105.
Rada,B.,K.Lekstrom,S.Damian,C.Dupuy,andT.L.Leto,
2008 The Pseudomonas toxin pyocyanin inhibits the dual
oxidase-based antimicrobial system as it imposes oxidative
stress on airway epithelial cells. J. Immunol. 181: 4883
4893.
Rahme, L. G., E. J. Stevens, S. F. Wolfort, J. Shao, R. G. Tompkins
et al., 1995 Common virulence factors for bacterial pathoge-
nicity in plants and animals. [see comments] Science 268:
18991902.
Seydoux, G., and M. A. Dunn, 1997 Transcriptionally repressed
germ cells lack a subpopulation of phosphorylated RNA poly-
merase II in early embryos of Caenorhabditis elegans and Dro-
sophila melanogaster. Development 124: 21912201.
Shapira, M., B. J. Hamlin, J. Rong, K. Chen, M. Ronen et al.,
2006 A conserved role for a GATA transcription factor in reg-
ulating epithelial innate immune responses. Proc. Natl. Acad.
Sci. USA 103: 1408614091.
Simmer, F., C. Moorman, A. M. van der Linden, E. Kuijk, P. V. van
den Berghe et al., 2003 Genome-wide RNAi of C. elegans us-
ing the hypersensitive rrf-3 strain reveals novel gene functions.
PLoS Biol. 1: E12.
Singh, V., and A. Aballay, 2012 Endoplasmic reticulum stress
pathway required for immune homeostasis is neurally con-
trolled by arrestin-1. J. Biol. Chem. 287: 3319133197.
Soudi, M., M. Zamocky, C. Jakopitsch, P. G. Furtmuller, and C.
Obinger, 2012 Molecular evolution, structure, and function
of peroxidasins. Chem. Biodivers. 9: 17761793.
Sumimoto, H., 2008 Structure, regulation and evolution of Nox-
family NADPH oxidases that produce reactive oxygen species.
FEBS J. 275: 32493277.
Tenor, J. L., B. A. McCormick, F. M. Ausubel, and A. Aballay,
2004 Caenorhabditis elegans-based screen identies Salmo-
nella virulence factors required for conserved host-pathogen in-
teractions. Curr. Biol. 14: 10181024.
Thein, M. C., A. D. Winter, G. Stepek, G. McCormack, G. Stapleton
et al., 2009 Combined extracellular matrix cross-linking activ-
ity of the peroxidase MLT-7 and the dual oxidase BLI-3 is critical
for post-embryonic viability in Caenorhabditis elegans. J. Biol.
Chem. 284: 1754917563.
Tsang, S. W., C. Q. Nguyen, D. H. Hall, and K. L. Chow, 2007 mab-7
encodes a novel transmembrane protein that orchestrates sensory
ray morphogenesis in C. elegans. Dev. Biol. 312: 353366.
van der Hoeven, R., K. C. McCallum, M. R. Cruz, and D. A. Garsin,
2011 Ce-Duox1/BLI-3 generated reactive oxygen species trig-
ger protective SKN-1 activity via p38 MAPK signaling during
infection in C. elegans. PLoS Pathog. 7: e1002453.
Wong,D.,D.Bazopoulou,N.Pujol,N.Tavernarakis,andJ.J.Ewbank,
2007 Genome-wide investigation reveals pathogen-specicand
shared signatures in the response of Caenorhabditis elegans to
infection. Genome Biol. 8: R194.
Zamocky, M., C. Jakopitsch, P. G. Furtmuller, C. Dunand, and C.
Obinger, 2008 The peroxidase-cyclooxygenase superfamily:
reconstructed evolution of critical enzymes of the innate im-
mune system. Proteins 72: 589605.
Communicating editor: B. Lazzaro
526 G. R. Tiller and D. A. Garsin
GENETICS
Supporting Information
http://www.genetics.org/lookup/suppl/doi:10.1534/genetics.113.160606/-/DC1
The SKPO-1 Peroxidase Functions in the Hypodermis
to Protect Caenorhabditis elegans From
Bacterial Infection
George R. Tiller and Danielle A. Garsin
Copyright © 2014 by the Genetics Society of America
DOI: 10.1534/genetics.113.160606
G.R.TillerandD.A.Garsin
 
2SI
FileS1
SUPPLEMENTARYMATERIALSANDMETHODS
RNAisolationandqRTPCR
PriortoRNAisolation,wildtypeandskpo1mutantanimalsweregrownoncdc25.1dsRNAexpressingE.coliHT115
fromtheeggtoL4larvalstage.L4animalswerethenexposedtoeithercdc25.1RNAiorE.faecalisOG1RFfor18hours.The
RNAwasisolatedusingTrizol(Invitrogen)accordingtothemanufacturer.RNAsamplesweretreatedwithDNaseItoeliminate
contaminatingDNAbytheTurboDNAfreekit(AppliedBiosystem)accordingtothemanufacturer.qRTPCRwasperformedas
described(VANDERHOEVENetal.2011).PrimersusedarelistedinTableS6(act1servedasthereferencegene).
BacterialColonization
TheCFUanalysiswasconductedinamannersimilartopastwork(GARSINetal.2001).Briefly,L4wildtypeandskpo1
mutantanimalsgrownonE.coliOP50wereexposedon100mmplatescontainingBHIagarwithgentamycin(10g/mL)seeded
with100lofE.faecalisOG1RFforeither12or36hoursat25˚C.Wormswerewashed3timeswithM9bufferat1.4rpm.
Wormswerethenwashedtwicewith25mMtetramisolehydrochloridetopreventingestionoftheantibiotictreatment.
Wormswereincubatedatroomtemperaturefor60minutesin25mMtetramisolehydrochloridesupplementedwithampicillin
andkanamycin,bothat1mg/mL,tokillsurfaceattachedE.faecalis.Wormswerecollectedat1.4rpmandwashedtwicewith
25mMtetramisolehydrochloridepriortogrinding.Tenwormsin10lweretransferredto200lofM9andgroundfor1
minuteusingamotorizedpestle(Kontescordlesscat#K7495400000andpestlescat#K7495211590).Serialdilutionswere
performedand100lofeachdilutionplatedonto100mmBHIgentamycin10g/mLplatesfor24hoursat37˚C.

G.R.TillerandD.A.Garsin
 
3SI
FigureS1Survivalofcdc25.1RNAitreatedskpo1animallinesonE.faecalis.Wildtypeandskpo1wormsweregrownoncdc
25.1dsRNAexpressingE.coliHT115fromL1throughL4larvalstagepriortoexposuretoE.faecalisOG1RF.Theskpo1worms
(GF89,GF90andGF91)weremoresusceptibletoE.faecalisrelativetowildtype(Pvalues<0.0001,0.0110,and<0.0001,
respectively).

G.R.TillerandD.A.Garsin
 
4SI
FigureS2Lifespandefectofskpo1mutantsonE.coliOP50withoutpriorcdc25.1exposure.Wildtypeandskpo1worms
weregrownonE.coliOP50fromL1throughL4larvalstagepriortothelongevityassayinwhichtheyweretransferredtoNGM
plateswithFUDR(100µg/ml)seededwithconcentrated(20X)E.coliOP50.Theaveragelifespanoftheskpo1wormswas
significantlyshorterthantheirwildtype,counterparts(Pvalue<0.0001).
G.R.TillerandD.A.Garsin
 
5SI
FigureS3skpo1mutantsdisplayalifespandefectonheatkilledE.coliOP50.Wildtypeandskpo1wormsweregrownoncdc
25.1dsRNAexpressingE.coliHT115fromL1throughL4larvalstagepriortothelongevityassay.Thewormsweretransferredto
NGMplateslikeinFigureS2;however,theE.coliOP50hadbeenheatkilled.Weobservedthattheskpo1wormsdisplayeda
longevitydefectonheatkilledOP50,relativetowildtypeworms(Pvalue<0.0001).
G.R.TillerandD.A.Garsin
 
6SI
FigureS4Theskpo1mutantdoesnothaveincreasedintestinalbacterialloadduringE.faecalisinfection.L4wildtypeand
skpo1mutantanimalswereexposedtoE.faecalisat25˚Cfor12or36hours,respectively.CFUvaluesforbothwildtypeand
skpo1animalsindicatedthatthereisnosignificantdifferenceinintestinalcolonizationbyE.faecalis.Threebiologicalreplicates
foreachstrainwereusedpertimepointandtheexperimentrepeatedtwice—i.eatotaln=60perstrain.Pvalueswere0.3913
and0.3592for12and36hours,respectively.
G.R.TillerandD.A.Garsin
 
7SI
FigureS5clec60expressioniselevatedinskpo1mutantanimals,relativetowildtype,inresponsetoE.faecalisinfection.L4
wildtypeandskpo1mutantanimalswereexposedtoE.faecalisfor18hoursat25˚C.Threetechnicalreplicatesperstrainper
genewereassayedforrelativeexpressionofinnateimmuneresponsegenesandtheexperimentwasrepeated,independently,
twice.ThesegeneswerepreviouslyfoundtodisplayincreasedexpressioninwildtypeanimalsinresponsetoE.faecalis
(ENGELMANNetal.2011).Ofthegenessurveyed,clec60wastheonlygenetodisplayasignificantincreaseinexpressionduring
infection,relativetoact1.Pvalueswere0.1912,0.1768,0.04432,and0.4684forclec35,42,60and71,respectively.

G.R.TillerandD.A.Garsin
 
8SI
TableS1MediansurvivalandPvaluesofE.faecalisOG1RFkillingassays
FigurenumberExperimentnumberStrain,exposureconditionsduring
development
Median
survival(days)
P
value
1A1wildtype,VCRNAi
wildtype,skpo1RNAi
8
7
C
=0.0404
2wildtype,VCRNAi
wildtype,skpo1RNAi
7
6
C
=0.0331
3er
i
1,VCRNAi
eri1,skpo1RNAi
8
7
C
=0.001
4er
i
1,VCRNAi
eri1,skpo1RNAi
7
5
C
=0.0053
5er
i
1,VCRNAi
eri1,skpo1RNAi
9
8
C
=0.0037
1C1wildtype,cdc25.1 RNAi
skpo1,cdc25.1RNAi
10
6
C
<0.0001
2wildtype,cdc25.1 RNAi
skpo1,cdc25.1RNAi
10
8
C
<0.0001
3wild type,cdc25.1 RNAi
skpo1,cdc25.1RNAi
13
8
C
=0.0016
4wildtype,cdc25.1 RNAi
skpo1,cdc25.1RNAi
13
8
C
=0.0005
5wildtype,cdc25.1 RNAi
skpo1,cdc25.1RNAi
13
7
C
<0.0001
4A1wrt2,VCRNAi
wrt2,skpo1RNAi
5
4
C
=0.0002
2wrt2,VCRNAi
wrt2,skpo1RNAi
4
3
C
=0.0176
3wrt2,VCRNAi
wrt2,skpo1RNAi
5
4
C
=0.0376
4wrt2,VCRNAi
wrt2,skpo1RNAi
4
4
C
=0.0058
4C1vha6,VCRNAi
vha6,skpo1RNAi
11
11
C
=0.6379
2vha6,VCRNAi
vha6,skpo1RNAi
11
11
C
=0.735
S11wildtype,OP50
skpo1*,OP50
8
3
C
<0.0001
2wildtype,OP50
skpo1*,OP50
8
3
C
<0.0001
C=control,*DataonlyshownforGF89skpo1strain.
G.R.TillerandD.A.Garsin
 
9SI
TableS2MediansurvivalandPvaluesofE.coliOP50longevityassays
FigurenumberExperiment
number
Strain,exposureconditionsduring
development
Mediansurvival
(Days)
P
value
1B1er
i
1,VCRNAi
eri1,skpo1RNAi
10
10
C
=0.3772
2er
i
1,VCRNAi
eri1,skpo1RNAi
10
10
C
=0.8948
1D1wildtype,cdc25.1 RNAi
skpo1,cdc25.1RNAi
10
8
C
<0.0001
2wildtype,cdc25.1 RNAi
skpo1,cdc25.1RNAi
14
11
C
<0.0001
3wildtype,cdc25.1 RNAi
skpo1,cdc25.1RNAi
14
11
C
<0.0001
4B1vha6,VCRNAi
vha6,skpo1RNAi
10
10
C
=0.9997
2vha6,VCRNAi
vha6,skpo1RNAi
11
10
C
=0.4484
4D1wrt2,VCRNAi
wrt2,skpo1RNAi
11
11
C
=0.6379
2wrt2,VCRNAi
wrt2,skpo1RNAi
11
11
C
=0.7235
S21wildtype,OP50
skpo1,OP50
14
11
C
<0.0001
2wildtype,OP50
skpo1,OP50
14
11
C
<0.0001
S3*1N2,cdc25.1 RNAi*
skpo1,cdc25.1RNAi*
16
13
C
<0.0001
2N2,cdc25.1 RNAi*
skpo1,cdc25.1RNAi*
14
12
C
<0.0001
C=control,*AssayedonheatkilledOP50
G.R.TillerandD.A.Garsin
 
10SI
TableS3DataforRelativeMortalityCalculation
FigurenumberExperiment
number
Strain Relativemortality
P
value
1FAverageof
experiments14
N2
skpo1
1
1
1
1
Avg=1
1.312
1.452
1.654
1.253
Avg=1.418
C
=0.0091
C=control

G.R.TillerandD.A.Garsin
 
11SI
TableS4MediansurvivalandPvaluesofP.aeruginosaPA14killingassays
FigureNumberExperiment
number
Strain,exposureconditions
duringdevelopment
Mediansurvival
(Hours)
P
value
1E1N2,cdc25.1 RNAi
skpo1,cdc25.1RNAi
91
88
C
=0.3783
2N2,cdc25.1 RNAi
skpo1,cdc25.1RNAi
89
69
C
=0.0639
3N2,cdc25.1 RNAi
skpo1,cdc25.1RNAi
72
72
C
=0.2613
G.R.TillerandD.A.Garsin
 
12SI
TableS5PrimersforcDNAinsertsligatedintopL4440constructfordsRNAproductioninE.coli
OligonameSequence
F32A52.A5F5’TTGCGGCCGCAATTCACCAGGGAATTTACACC3’
F32A52.A3R5’CCTCGAGGGTGGTCTTTTCACATTCTGG3’
F32A52.B5F5’TTGCGGCCGCAACAACCACTCATTTCACCAGG3’
F32A52.B3R5’CCTCGAGGCTGTAGTTGTTACTCTTTGTGG3’
p
xn15F5’TTGCGGCCGCAAGGACTCTGGAAGGTACAC3’
p
xn13R5’CCTCGAGGATACTTGGCACTTCCACTCT3’
G.R.TillerandD.A.Garsin
 
13SI
TableS6qRTPCRprimersforassessingexpressionofinnateimmuneresponsegenesinresponsetoE.faecalis
OligonameSequence
clec355F5’AGATGCTGGACAGTGGAAAAG3’
clec353R5’GTGCGGAGTATTGTAGCGTAG3’
clec425F5’GTAACTCCGTATTGGCTGGG3’
clec423R5’GTAAACGCAGCTTCCAATCTC3’
clec605F5’TGTAAGAGAACAGTTGGAACCC3’
clec603R5’TATGTGCATGGGTACTGATCG3’
clec715F5’ACGACAGGAAGTGATGTATTGG3’
clec713R5’TTGACGGACTTTAGCCACTG3’
act15F5’ACCATGTACCCAGGAATTGC3’
act13R5’TGGAAGGTGGAGAGGGAAG3’
G.R.TillerandD.A.Garsin
 
14SI
SUPPLEMENTARYLITERATURECITED
Engelmann,I.,A.Griffon,L.Tichit,F.MontananaSanchis,G.Wangetal.,2011Acomprehensiveanalysisofgeneexpression
changesprovokedbybacterialandfungalinfectioninC.elegans.PLoSOne6:e19055.
Garsin,D.A.,C.D.Sifri,E.Mylonakis,X.Qin,K.V.Singhetal.,2001AsimplemodelhostforidentifyingGrampositivevirulence
factors.ProceedingsoftheNationalAcademyofSciencesoftheUnitedStatesofAmerica98:1089210897.
vanderHoeven,R.,K.C.McCallum,M.R.CruzandD.A.Garsin,2011CeDuox1/BLI3GeneratedReactiveOxygenSpecies
TriggerProtectiveSKN1Activityviap38MAPKSignalingduringInfectioninC.elegans.PLoSPathog7:e1002453.
... Portions of the ROS production pathways are largely conserved in C. elegans and vertebrate animals, including humans. For example itr-1, the inositol trisphosphate receptor is required for intracellular calcium release in response to infection [1], an aquaporin, aqp-1, is required for the full anti-pathogen response [11], and a dual oxidase/peroxidase, bli-3 and a secreted peroxidase, skpo-1, are important for the defense against pathogens [12,13]. Importantly, these pathways are simplified in C. elegans with minimal redundancy. ...
... To test this notion, we fed wild type and pbo-1 mutants the opportunistic pathogen Enterococcus faecalis (Fig 2). Similar to previous studies [13,22], we found that 50% of wild-type animals fed E. faecalis on nematode growth media (NGM) were killed with a median survival (LT 50 ) of 9.5 ± 1 days, post infection (Fig 2A). By contrast, 50% of pbo-1 mutants were killed after an average of 6 ± 1 days of E. faecalis feeding, which was significantly shorter than the wild-type animals (p<0.005) ...
... Work from the Ausubel and Garsin labs have shaped our understanding of the molecular events of pathogenesis, and specifically how E. faecalis and C. elegans interact during infection [7,12,13,22]. Briefly, recent work has identified that infection of C. elegans results in the production of H 2 O 2 as part of the innate immune system [12]. ...
Article
Full-text available
Caenorhabditis elegans are soil-dwelling nematodes and models for understanding innate immunity and infection. Previously, we developed a novel fluorescent dye (KR35) that accumulates in the intestine of C. elegans and reports a dynamic wave in intestinal pH associated with the defecation motor program. Here, we use KR35 to show that mutations in the Ca²⁺-binding protein, PBO-1, abrogate the pH wave, causing the anterior intestine to be constantly acidic. Surprisingly, pbo-1 mutants were also more susceptible to infection by several bacterial pathogens. We could suppress pathogen susceptibility in pbo-1 mutants by treating the animals with pH-buffering bicarbonate, suggesting the pathogen susceptibility is a function of the acidity of the intestinal pH. Furthermore, we use KR35 to show that upon infection by pathogens, the intestinal pH becomes neutral in a wild type, but less so in pbo-1 mutants. C. elegans is known to increase production of reactive oxygen species (ROS), such as H2O2, in response to pathogens, which is an important component of pathogen defense. We show that pbo-1 mutants exhibited decreased H2O2 in response to pathogens, which could also be partially restored in pbo-1 animals treated with bicarbonate. Ultimately, our results support a model whereby PBO-1 functions during infection to facilitate pH changes in the intestine that are protective to the host.
... Loss of hyphal growth was reported to attenuate killing in both C. elegans and in mammals [17,65]. Although there has been considerable study of the host response to E. faecalis and some virulence determinants in C. elegans have been identified [34,[66][67][68], much of this work has been done on agar. At least one acute virulence factor, the protease cytolysin, is shared by C. elegans and mammalian models [34], but further study will be necessary to evaluate whether this factor is important in this liquid assay. ...
Article
Full-text available
Colonization is generally considered a prerequisite for infection, but this event is context-dependent, as evidenced by the differing ability of the human pathogen Pseudomonas aeruginosa to efficiently colonize Caenorhabditis elegans on agar but not in liquid . In this study, we examined the impact of the environment, pathogen, host, and their interactions on host colonization. We found that the transition to a liquid environment reduces food uptake by about two-fold. Also expression of specific adhesins was significantly altered in liquid-based assays for P. aeruginosa, suggesting that it may be one factor driving diminished colonization. Unexpectedly, host immune pathways did not appear to play a significant role in decreased colonization in liquid. Although knocking down key immune pathways (e.g. daf-16 or zip-2), either alone or in combination, significantly reduced survival, the changes in colonization were very small. In spite of the limited bacterial accumulation in the liquid setting, pathogenic colonization was still required for the virulence of Enterococcus faecalis. In addition, we found that a pathogen-induced dormancy was displayed by C. elegans in liquid medium after pathogen exposure, resulting in cessation of pharyngeal pumping and a decrease in bacterial intake. We conclude that poor colonization in liquid is likely due to a combination of environmental factors and host-pathogen interactions. These results provide new insights into mechanisms for colonization in different models, enabling pathogenesis models to be fine-tuned to more accurately represent the conditions seen in human infections so that new tools for curbing bacterial and fungal infections can be developed.
... Do the ShKT domains of the Tc-MUC-1 protein play roles in altering innate immune responses of host animals?-as ShKT domain-containing peroxidase and associated cuticle development of C. elegans has been reported to be involved in pathogen resistance [64][65][66]. Are mucins involved in the tuft cell activation?-which has been emerging a novel aspect for understanding intestinal immune responses to parasitic infections [67]. Addressing these questions should improve our understanding of the interplay between T. canis larvae and host cells. ...
Article
Full-text available
Toxocariasis is a neglected parasitic disease caused predominantly by larvae of Toxocara canis . While this zoonotic disease is of major importance in humans and canids, it can also affect a range of other mammalian hosts. It is known that mucins secreted by larvae play key roles in immune recognition and evasion, but very little is understood about the molecular interactions between host cells and T . canis . Here, using an integrative approach (affinity pull-down, mass spectrometry, co-immunoprecipitation and bioinformatics), we identified 219 proteins expressed by a murine macrophage cell line (RAW264.7) that interact with prokaryotically-expressed recombinant protein (r Tc -MUC-1) representing the mucin Tc -MUC-1 present in the surface coat of infective larvae of T . canis . Protein-protein interactions between r Tc -MUC-1 and an actin binding protein CFL1 as well as the fatty acid binding protein FABP5 of RAW264.7 macrophages were also demonstrated in a human embryonic kidney cell line (HEK 293T). By combing predicted structural information on the protein-protein interaction and functional knowledge of the related protein association networks, we inferred roles for Tc -MUC-1 protein in the regulation of actin cytoskeletal remodelling, and the migration and phagosome formation of macrophage cells. These molecular interactions now require verification in vivo . The experimental approach taken here should be readily applicable to comparative studies of other ascaridoid nematodes (e.g. T . cati , Anisakis simplex , Ascaris suum and Baylisascaris procyonis ) whose larvae undergo tissue migration in accidental hosts, including humans.
... ROS generated in this way activate SKN-1/Nrf via the PMK-1/p38 MAPK pathway, and SKN-1 activity promotes resistance to E. faecalis (Hoeven et al., 2011). Later work showed an unexpected role in the epidermis for two heme peroxidases, SKPO-1, a C. elegans ortholog of human lactoperoxidase, and HPX-2, which is homologous to several human heme-containing peroxidases, to protect the host against intestinal E. faecalis infection (Liu et al., 2019;Tiller and Garsin, 2014). ...
... ROS generated in this way activate SKN-1/Nrf via the PMK-1/p38 MAPK pathway, and SKN-1 activity promotes resistance to E. faecalis (Hoeven et al., 2011). Later work showed an unexpected role in the epidermis for two heme peroxidases, SKPO-1, a C. elegans ortholog of human lactoperoxidase, and HPX-2, which is homologous to several human heme-containing peroxidases, to protect the host against intestinal E. faecalis infection (Liu et al., 2019;Tiller and Garsin, 2014). ...
Chapter
In its natural habitat, C. elegans encounters a wide variety of microbes, including food, commensals and pathogens. To be able to survive long enough to reproduce, C. elegans has developed a complex array of responses to pathogens. These activities are coordinated on scales that range from individual organelles to the entire organism. Often, the response is triggered within cells, by detection of infection-induced damage, mainly in the intestine or epidermis. C. elegans has, however, a capacity for cell non-autonomous regulation of these responses. This frequently involves the nervous system, integrating pathogen recognition, altering host biology and governing avoidance behavior. Although there are significant differences with the immune system of mammals, some mechanisms used to limit pathogenesis show remarkable phylogenetic conservation. The past 20 years have witnessed an explosion of host-pathogen interaction studies using C. elegans as a model. This review will discuss the broad themes that have emerged and highlight areas that remain to be fully explored.
... Another hypodermally localized heme peroxidase-SKPO-1-was also discovered by RNAi screening that was found to be important in maintaining normal cuticle phenotype. SKPO-1 was also claimed to play a role-along with BLI-3-in host defense against pathogenic bacteria (56,66). ...
Article
Full-text available
Peroxidase enzymes can oxidize a multitude of substrates in diverse biological processes. According to the latest phylogenetic analysis, there are four major heme peroxidase superfamilies. In this review, we focus on certain members of the cyclooxygenase-peroxidase superfamily (also labeled as animal heme peroxidases) and their connection to specific NADPH oxidase enzymes which provide H2O2 for the one- and two-electron oxidation of various peroxidase substrates. The family of NADPH oxidases is a group of enzymes dedicated to the production of superoxide and hydrogen peroxide. There is a handful of known and important physiological functions where one of the seven known human NADPH oxidases plays an essential role. In most of these functions NADPH oxidases provide H2O2 for specific heme peroxidases and the concerted action of the two enzymes is indispensable for the accomplishment of the biological function. We discuss human and other metazoan examples of such cooperation between oxidases and peroxidases and analyze the biological importance of their functional interaction. We also review those oxidases and peroxidases where this kind of partnership has not been identified yet.
Article
The entomopathogenic nematode Steinernema hermaphroditum was recently rediscovered and is being developed as a genetically tractable experimental system for the study of previously unexplored biology, including parasitism of its insect hosts and mutualism with its bacterial endosymbiont Xenorhabdus griffiniae. Through whole-genome re-sequencing and genetic mapping we have for the first time molecularly identified the gene responsible for a mutationally defined phenotypic locus in an entomopathogenic nematode. In the process we observed an unexpected mutational spectrum following EMS mutagenesis in this species. We find that the ortholog of the essential C. elegans peroxidase gene skpo-2 controls body size and shape in S. hermaphroditum. We confirmed this identification by generating additional loss-of-function mutations in the gene using CRISPR-Cas9. We propose that the identification of skpo-2 will accelerate gene targeting in other Steinernema entomopathogenic nematodes used commercially in pest control, as skpo-2 is X-linked and males hemizygous for loss of its function can mate, making skpo-2 an easily recognized and maintained marker for use in co-CRISPR.
Chapter
Caenorhabditis elegans (C. elegans) is a key model organism that has been used to elucidate conserved mechanisms and components involved in development, immune response, and aging. Relevant to this chapter, C. elegans has been used to study the roles and functions of NADPH oxidases and has contributed to the overall understanding of these enzymes. Unlike mammals that have 5 NOX and 2 DUOX NADPH oxidases, the C. elegans genome only encodes two DUOX enzymes. One of these, BLI-3, is critical for viability and has ascribed biological roles in development, immune response, and aging. The second, DUOX-2, arises from a weakly expressed gene and only one report describes a phenotype associated with its loss. Therefore, this chapter will focus mostly on BLI-3, examining its sequence and structural features, its expression with respect to developmental stage and tissue location, its biological roles in development, immune response, aging, and stress response, and its regulators and functional interactors. BLI-3 structure, and function will be compared to mammalian DUOX enzymes to highlight and explore relevant similarities and differences. Finally, important gaps in knowledge and avenues for future investigation will be discussed.Keywords Caenorhabditis elegans BLI-3DUOXNADPH oxidaseBlisteredCuticle developmentImmunityAging
Article
Full-text available
The nematode Caenorhabditis elegans is commonly used as a model organism in studies of the host immune response. The worm encodes twelve peroxidase-cyclooxygenase superfamily members, making it an attractive model in which to study the functions of heme peroxidases. In previous work, loss of one of these peroxidases, SKPO-1 (ShkT-containing peroxidase), rendered C. elegans more sensitive to the human, Gram-positive pathogen Enterococcus faecalis. SKPO-1 was localized to the hypodermis of the animals where it also affected cuticle development as indicated by a morphological phenotype called “dumpy.” In this work, a better understanding of how loss of skpo-1 impacts both sensitivity to pathogen as well as cuticle development was sought by subjecting a deletion mutant of skpo-1 to transcriptome analysis using RNA sequencing following exposure to control (Escherichia coli) and pathogenic (E. faecalis) feeding conditions. Loss of skpo-1 caused a general upregulation of genes encoding collagens and other proteins related to cuticle development. On E. faecalis, these animals also failed to upregulate guanylyl cyclases that are often involved in environmental sensing. Hoechst straining revealed increased permeability of the cuticle and atomic force microscopy exposed the misalignment of the cuticular annuli and furrows. These findings provide a basis for better understanding of the morphological as well as the pathogen sensitivity phenotypes associated with loss of SKPO-1 function.
Chapter
In this chapter, we focused on the introduction of epidermal signaling pathways involved in the control of toxicity of environmental toxicants or stresses. We first introduced epidermal molecular signaling pathways, including insulin signaling pathway, peroxidase SKPO-1, collagens, SNF-12-STA-2 signaling cascade, G-protein-coupled receptors (GPCRs), and G proteins, in the regulation of toxicity of environmental toxicants or stresses. The potential effects of epidermal signals on other tissues or organs, such as neurons and intestine, were further introduced and discussed.
Article
Full-text available
Intramolecular disulfide bond formation is promoted in oxidizing extracellular and endoplasmic reticulum compartments and often contributes to protein stability and function. DUOX1 and DUOX2 are distinguished from other members of the NOX protein family by the presence of a unique extracellular N-terminal region. These peroxidase-like domains lack the conserved cysteines that confer structural stability to mammalian peroxidases. Sequence-based structure predictions suggest that the thiol groups present are solvent-exposed on a single protein surface and are too distant to support intramolecular disulfide bond formation. To investigate the role of these thiol residues, we introduced four individual cysteine to glycine mutations in the peroxidase-like domains of both human DUOXs and purified the recombinant proteins. The mutations caused little change in the stabilities of the monomeric proteins, supporting the hypothesis that the thiol residues are solvent-exposed and not involved in disulfide bonds that are critical for structural integrity. However, the ability of the isolated hDUOX1 peroxidase-like domain to dimerize was altered, suggesting a role for these cysteines in protein-protein interactions that could facilitate homodimerization of the peroxidase-like domain or, in the full-length protein, heterodimeric interactions with a maturation protein. When full-length hDUOX1 was expressed in HEK293 cells, the mutations resulted in decreased H2O2 production that correlated with a decreased amount of the enzyme localized to the membrane surface rather than with a loss of activity or with a failure to synthesize the mutant proteins. These results support a role for the cysteine residues in intermolecular disulfide bond formation with the DUOX maturation factor DUOXA1. Background: Dual oxidases (DUOXs) are membrane-bound ROS-generating enzymes. Results: Conserved DUOX cysteines localized in an N-terminal domain contribute to enzymatic maturation, independent of structural stabilization. Conclusion: Intermolecular disulfides support the interaction between DUOX enzymes and their maturation factors. Significance: This study reflects a complex profile of protein interactions required for activity and localization of the DUOX enzymes.
Article
Full-text available
The Gram-positive bacterium Enterococcus faecalis and the fungus Candida albicans are both found as commensals in many of the same niches of the human body, such as the oral cavity and gastrointestinal (GI) tract. However, both are opportunistic pathogens and have frequently been found to be coconstituents of polymicrobial infections. Despite these features in common, there has been little investigation into whether these microbes affect one another in a biologically significant manner. Using a Caenorhabditis elegans model of polymicrobial infection, we discovered that E. faecalis and C. albicans negatively impact each other's virulence. Much of the negative effect of E. faecalis on C. albicans was due to the inhibition of C. albicans hyphal morphogenesis, a developmental program crucial to C. albicans pathogenicity. We discovered that the inhibition was partially dependent on the Fsr quorum-sensing system, a major regulator of virulence in E. faecalis. Specifically, two proteases regulated by Fsr, GelE and SerE, were partially required. Further characterization of the inhibitory signal revealed that it is secreted into the supernatant, is heat resistant, and is between 3 and 10 kDa. The substance was also shown to inhibit C. albicans filamentation in the context of an in vitro biofilm. Finally, a screen of an E. faecalis transposon mutant library identified other genes required for suppression of C. albicans hyphal formation. Overall, we demonstrate a biologically relevant interaction between two clinically important microbes that could affect treatment strategies as well as impact our understanding of interkingdom signaling and sensing in the human-associated microbiome.
Article
Full-text available
Peroxidasins represent the subfamily 2 of the peroxidase-cyclooxygenase superfamily and are closely related to chordata peroxidases (subfamily 1) and peroxinectins (subfamily 3). They are multidomain proteins containing a heme peroxidase domain with high homology to human lactoperoxidase that mediates one- and two-electron oxidation reactions. Additional domains of the secreted and glycosylated metalloproteins are type C-like immunoglobulin domains, typical leucine-rich repeats, as well as a von Willebrand factor C module. These are typical motifs of extracellular proteins that mediate protein-protein interactions. We have reconstructed the phylogeny of this new family of oxidoreductases and show the presence of four invertebrate clades as well as one vertebrate clade that includes also two different human representatives. The variability of domain assembly in the various clades was analyzed, as was the occurrence of relevant catalytic residues in the peroxidase domain based on the knowledge of catalysis of the mammalian homologues. Finally, the few reports on expression, localization, enzymatic activity, and physiological roles in the model organisms Drosophila melanogaster, Caenorhabditis elegans, and Homo sapiens are critically reviewed. Roles attributed to peroxidasins include antimicrobial defense, extracellular matrix formation, and consolidation at various developmental stages. Many research questions need to be solved in future, including detailed biochemical/physical studies and elucidation of the three dimensional structure of a model peroxidasin as well as the relation and interplay of the domains and the in vivo functions in various organisms including man.
Article
Full-text available
In response to pathogen infection, the host innate immune system activates microbial killing pathways and cellular stress pathways that need to be balanced because insufficient or excessive immune responses have deleterious consequences. Recent studies demonstrate that two G protein-coupled receptors (GPCRs) in the nervous system of Caenorhabditis elegans control immune homeostasis. To investigate further how GPCR signaling controls immune homeostasis at the organismal level, we studied arrestin-1 (ARR-1), which is the only GPCR adaptor protein in C. elegans. The results indicate that ARR-1 is required for GPCR signaling in ASH, ASI, AQR, PQR, and URX neurons, which control the unfolded protein response and a p38 mitogen-activated protein kinase signaling pathway required for innate immunity. ARR-1 activity also controlled immunity through ADF chemosensory and AFD thermosensory neurons that regulate longevity. Furthermore, we found that although ARR-1 played a key role in the control of immunity by AFD thermosensory neurons, it did not control longevity through these cells. However, ARR-1 partially controlled longevity through ADF neurons.
Article
Full-text available
The conidia of the endoparasitic fungus Meria coniospora (Deuteromycetes) had different patterns of adhesion to the cuticles of the several nematode species tested; adhesion in some species was only to the head and tail regions, on others over the entire cuticle, whereas on others there was a complete lack of adhesion. After adhesion, the fungus usually infected the nematode. However, adhesion to third-stage larvae of five animal parasitic nematodes, all of which carry the cast cuticle from the previous molt, did not result in infection. M. coniospora infected animal parasitic nematodes when this protective sheath was removed. Seven preparations of sialic acid (N-acetylneuraminic acid) gave three types of response in adhesion-infection of nematodes: (i) a significant reduction in conidial adhesions; (ii) no interference with adhesion, but a 10-day delay in infection; and (iii) a delay in infection by 2 to 3 days. The current results support previous findings indicating involvement of sialic acids localized on nematode cuticles in recognition of prey by M. coniospora.
Article
Full-text available
The nematode C. elegans is attracted to nutritious bacteria and is repelled by pathogens and toxins. Here we show that RNAi and toxin-mediated disruption of core cellular activities, including translation, respiration, and protein turnover, stimulate behavioral avoidance of normally attractive bacteria. RNAi of these and other essential processes induces expression of detoxification and innate immune effectors, even in the absence of toxins or pathogens. Disruption of core processes in non-neuronal tissues was sufficient to stimulate aversion behavior, revealing a neuroendocrine axis of control that additionally required serotonergic and Jnk kinase signaling pathways. We propose that surveillance pathways overseeing core cellular activities allow animals to detect invading pathogens that deploy toxins and virulence factors to undermine vital host functions. Variation in cellular surveillance and endocrine pathways controlling behavior, detoxification, and immunity selected by past toxin or microbial interactions could underlie aberrant responses to foods, medicines, and microbes.
Article
Full-text available
Infected animals will produce reactive oxygen species (ROS) and other inflammatory molecules that help fight pathogens, but can inadvertently damage host tissue. Therefore specific responses, which protect and repair against the collateral damage caused by the immune response, are critical for successfully surviving pathogen attack. We previously demonstrated that ROS are generated during infection in the model host Caenorhabditis elegans by the dual oxidase Ce-Duox1/BLI-3. Herein, an important connection between ROS generation by Ce-Duox1/BLI-3 and upregulation of a protective transcriptional response by SKN-1 is established in the context of infection. SKN-1 is an ortholog of the mammalian Nrf transcription factors and has previously been documented to promote survival, following oxidative stress, by upregulating genes involved in the detoxification of ROS and other reactive compounds. Using qRT-PCR, transcriptional reporter fusions, and a translational fusion, SKN-1 is shown to become highly active in the C. elegans intestine upon exposure to the human bacterial pathogens, Enterococcus faecalis and Pseudomonas aeruginosa. Activation is dependent on the overall pathogenicity of the bacterium, demonstrated by a weakened response observed in attenuated mutants of these pathogens. Previous work demonstrated a role for p38 MAPK signaling both in pathogen resistance and in activating SKN-1 upon exposure to chemically induced oxidative stress. We show that NSY-1, SEK-1 and PMK-1 are also required for SKN-1 activity during infection. Evidence is also presented that the ROS produced by Ce-Duox1/BLI-3 is the source of SKN-1 activation via p38 MAPK signaling during infection. Finally, for the first time, SKN-1 activity is shown to be protective during infection; loss of skn-1 decreases resistance, whereas increasing SKN-1 activity augments resistance to pathogen. Overall, a model is presented in which ROS generation by Ce-Duox1/BLI-3 activates a protective SKN-1 response via p38 MAPK signaling.
Article
Caenorhabditis Elegans has been a popular model organism for biological research for over thirty years and has been used to investigate many aspects of animal development, for example apoptosis, the Hox genes, signal transduction pathways, and the development of the nervous system. It has recently taken on new importance with the publication of the entire genome sequence in 1998. The first chapter gives all the basic information on C. elegans required to use it: it's natural history, anatomy, life cycle, development, and evolution. Information on how to obtain, grow, and maintain C. elegans for use as a model system is given in Chapter 4. Chapters 2 and 3 describe the genome project and show how to use genome sequence information by searching the database for homologues using different search methods and then how to analyse the search data. The next chapter gives the essential practical details of transformation and common uses for the technique. Chapter 6 covers reverse genetics and describes strategies for gene inactivation that are known to work in C elegans: epigenetic inactivation and mutational germ line inactivation. Chapter 7 is designed to help the user analyse phenotype by microscopy and includes Normaski, fluorescence, 4-dimensional, and electron microscopy. Techniques for studying the neurobiology of C. elegans are given in chapter 8. Chapter 9 describes the three commonly used approaches for studying gene expression and Chapter 10 deals with the common methods of molecular biology essential for gene characterization. C. elegans is not the ideal organism for biochemical studies, but chapter 11 describes several procedures for producing biochemically useful quantities of pure tissues. The final chapter is about conventional genetics and details the standard procedures for selfing and crossing; mutagenesis and mutant screening; characterization of mutants; gene mapping; temperature-shift experiments and mosaic analysis. Caenorhabditis Elegans: A Practical Approach will therefore provide all the background information necessary for use of C. elegans as a model system.
Article
The CYP4 family of cytochrome P450 enzymes and the mammalian peroxidases covalently bind their heme groups. In the CYP4 enzymes this involves one, and in the peroxidases two, covalent ester links between heme methys and carboxylic acid side-chains. In addition, in myeloperoxidase, a methionine forms a sulfonium link to a heme vinyl group. The ester links in both classes of proteins are autocatalytically formed. One role of the covalent bonds in peroxidases is to protect the heme group from modification by catalytically generated reactive products. The covalent heme bond in CYP4 enzymes contributes to their high preference for omega- over (omega-1)-hydroxylation.