Serovar-Dependent Differences in Hfq-Regulated Phenotypes in Actinobacillus Pleuropneumoniae

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Pathogens and Disease, 78, 2020, ftaa066

doi: 10.1093/femspd/ftaa066
Advance Access Publication Date: 23 October 2020
Research Article

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


RESEARCH ARTICLE

Serovar-dependent differences in Hfq-regulated


phenotypes in Actinobacillus pleuropneumoniae
Josicelli Souza Crispim1,† , Thyara Ferreira da Silva1 , Newton
Moreno Sanches1 , Giarlã Cunha da Silva1 , Monalessa Fábia Pereira1 , Ciro
César Rossi1 , Yanwen Li2 , Vanessa Sofia Terra3 , Prerna Vohra3 , Brendan
W. Wren3 , Paul R. Langford2 , Janine T. Bossé2 and Denise Mara
Soares Bazzolli1, *,‡
1
Laboratório de Genética Molecular de Bactérias, Departamento de Microbiologia, Instituto de Biotecnologia
Aplicada à Agropecuária – BIOAGRO, Universidade Federal de Viçosa, Viçosa, 36570–900, Brazil, 2 Section of
Paediatric Infectious Disease, Imperial College London, St Mary’s Campus, London W2 1PG, UK and
3
Department of Pathogen Molecular Biology, London School of Hygiene and Tropical Medicine, Keppel Street,
London, WC1E 7HT, UK

Corresponding author: Laboratório da Genética Molecular de Bactérias, Departamento de Microbiologia, Instituto de Biotecnologia Aplicada à
Agropecuária - BIOAGRO, Universidade Federal de Viçosa, Viçosa, MG - Brazil. CEP: 36570-900. E-mail: dbazzolli@ufv.br
One sentence summary: Serovar-dependent differences identified in regulation of complex phenotypes by the RNA chaperone Hfq in the pig pathogen
A. pleuroneumoniae indicate the importance of strain selection and interpretation of results when analysing global gene regulator function.
Editor: Juliana Campos Junqueira.

Josicelli Souza Crispim, http://orcid.org/0000-0002-1625-1088

Denise Mara Soares Bazzolli, http://orcid.org/0000-0002-0371-6966

ABSTRACT
The RNA chaperone Hfq regulates diverse processes in numerous bacteria. In this study, we compared phenotypes (growth
rate, adherence, response to different stress conditions and virulence in Galleria mellonella) of wild-type (WT) and isogenic
hfq mutants of three serovars (1, 8 and 15) of the porcine pathogen Actinobacillus pleuropneumoniae. Similar growth in rich
broth was seen for all strains except Ap1hfq, which showed slightly reduced growth throughout the 24 h time course, and
the complemented Ap8hfqC mutant had a prolonged lag phase. Differences were seen between the three serovar WT
strains regarding adherence, stress response and virulence in G. mellonella, and deletion of hfq affected some, but not all of
these phenotypes, depending on serovar. Complementation by expression of cloned hfq from an endogenous promoter only
restored some WT phenotypes, indicating that complex regulatory networks may be involved, and that levels of Hfq may be
as important as presence/absence of the protein regarding its contribution to gene regulation. Our results support that Hfq
is a pleiotropic global regulator in A. pleuropneumoniae, but serovar-related differences exist. These results highlight the
importance of testing multiple strains/serovars within a given species when determining contributions of global regulators,
such as Hfq, to expression of complex phenotypes.

Keywords: Pasteurellaceae; RNA chaperone; Galleria mellonella; virulence; stress

Received: 22 July 2020; Accepted: 21 October 2020



C The Author(s) 2020. Published by Oxford University Press on behalf of FEMS. All rights reserved. For permissions, please e-mail:

journals.permissions@oup.com

1
2 Pathogens and Disease, 2020, Vol. 78, No. 9

INTRODUCTION the E. coli strains in LB broth and agar. Chloramphenicol (1 or


20 μg/mL, for A. pleuropneumoniae and E. coli, respectively) or
The Gram-negative bacterium Actinobacillus pleuropneumoniae
kanamycin (75 μg/mL) was added to the medium when required.
causes porcine pleuropneumonia, a disease that has a nega-
Salt-free LB agar (10 g tryptone, 5 g yeast extract and 15 g agar per
tive economic impact on the worldwide swine industry (Sassu
L) supplemented with 10% filter-sterilized sucrose, 10% horse
et al. 2018). Currently, eighteen serovars are recognized based
serum (TCS Biosciences) and 10 μg/mL NAD (Sigma-Aldrich) was

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


on capsular polysaccharides (Bossé et al. 2018). All serovars are
used for counter selection of A. pleuropneumoniae mutants, as
pathogenic, but some are more virulent than others, e.g. serovar
previously described (Bossé et al. 2014). E. coli MFDpir (Ferrières
3 is rarely pathogenic, but serovar 1 is considered of high viru-
et al. 2010) and Stellar (Clontech) strains were used in conjuga-
lence (Rogers et al. 1990; Frey 2011). In part, this is related to the
tion and transformation assays, respectively.
combinations of repeat in toxin (RTX) toxins (ApxI-III) present in
different serovars (Frey 2011). Other virulence factors, some of
which also differ depending on serovar, have been reported for Strain construction
A. pleuropneumoniae including: capsule, lipopolysaccharide (LPS),
fimbriae, outer membrane proteins, iron-binding proteins and Unless otherwise stated, all PCRs were performed using
the ability to form biofilms [reviewed in Bossé et al. 2002; Chiers CloneAmpTM HiFi PCR Premix (Takara), and genomic DNA from
et al. 2010]. In addition, roles in virulence have been indicated for the serovar 8 strain MIDG2331 (Bossé et al. 2016) was used as the
global regulators of gene expression such as RpoE (Bossé et al. template for amplification of A. pleuropneumoniae products for
2010), HlyX (Buettner et al. 2009), ArcA (Buettner et al. 2008) and cloning. For direct cloning into the T-vector, pGEM-T (Promega),
Hfq (Zhou et al. 2008; Subashchandrabose et al. 2013), the latter products amplified with the CloneAmpTM HiFi polymerase were
being the subject of this study. first A-tailed by incubation at 70◦ C for 30 minutes with 0.2 mM
Hfq was first identified in 1972 as regulator of phage Qβ dATP and 5 U of Taq polymerase (Promega), according to manu-
RNA replication in Escherichia coli (Franze de Fernandez, Hay- facturer’s instructions. All initial constructs were transformed
ward and August 1972). It is now known that, through its inter- into E. coli Stellar cells (Takara), according to manufacturer’s
actions with small RNAs (sRNAs), Hfq is a major global regulator protocol, with selection of clones on media containing chlo-
of gene expression in a wide variety of bacteria (Vogel and Luisi ramphenicol or kanamycin, as appropriate. A description of all
2011; Sobrero and Valverde 2012; Feliciano et al. 2016; Dos San- primers used in this study is given in Table 2.
tos, Arraiano & Andrade 2019). In E. coli, deletion of the hfq gene The hfq and hfq::3XFLAG strains of A. pleuropneu-
results in pleiotropic changes when compared to wild-type (WT), moniae serovars 8 and 15, and the Ap1hfqcatsacB and
including increased cell size, reduced growth rate, increased Ap1hfq::3XFLAGcat strains of serovar 1, were obtained using the
sensitivity to ultraviolet light and other processes (Tsui, Leung previously described natural transformation technique (Bossé
and Winkler 1994; Kendall et al. 2011). A role for Hfq in viru- et al. 2014). Briefly, the sequence comprising the hfq gene, and
lence, as adjudged in vivo or by surrogate markers such as tol- ∼600 bp to either side, was amplified using primers 1 and 2
erance to stress and ability to form biofilms, has been shown (Table 2), A-tailed and cloned in pGEM-T (Promega), resulting
for many Gram-negative bacteria including: Neisseria meningitidis in pThfqFlank. A selection/counterselection cassette, catsacB,
(Fantappiè et al. 2009), Haemophilus influenzae (Hempel et al. 2013), was amplified from pUSScatsac (Bossé et al. 2014) using primers
Yersinia enterocolitica (Kakoschke et al. 2014), Brucella melitensis 3 and 4. pThfqFlank was opened by inverse PCR using primers
(Cui et al. 2013), Salmonella enterica serovar Typhimurium (Behere 5 and 6 designed with 15 bp overhangs to allow In-Fusion
et al. 2016), Pasteurella multocida (Mégroz et al. 2016), Xanthomonas (Takara) cloning, according to manufacturer’s instructions, of
campestris (Lai et al. 2018) and Bordetella pertussis (Hayes et al. the catsacB cassette in place of the deleted hfq gene to generate
2020). plasmid pThfqcatsacB. This plasmid was transformed into
With A. pleuropneumoniae, it has also been established that A. pleuropneumoniae serovars 1, 8 and 15 to obtain hfqcatsacB
Hfq has a role in virulence. Both Zhou et al. (2008) and Sub- mutants, as previously described (Bossé et al. 2014).
ashchandrabose et al. (2013) demonstrated that A. pleuropneumo- An unmarked deletion construct was made by amplifying
niae hfq mutants of serovar 1 strains Shope 4074 and AP 93-9, the flanking regions to either side of hfq, using primers 1 and
respectively, were less virulent in pigs. In addition, an hfq mutant 7 for the left flank, and 2 and 8 for the right flank. Primers 7
of a clinical serovar 8 isolate, MIDG2331, was attenuated in the and 8 contain 15 bp overhangs to allow direct fusion of the two
Galleria mellonella (wax moth) model of infection (Pereira et al. amplicons by over-lap extension (OE) PCR (Bossé et al. 2014).
2015). In vitro, the AP 93-9 serovar 1 hfq mutant was defective in The OE PCR product was cloned into pGEM-T (Promega), result-
biofilm formation and was more sensitive to superoxide stress ing in pThfq. A construct containing hfq with a 3 fusion to a
(Subashchandrabose et al. 2013). In this study, we undertook a 3XFLAG tag (3x GAT TAC AAG GAT GAC GAT GAC AGG) was also
comparative analysis of the effect of hfq mutagenesis on three generated. The 3XFLAG tag was amplified from pDOC-F (acces-
different serovars of A. pleuropneumoniae to determine if regula- sion number GQ889496), a generous gift from S. Wigneshweraraj,
tion of different Hfq phenotypes is serovar dependent. using primers 9 and 10. The pThfqFlank construct was opened by
inverse PCR using primers 11 and 12, and the 3XFLAG amplicon
was inserted by In-Fusion cloning, creating pThfq::3XFLAG.
MATERIALS AND METHODS To obtain the unmarked hfq and hfq::3XFLAG mutant
strains, the hfqcatsacB mutants were subjected to a second nat-
Bacterial strains, growth conditions and maintenance
ural transformation with linearized plasmids, either pThfq or
The A. pleuropneumoniae strains used in this study (listed in pThfq::3XFLAG, with counterselection on LB-SSN plates (Bossé
Table 1) were routinely grown at 37◦ C with 5% CO2 in brain heart et al. 2014). As counterselection with the unmarked deletion con-
infusion (BHI; Difco) broth and agar supplement with 10 μg/mL structs was not successful with the Ap1hfqcatsacB mutant, an
nicotinamide adenine dinucleotide (NAD; Sigma-Aldrich) and alternate construct, pThfq::3XFLAGcat, was used to obtain the
Crispim et al. 3

Table 1. Strains and plasmids used in this study.

Strains and plasmids Description Source or reference

A. pleuropneumoniae
Serotype 1
Shope 4074 WT Shope 4074 WT ATCC 27 088
Ap1hfqcatsacB hfq mutant of Shope 4074 This study

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


Ap1hfqcatsacBC Complemented strain This study
Ap1hfq::3XFLAGcat WT containing a 3XFLAG tag replacing the last codon of the hfq gene. This study
Serotype 8
MIDG2331 WT Serotype 8 clinical isolate from UK (Bossé et al. 2016)
Ap8hfqa hfq mutant of MIDG2331 This study
Ap8hfqC Complemented strain This study
Ap8hfq::3XFLAG WT containing a 3XFLAG tag replacing the last codon of the hfq gene. This study
Serotype 15
HS143 WT HS143 WT (Blackall et al. 2002)
Ap15hfqcat hfq mutant This study
Ap15hqC Complemented strain This study
Ap15hfq::3XFLAG WT containing a 3XFLAG tag replacing the last codon of the hfq gene. This study
E. coli
Stellar Competent cell: F–, endA1, supE44, thi-1, recA1, relA1, gyrA96, phoA, Takara
80d lacZ M15,  (lacZYA—argF) U169,  (mrr-hsdRMS-mcrBC), mcrA,
λ–
MFDpir Conjugative cell: MG1655 RP4-2-Tc::[Mu1::aac(3)IV- aphA- nic35- (Ferrières et al. 2010)
Mu2::zeo] dapA::(erm-pir) recA. Strain used to introduce pMIDG hfq
in A. pleuropneumoniae hq strains.
Plasmids
pUSScatsac Template DNA for amplification of catsacB cassette which contains (Bossé et al. 2014)
DNA uptake sequences for natural transformation into A.
pleuropneumoniae.
pThfqFlank Plasmid pGEM-T containing 600 nucleotides upstream hfq gene, hfq This study
gene and 600 nucleotides downstream hfq gene.
pThfqcatsacB Plasmid pThfq containing the hfq gene disrupted by catsacB cassette. This study
pThfq3XFLAG Plasmid pGEM-T containing the hfq gene with a 3XFLAG tag in the This study
region 3 of the hfq gene.
pThfq3XFLAGcat Plasmid pGEM-T containing the hfq gene with a 3XFLAG tag in the This study
region 3 of the hfq gene followed by the cat gene.
pThfq::3XFLAGcatsacB Plasmid pGEM-T containing the hfq gene with a 3XFLAG tag in the This study
region 3 of the hfq gene disrupted by catsacB cassette.
pMIDG hfq pMIDG plasmid (Bossé et al. 2009) containing the hfq gene under This study
promoter inside of the miaA gene. Strain used to complement the
hfq strains.

a
Although this mutant strain has been used in a previous study by our group (Pereira et al. 2015), this is the first description of the generation of the mutation.

FLAG-tagged mutant. Primers 13 and 14 were used for ampli- Hfq promoter analysis and mutation complementation
fication of the cat cassette of plasmid pUSScatsac. The 3xFLAG
tag was amplified from pDOC-F using primers 9 and 15. Primers For a better understanding of the promoter(s) involved in the
14 and 15 contain 15 bp overhangs to allow direct fusion of the transcription of the hfq gene, a prediction of the hfq operon
two amplicons by OE PCR, as above. The pThfqFlank construct was performed using DOOR (Database for prOkaryotic OpeR-
was opened by inverse PCR using primers 11 and 12, and the ons) (Mao et al. 2009), followed by prediction of promoters using
3xFLAGcat amplicon was inserted by In-Fusion cloning, creating BPROM (Solovyev, Shahmuradov and Salamov 2010) and visual
pThfq::3XFLAGcat. This plasmid was transformed, as previously analysis of the sequences.
described (Bossé et al. 2014), into A. pleuropneumoniae serovar 1 to Complementation of the hfq mutants was achieved by
obtain the Ap1hfq::3XFLAGcat mutant. Deletion of hfq, or the cloning the hfq gene, with three of the predicted endogenous
presence of FLAG-tagged hfq, in the chromosome of respective promoters, into the low copy plasmid pMIDG100 (O’Dwyer et al.
mutants was confirmed by PCR and sequencing using primers 2004; Bossé et al. 2009). Primers 22 and 23 were used to amplify
1 and 2. RT-PCR analysis using cDNA from both WT and hfq the sequence from 850 bp upstream, to 72 bp downstream, of hfq.
mutant strains was performed with primer pairs 16 and 17, 18 The vector pMIDG100 was digested with EcoRI and BstBI (New
and 19, as well as 20 and 21 (for detection of expression of hflX, England Biolabs) and the 1.2 kb PCR product was inserted using
miaA and hfq, respectively) in order to confirm that deletion of In-Fusion cloning, as above. The plasmid pMIDG hfq was trans-
hfq did not affect expression of the flanking genes. As a positive formed into E. coli MFDpir (Ferrières et al. 2010) with selection
control for each primer pair, gDNA from the WT strain was used. on LB agar containing kanamycin (75 μg/mL), prior to conjuga-
The presence of expressed FLAG-tagged Hfq was confirmed by tion into the A. pleuropneumoniae hfq strains to obtain the com-
Western blot using anti-FLAG antibodies (see below). plemented (hfqC) strains. Confirmation of the presence of the
4 Pathogens and Disease, 2020, Vol. 78, No. 9

Table 2. Primers used in this study.

N◦ Primer Oligonucleotide sequence (5 to 3 ) Description

1 hfqflank for TTCCGGTGGAAGTAATTAGCGTAGA For amplification of the hfq cassette.


2 hfqflank rev ATATCCGCTTTCTGACGAGTTTTGC
3 cat deltahfq ATCTTTACAAGATCCTACAAGCGGTCGGCAATAAGTTACC For amplification of catsacB cassette
containing 15 bp overhangs (underlined)

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


complimentary to pThfqFlank opened by
inverse PCR with primers 5 and 6.
4 sac deltahfq CGCAACCGCTTCAACGAATTGCGTGAAGCTCGAGGTATG
5 deltahfq invcat TGCCGACCGCTTGTAGGATCTTGTAAAGATTGACCTTTTGC For inverse PCR of pThfqFlank to remove all
but 59 bp of the hfq gene and adding 15 bp
overhangs (underlined) complementary to
the catsacB cassette generated with primers
3 and 4.
6 deltahfq invsac AGCTTCACGCAATTCGTTGAAGCGGTTGCGGATAAAGC
7 deltahfq 1 CGCAACCGCTTCAACGGATCTTGTAAAGATTGACCTTTTGC For generation of hfq construct. Addition
of 15 bp overhangs (underlined) allow
direct fusion of left flank amplified using
primers 1 and 7 to right flank generated
using primers 2 and 8.
8 deltahfq 2 ATCTTTACAAGATCCGTTGAAGCGGTTGCGGATAAAGC
9 FLAG hfq GTTGCGGATAAAGCGGGTACCGACTACAAAGACCATGAC For amplification of the 3XxFLAG tag
containing 15 bp overhangs (underlined)
complementary to pThfqFlank opened by
inverse PCR with primers 11 and 12.
10 FLAG hflX TTGGTATCTGATCGGCTCCAGCCTACATTACTATTTATCG
11 hfq inv1 CGCTTTATCCGCAACCGCTTCAAC For generation of pThfq::3XFLAG
12 hfq inv2 CCGATCAGATACCAAATACAGATG
13 cat FLAG TAATGTAGGCTGGAGGTACAAGCGGTCGGCAATAGTTACC For amplification of the cat cassette
containing 15 bp overhangs (underlined)
complimentary to 3XFLAG and pThfqFlank
opened by inverse PCR with primers 11 and
12.
14 cat hflX TTGGTATCTGATCGGGAAGTGCGGTATGCCGTCTGAAC
15 FLAG cat GCCGACCGCTTGTACCTCCAGCCTACATTACTATTTATCG For amplification, in combination with
primer 9, of the 3xFLAG cassette containing
15 bp overhangs (underlined)
complementary to cat cassette and
pThfqFlank opened by inverse PCR with
primers 11 and 12.
16 hflX for CACGAGCTTAGTCCGTCACA For RT-PCR analysis of hflX expression.
17 hflX rev AATGCTACCCGCTGTATGCT
18 miaA for TAATGGGTCCAACGGCTTCG For RT-PCR analysis of miaA expression.
19 miaA rev CACTGTTCCAACCTCGCAGCCAAG
20 EcoRI hfq GCGCGAATTCAGGAAAAGAAAATGGCAAAAGGTCAATCT For RT-PCR analysis of hfq expression.
21 hfq SacI GCGCGAGCTCATTATTCCGCTTTATCCGCAACCGC
22 hfqMIDG for GCTCAAGCTTCGAATTCGAGCTTGCCCCTCACCGCTTGATTG For amplification of hfq gene with its own
promoter region and containing 15 bp
overhangs complementary to pMIDG100
cut with EcoRI and BstBI.
23 hfqMIDG rev TTGGGATCTTTCGAAGCGTTTTCATCTGTATTTGGTATCTG

gene was performed by PCR and sequencing of the intact gene In order to verify expression of Hfq during growth in broth
using the same primers described above. culture, each of the three serovar hfq::3XFLAG strains were inoc-
ulated into 200 mL of broth (initial OD600 0.01) and then aliquoted
into seven flasks of 20 mL each. At time points (1, 2, 3, 4, 6, 8
Growth rate and Hfq expression
and 12 h), one of each serovar culture was centrifuged at 9000x
For growth curves, the A. pleuropneumoniae WT and mutant g, and the resulting pellets were re-suspended in 1 mL of lysis
strains were cultivated in 20 mL of broth in Erlenmeyer flasks buffer (20 mM Tris-HCl, 1 mM EDTA and pH 7.4) and disrupted by
incubated at 37◦ C for 24 h with agitation (180 rpm). Optical den- mechanical lysis using Matrix B tubes (MP Biomedicals). For each
sity at 600 nm (OD600 ) was measured every hour for the first 12 sample, 10 μg of soluble protein were applied to wells of 4%–
h, and then at 24 h, using an Ultrospec 10 (GE Healthcare Life 12% NuPAGE Bolt BisTris Plus (Life Technologies—BG04120BOX)
Sciences). gels. Following electrophoretic separation, the proteins were
Crispim et al. 5

transferred to nitrocellulose membrane (iBlot 2 NC Regular Statistical analysis


Stacks; Life Technologies—IB23001) using the iBlot 2 system (Life
Technologies—IB21001). The membrane was processed, as pre- Data from growth curves and adhesion to A549 cells and
viously described (Beddek et al. 2004), using an anti-FLAG mon- polystyrene microtiter plates were analyzed by Tukey’s test used
oclonal (Sigma) as the primary antibody, and detection using to compare means using R v.2.13.0. The differences in G. mel-
ECLTM Western blotting detection reagents (GE Healthcare) and lonella survival were calculated by using the log-rank test using
Hyperfilm ECL (GE Healthcare). R v.2.13.0. A P < 0.05 was considered to be statistically signifi-
cant. All the assays were done in experimental and biologic trip-

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


licates.
Bacterial adhesion
Bacterial adhesion to three different surfaces was investigated. RESULTS
Adhesion to epithelial A549 cells was determined as previously Construction A. pleuropneumoniae hfq mutants
described by Cuccui et al. (2017), and adherence to polystyrene
microtiter plates (Kasvi—K12-096) following growth for 24 h at As previously reported (Subashchandrabose et al. 2013), the hfq
37◦ C was visualized using crystal violet, as described by Kaplan gene in A. pleuropneumoniae is located in the miaA-hfq-hflX locus,
and Mulks (2005). For the third adhesion assay, strains were inoc- as it is in E. coli (Tsui, Leung and Winkler 1994). This locus is
ulated in vials containing 1 cm2 steel coupons, as described pre- shown in Fig. 1A, with all detected promoters indicated, as well
viously by Moen et al. (2015). Briefly, the vials were incubated at as locations of the priming sites used for PCR amplification of
37◦ C for a period of 24 h. Cultures were then fixed to the steel products used to construct the various plasmids. In order to
coupons with 2.5% glutaraldehyde in 0.05 M phosphate buffered determine the role of Hfq in A. pleuropneumoniae serovars 1, 8
saline (PBS) and dehydrated in a graded ethanol series up to and 15, we generated isogenic mutants lacking 220 nucleotides,
100%. The cells were dried using a CPD 030 critical point dryer leaving a truncated hfq gene having only 29 nucleotides in the
(Bal-Tec) and shadowed with gold using a Sputter Coater (Elec- 5 region and 30 nucleotides in the 3 region. Clean deletion
tron Microscopy Sciences) prior to visualization with a scanning mutants were generated for serovars 8 and 15, whereas counter
electron microscope (VP1430; LEO). selection was not successful with the serovar 1 mutant, leaving
the catsacB insertion in place of the deleted 220 bases. In order
to aid in evaluation of Hfq expression under stress conditions,
Stress tolerance we also generated isogenic strains where the native hfq was
replaced with hfq additionally encoding a C-terminal 3XFLAG tag
The following agents and their concentrations were used in BHI- (followed by the cat gene in the serovar 1 strain). Absence of any
NAD agar to investigate the sensitivity of the hfq strains to polar effects on miaA and hflX expression in the hfq mutants
different stress conditions: 1.5% NaCl; pH 6.0 and 6.5 (adjusted was confirmed by RT-PCR, with representative results for serovar
using HCl); 1.25 mM H2 O2 ; 4% ethanol; and cultivation at 42◦ C. 8 shown in Fig. 1B. Expression of Hfq by the serovar 1, 8 and
Bacterial cultures with initial OD600 of 1.0 were serially diluted 15 Hfq::3XFLAG strains during growth in broth culture was con-
in PBS to 10−7 , and 10 μL of each 10-fold dilution were applied firmed by Western blotting (Fig. 1C).
on each selective stress agar in square plates (688 102; Greiner
Bio-One). As control, cultures were similarly plated on BHI-NAD
agar containing no stress agent. All plates were cultured at 37◦ C, Growth rate and Hfq expression
except the temperature stress plate, which was incubated at
Only the Ap1hfqstrain had a reduced growth rate in BHI-NAD,
42◦ C. The growth of strains was compared between the control
and the complemented Ap8hfqC strain had a prolonged lag
and test plates.
phase, compared to its isogenic WT strain, which was signif-
icant using the Tukey’s test (P < 0.05) (Fig. 2A). Western blot
results for the FLAG-tagged mutants of each serovar showed
Virulence in G. mellonella
that Hfq expression was detectable at all time points assayed
The Galleria mellonella larvae used in this study were reared in (Fig. 2B), with apparent slight increase in Hfq expression for the
our laboratory, kept at 28◦ C in darkness and fed an artificial diet. Ap8hfq::3XFLAG and Ap15hfq::3XFLAG, but not Ap1hfq::3XFLAG
On the day of the experiment, last-instar larvae, each weigh- strains over the time course.
ing 250–300 mg, were selected and kept in the same environ-
mental conditions until inoculation, following our previously
Bacterial adhesion
described methods (Pereira et al. 2015; Pereira et al. 2018, Blanco
et al. 2017). Briefly, A. pleuropneumoniae cultures were grown to The WT strains of the different serovars tested showed marked
mid-exponential phase and inocula consisting of 10 μl of seri- differences in adhesion to A549 epithelial cells, with serovar 8
ally diluted cell suspensions, varying from 103 to 107 CFU per being most, and serovar 15 least, adherent (Fig. 3A). Reduction of
larva (n = 10 larvae per dilution), were injected into the haemo- adherence was significant for the Ap1hfq and Ap15hfq strains
coel of the first right pro-leg. The larvae were incubated at 37◦ C, (P < 0.05), but not Ap8hfq, relative to their isogenic WT strains
in the dark, and analyzed according to survival at 24, 48, 72 and (Fig. 3A). Instead of restoring WT levels, expression of hfq from
96 h post infection. Larvae were considered as dead if they did the complementation vector further reduced adherence for all
not respond to touch stimuli. Survival curves were plotted using serovars (Fig. 3A), though the difference was only significant for
the Kaplan-Meier method (Goel, Khanna and Kishore 2010). For Ap15hfqC (P < 0.05). All hfq mutants had reduced adhesion to
the evaluation of bacterial load, the larval haemolymph was col- polystyrene compared to their WT strains (P < 0.05) (Fig. 3B), with
lected at 0, 1, 2, 4 and 24 h after infection. Thereafter, the CFU/mL the Ap8hfq strain showing the greatest reduction. In contrast
were determined. Larvae inoculated with PBS were used as neg- to the assay using A549 epithelial cells, all of the complemented
ative controls for the assay. strains showed increased adhesion to polystyrene compared to
6 Pathogens and Disease, 2020, Vol. 78, No. 9

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


Figure 1. Generation and confirmation of hfq mutant strains. (A) Genomic organization of the miaA, hfq and hflX genes in A. pleuropneumoniae. Predicted promoter
sequences are indicated by the bent arrows labelled P1 to P4, and a predicted transcriptional terminator is indicated by a stem-loop structure downstream of hfq. The
primers used in mutant construction, cloning and RT-PCR, are represented by arrows bellow their targets, numbered according to their identification in Table 2. (B)
RT-PCR analysis of possible polar effects due to deletion of hfq, showing representative results for MIDG2331. PCR was performed with the products of cDNA synthesis
from RNA template of either the WT or hfq strain (as indicated), both with (RT+) and without (RT-) the addition of reverse transcriptase. Note that the genomic DNA
(gDNA) control used to confirm primer function and product size for each primer pair was from the WT strain only, thus a product for hfq amplification is seen as a
comparison for the lack of amplification by RT-PCR from the hfq strain. Amplification of the target sequences in hfq, miaA and hflX was achieved with the primer pairs
20/21, 18/19 and 16/17, respectively. M = molecular weight marker (DNA Marker Quick-load 100bp DNA ladder, Neb Biolabs). (C) Western blot showing the detection of
the 14 kDa Hfq::3XFLAG protein. For each hfq::3XFLAG strain of A. pleuropneumoniae serovars 1 (Ap1), 8 (Ap8) and 15 (Ap15), ten micrograms of soluble protein from early
stationary phase culture were separated by SDS-PAGE and transferred to nitrocellulose membrane for detection using an anti-Flag antibody. The molecular weight
marker lane (M = SeeBlue Plus2; Invitrogen) from the corresponding stained gel is shown next to the blot.

Figure 2. Growth of A. pleuropneumoniae WT, hfq mutants and complemented strains. (A) Growth curve of A. pleuropneumoniae serovars 1, 8 and 15 strains. (B) Hfq::3XFLAG
expression analysis during the growth curve of the A. pleuropneumoniae strains. WT, hfq::3XFLAG (strain that express Hfq::3XFLAG), hfq (hfq mutant), hfqC (comple-
mented strain). Error bars are shown for all points in the graphs, but may not be visible in some cases.

their respective hfq mutants, however these increases were not Stress tolerance
significant and did not restore WT levels. The images of steel
coupons obtained by electron microscopy indicated that all hfq We investigated the responses of A. pleuropneumoniae serovars 1,
strains had lower adherence capacity to this surface than their 8 and 15, and their respective hfq mutants, to a variety of stress
respective WT strains (Fig. 3C). This was particularly marked inducing agents or physical stress (higher temperature) whilst
with Ap1hfq where there were, in contrast to WT, few adher- growing on BHI-NAD-agar plates, as shown in Fig. 4. The WT
ent cells, with clear complementation in the Ap1hfqC strain. strains of serovars 1, 8 and 15 showed different levels of resis-
Although adherence of the Ap8hfq and Ap15hfq mutants was tance to the different stresses, with serovar 8 being more sensi-
not completely abolished, complementation did not restore WT tive to NaCl, and serovar 15 being more resistant to ethanol, but
levels (Fig. 3C). more sensitive to elevated temperature (42◦ C) and pH 6.0, than
Crispim et al. 7

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020

Figure 3. Effect of Hfq on adherence of A. pleuropneumoniae serovars 1, 8 and 15 to biotic and abiotic surfaces. (A) The adherence to eukaryotic cells. (B) The adherence
to polystyrene microplate was examined by crystal violet reading in OD600 and the adherence capacity was determined according to WT strains. (C) The adherence to
steel coupons was examined by scanning electron microscopy. Bars: 10 μ m. Different letters inside of the Fig represent statistical significance difference among the
strains in relation the cell length. The statistical analysis was performed using Tukey’s test with P < 0.05. All the assays were conducted in experimental and biological
triplicates. WT, hfq (hfq mutant), hfqC (complemented strain).
8 Pathogens and Disease, 2020, Vol. 78, No. 9

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


Figure 4. Effect of Hfq on stress tolerance in A. pleuropneumoniae serovars 1, 8 and 15. Exponentially growing A. pleuropneumoniae strains (OD600 = 1.0;  108 cell/mL)
were exposed to different stress conditions: oxidative (1.25 mM H2 O2 ), osmotic (1.5% NaCl), alcoholic (4% ethanol), temperature (42◦ C) and pH (6.5 and 6.0). As control,
the strains were grown in BHI-NAD-agar at 37◦ C, 5% CO2 and no stressor agent. The numbers 1, 2 and 3 indicate the WT, hfq (hfq mutant) and hfqC (complemented
strain), respectively.

the other serovars. Furthermore, the respective hfq mutants serovar. At 24 h, larvae inoculated with serovar 1 strains showed
also showed differences. Unlike Ap8hfq, no growth of Ap1hfq 4% survival for the WT, 22% for hfq and 25% for hfqC indi-
and Ap15hfqwas found in the presence of 1.25 mM H2 O2 . Com- cating that the serovar 1 WT was highly virulent in this infec-
pared to the WT strains, all hfq mutants were more sensitive tion model, and the hfq mutant only slightly attenuated com-
to 1.5% NaCl, although with differing degrees of growth reduc- pared to the WT (P < 0.05), but complementation did not restore
tion. Ap8hfq and Ap15hfq were sensitive to the presence of the WT level of virulence (Fig. 1A). Similar survival rates were
ethanol, whereas only Ap15hfq showed a slight reduction in found at 96 h. The serovar 8 WT strain was not as virulent as the
growth at 42◦ C. Ap1hfq and Ap15hfq, in contrast to Ap8hfq, serovar 1 WT, with survival of G. mellonella of 60% at 24 h, and
were more sensitive to growth at pH 6.5 than their WT strains. 26% at 96 h (Fig. 1A). However, the Ap8hfq mutant was fully
Except in the case of the serovar 8 WT and mutant strains grown attenuated, with 100% survival of G. mellonella through the 96
in the presence of NaCl, restoration of WT levels of growth was h test period. Partial complementation was seen for Ap8hfqC,
achieved by complementation for all other conditions where with 51% survival of G. mellonella at 96 h. The serovar 15 WT did
deletion of hfq resulted in increased sensitivity. not appear to be virulent in the G. mellonella infection model
(over 90% survival through 96 h), and no difference was seen
for the AP15hfq and AP15hfqC strains (Fig. 1A). Analysis of
Virulence in G. mellonella
bacterial load also showed a similar decrease over time in the
The results of the virulence assay using the G. mellonella infec- larvae infected with the serovar 15 WT, hfq and hfqC strains
tion model are shown in Fig. 5. The concentration of 1.0 × 105 (Fig. 5B). The Ap1hfq and Ap8hfq strains both showed a five-
CFU per larva was chosen for graphic representation, as it was log decrease in the number of colonies per larva in the course of
found to be the best dose to allow visualization of the differences 24 h, and few bacterial (approximately 101 ) cells were observed
in the virulence profiles between WT, hfq and hfqC for each in the haemolymph after of 24 h of the experiment (Fig. 5B). In
Crispim et al. 9

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


Figure 5. Effect of Hfq on the virulence of A. pleuropneumoniae serovars 1, 8 and 15 in G. mellonella. (A) Killing was monitored after larval infection with 1 × 105 CFU of
WT, hfq and hfqC A. pleuropneumoniae strains from serovars 1, 8 and 15. The virulence attenuation was verified in the hfq strains of serovars 1 and 8 (P < 0,05). (B)
Determining of the bacterial load in G. mellonella hemolymph at 0, 1, 2, 4 and 24 h of the assay of (A) in three biological replicates. Larvae inoculated with PBS 1X were
used as negative control. WT, hfq (hfq mutant) and hfqC (complemented strain).

contrast, larvae infected with the serovar 1 and 8 WT strains are serovar specific, also contribute to virulence (Bossé et al.
showed an increase of bacterial load between 1 and 4 h, fol- 2002; Chiers et al. 2010). Initially, our goal had been to character-
lowed by less than a two-log reduction by 24 h post-infection. ize the influence of Hfq on several aspects of the physiology of
In contrast to the results for the G. mellonella survival assay, the A. pleuropneumoniae serovar 8, using MIDG2331 as a model, with
Ap1hfqC showed partial, whereas the Ap8hfqC showed no, serovar 1 and 15 strains used as controls for hfq mutation (Zhou
complementation in regard to bacterial load. et al. 2008; Subashchandrabose et al. 2013) and natural trans-
formation (Bossé et al. 2009), respectively. However, as different
DISCUSSION phenotypes for the mutants became apparent, along with dif-
ferences in the virulence profiles and other features of the WT
The role of the RNA chaperone Hfq in different bacterial species strains, we shifted our efforts towards comparing the differen-
can be variable. For example, Francisella novicida (Chambers and tial influence of the lack of Hfq in strains from these distinct
Bender 2011) and Cronobacter sakazakii (Kim et al. 2015) hfq serovars.
mutants are less resistant to oxidative stress, by contrast Staphy- A previous study by Subashchandrabose et al. (2013), char-
lococcus aureus mutants are more resistant to oxidative stress acterizing an hfq mutant of a clinical serovar 1 strain (AP 93-9),
(Liu et al. 2010). Hfq mediates the interaction of many sRNAs with showed a slight reduction in growth rate compared to the WT
their target mRNAs, in some cases leading to repression and during cultivation in rich broth, which could be complemented
in others activation of target gene expression (Vogel and Luisi by expression of hfq from a plasmid. In our current study, we
2011; Feliciano et al. 2016). The distribution of specific genes and found similar results for a hfq mutant of the serovar 1 refer-
sRNAs involved in encoding and regulating expression of com- ence strain (Shope 4074), however deletion of hfq in the serovar
plex phenotypes such as growth, biofilm formation, stress resis- 8 and 15 strains tested had no effect on growth in rich broth,
tance, and virulence can vary between different serovars/strains indicating a possible serovar-related effect.
of the same species, so it is not surprising to find the effects of The majority of clinical isolates of A. pleuropneumoniae readily
global regulators can be significantly strain as well as species form biofilms, but this phenotype tends to be lost after passage
dependent. in broth culture, suggesting repression in vitro (Kaplan and Mulks
In this study, we compared the effects of hfq mutation in 2005). Of the twelve serovar reference strains tested, only the
serovars 1 (Shope 4074; reference strain), 8 (MIDG2331; clinical serovar 5b and 11 strains (L20 and 56 513, respectively) retained
isolate) and 15 (HS143; reference strain) of A. pleuropneumoniae, the ability to adhere to glass tubes or polystyrene plates, indi-
an important swine pathogen for which there are 18 known cating possible serovar-related differences in regulation of this
serovars (Bossé et al. 2018) that can vary in their degree of vir- phenotype (Kaplan and Mulks 2005). Production of a poly-1,6-N-
ulence in pigs (Rogers et al. 1990; Sassu et al. 2018). Serovar 1 acetylglucosamine (PNAG) exopolysaccharide matrix has been
isolates, expressing Apx toxins I and II, are typically charac- shown to be the main contributor to A. pleuropneumoniae biofilm
terized by high virulence, whereas serovars 8 and 15, express- formation on abiotic surfaces (Kaplan et al. 2004; Izano et al.
ing ApxII and III, are characterized by moderate virulence (Frey 2007), with the O-antigen component of LPS also shown to
2011). In addition, factors other than RTX toxins, some of which contribute (Hathroubi et al. 2015). Components of LPS, PNAG,
10 Pathogens and Disease, 2020, Vol. 78, No. 9

pili, outer membrane proteins and glycoproteins have also been et al. 2003; Klitgaard et al. 2012). Numerous genes involved
implicated in binding of A. pleuropneumoniae to various cell lines in stress response have been identified, and their expression
(Paradis et al. 1994; Rioux et al. 1999; Van Overbeke et al. 2002; has been shown to be regulated by factors including RpoE
Auger et al. 2009; Li et al. 2012; Liu et al. 2015; Cuccui et al. 2017; and (p)ppGpp (Bossé et al. 2010; Li et al. 2015). In the study by
Liu et al. 2018), indicating a more complex phenotype than bind- Subashchandrabose et al. (2013), Hfq was shown to contribute to
ing to abiotic surfaces. resistance of their clinical serovar 1 isolate to oxidative stress,
Deletion of hfq in the AP 93-9 clinical serovar 1 strain, a strong but other sources of stress were not investigated. Here we have
biofilm former, was shown to reduce expression of pgaC, encod- shown that there was variation in the response of the three

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


ing the glycosyltransferase involved in PNAG biosynthesis, and different serovar WT strains, as well as their hfq mutants, to
completely abrogated the ability to adhere to polystyrene (Sub- different stressors. For example, the serovar 8 WT was more sen-
ashchandrabose et al. 2013). In our study, we further investigated sitive to NaCl stress, whereas the serovar 15 WT was more sen-
the contribution of Hfq to regulation of adherence of A. pleu- sitive to heat stress at 42◦ C, but more resistant to ethanol stress,
ropneumoniae to biotic and abiotic surfaces. We found that the than the other two WT strains. The hfq mutants of serovars 1
serovar 1, 8 and 15 WT strains tested showed different levels of and 15, but not serovar 8, were more sensitive to H2 O2 and pH
adhesion to various surfaces. The WT serovar 8 clinical isolate 6.5 compared to their WT parental strains. In contrast to the
(MIDG2331) showed the highest level of adherence to the A549 adhesion experiments, the stress susceptible phenotypes were
human alveolar basal epithelial cell line, which we have previ- complemented by expression of the plasmid-encoded hfq gene.
ously used for A. pleuropneumoniae adhesion assays (Cuccui et al. Although Subashchandrabose et al. (2013) previously
2017), as well as to polystyrene plates. Furthermore, the biofilm reported that deletion of hfq in A. pleuropneumoniae did not
formed by the serovar 8 WT on steel coupons showed a more result in increased sensitivity to H2 O2 or cumene hydroperox-
mature 3-D architecture, compared to those of the serovar 1 and ide, it did increase sensitivity to methyl viologen and potassium
15 WT strains. Although the serovar 15 reference strain showed tellurite—both known to generate superoxide radicals within
similar adherence to polystyrene and steel coupons when com- bacterial cells. In their study, sensitivity to these agents was
pared to the serovar 1 reference strain, it also showed the lowest tested using a disk diffusion assay, and they used a strong
level of adherence to A549 cells. biofilm forming clinical isolate of serovar 1 (AP 93-9) of A.
Deletion of hfq resulted in varying degrees of adherence pleuropneumoniae. Both of these factors could explain the dif-
reduction to the different surfaces depending on the serovar. ferences in results found in our current study. Overall, the
All three serovar hfq mutants showed slight, but significant data indicate that, as for adhesion and biofilm formation, Hfq
(P < 0.05), reduction of adherence to polystyrene, with some plays a role in stress resistance, but there are serovar- or even
restoration (not significant) of binding in each following expres- strain-dependent differences in regulation of this complex
sion of the hfq gene from the complementation vector. It is not phenotype.
clear why we did not see complete abrogation of binding to Finally, we compared virulence of serovar 1, 8 and 15 A. pleu-
polystyrene with our hfq mutants, or full complementation, as ropneumoniae WT and hfq mutants in the G. mellonella infection
was seen in the study by Subashchandrabose et al. (2013), but this model that we previously described (Pereira et al. 2015). As for the
may have been due to differences in the isolates and/or how the other phenotypes tested, there were variations between the dif-
assays were performed. Results of the adhesion assay using steel ferent WT serovars and hfq mutant strains with regards to viru-
coupons showed all hfq strains had lower adherence capac- lence in this model. Since the serovar 15 WT was avirulent under
ity to this surface than their respective WT strains, but restora- the conditions tested, no difference was seen following deletion
tion of the WT adherence phenotype was only seen with the of the hfq gene in this strain. The serovar 1 WT was the most
Ap1hfqC strain. In the assay using A549 cells, all three hfq virulent, but only showed a slight decrease, whereas the moder-
mutants showed reduced adherence compared to their respec- ately virulent serovar 8 WT was completely attenuated follow-
tive WT strains, but the level of reduction was only significant ing deletion of its hfq gene. Furthermore, complementation was
(P < 0.05) for the serovar 1 and 15 mutants. As opposed to no, or not successful for the serovar 1 mutant expressing the plasmid-
partial, complementation of the binding phenotypes, each of the encoded hfq gene, but restored almost full WT level of virulence
three serovar hfq mutants expressing the plasmid encoded hfq for the serovar 8 mutant, indicating possible differences in genes
gene showed even further reductions in binding to A549 cells, (and possible differences in gene regulation) contributing to vir-
though this was only significant (P < 0.05) for the Ap15hfqC ulence of these serovars in this infection model.
strain. Complementation of mutated phenotypes using cloned
It is difficult to determine from the current studies whether genes expressed from shuttle plasmids is always challenging,
the different in vitro adhesion phenotypes are due to serovar as factors including level of expression (due to copy number
related differences in encoded adhesion genes, or to changes in of plasmid and strength of promoter used for expression) and
gene regulation following passage of clinical isolates in the lab- indirect effects of interaction of the expressed gene with reg-
oratory, or both. Clearly the clinical serovar 8 WT strain showed ulatory network(s) can influence the overall success of restor-
the greatest adherence to all of the surfaces tested compared to ing the WT phenotype. This is especially true for complementa-
the WT serovar 1 and 15 reference strains, and deletion of hfq tion of genes encoding global regulators, such as Hfq. We tried
reduced adherence in all cases, but to different extents. These account for possible confounding issues by cloning the hfq gene
data support a role for Hfq in regulating at least some of the gene into a low copy number plasmid (pMIDG100), with expression
products contributing to adherence to the different surfaces in possible from the endogenous sigma 70 and/or sigma E pro-
each of the serovar strains tested, but other regulators such as moters included in the upstream sequence. However, we still
RpoE and H-NS (Bossé et al. 2010) and the two component sys- found that pMIDG hfq was able to complement some, but not
tems CpxA/CpxR (Li et al. 2018) and QseA/QseB (Liu et al. 2015) all of the hfq mutant phenotypes, and this was sometimes
have also been shown to be involved, and their relative contri- serovar-dependent. In the case of binding to A549 epithelial
butions to regulating this complex phenotype are unresolved. cells, expression of the plasmid copy of hfq resulted in fur-
The ability to respond to and repair damage caused by a ther reductions rather than restoration of WT levels in adher-
variety of stresses is important for the survival of A. pleuropneu- ence for all 3 serovar strains. Each of the phenotypes ana-
moniae within its host, especially during acute disease (Sheehan lyzed in this study are complex, and result from coordinated
Crispim et al. 11

expression of different genes, some of which may be regulated Blanco LAA, Crispim JS, Fernandes KM et al. Differential cellular
by Hfq-dependent sRNAs and others not. Adding to this com- immune response of Galleria mellonella to Actinobacillus pleu-
plex network, other regulators such as sigma factors and DNA ropneumoniae. Cell Tissue Res 2017;370:1–17.
binding proteins may also be involved, and the balance of these Bossé JT, Chaudhuri RR, Li Y et al. Complete genome sequence
factors likely determines the resulting phenotype. Similar obser- of MIDG2331, a genetically tractable serovar 8 clinical
vations of partial complementation and/or exacerbation of phe- isolate of Actinobacillus pleuropneumoniae. Genome Announc
notype have been made by others when expressing hfq on plas- 2016;4:e01667–15.
mids, either from its own promoter or an inducible one (Fan- Bossé JT, Durham AL, Rycroft AN et al. New plasmid tools for

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


tappiè et al. 2009; Schilling and Gericher 2009; Bai et al. 2010; genetic analysis of Actinobacillus pleuropneumoniae and other
Chambers and Bender 2011). It is possible that, even though Pasteurellaceae. Appl Environ Microbiol 2009;75:6124–31.
we deliberately cloned the hfq gene along with three possible Bossé JT, Janson H, Sheehan BJ et al. Actinobacillus pleuropneu-
endogenous promoters, on a low copy number plasmid, the moniae: pathobiology and pathogenesis of infection. Microbes
intracellular levels of Hfq were either lower or higher than those Infect 2002;4:225–35.
present in the WT strains, resulting in differential regulation of Bossé JT, Li Y, Sárközi R et al. Proposal of serovars 17 and 18
genes affecting the different phenotypes. of Actinobacillus pleuropneumoniae based on serological and
In summary, we found that Hfq contributes to regulation of genotypic analysis. Vet Microbiol 2018;217:1–6.
adhesion to biotic and abiotic surfaces, resistance to various Bossé JT, Sinha S, Li M-S et al. Regulation of pga operon expres-
stress conditions and virulence in a surrogate model of infec- sion and biofilm formation in Actinobacillus pleuropneumoniae
tion, to differing extents in the three serovars of A. pleurop- by sigmaE and H-NS. J Bacteriol 2010;192:2414–23.
neumoniae studied. The full set of genes and sRNAs contribut- Bossé JT, Soares-Bazzolli DM, Li Y et al. The generation of suc-
ing to each of these phenotypes, and how these differ between cessive unmarked mutations and chromosomal insertion of
serovar/strains of A. pleuropneumoniae, remain to be determined. heterologous genes in Actinobacillus pleuropneumoniae using
We conclude the need for caution in extrapolating the effects natural transformation. PLoS One 2014;9:e111252.
of deletion of global regulators, and hfq in particular, to other Buettner FFR, Bendallah IM, Bossé JT et al. Analysis of the Acti-
strains of the same species, especially regarding complex phe- nobacillus pleuropneumoniae ArcA regulon identifies fumarate
notypes. reductase as a determinant of virulence. Infect Immun
2008;76:2284–95.
Buettner FFR, Bendalla IM, Bossé JT et al. Analysis of the Acti-
ACKNOWLEDGMENTS nobacillus pleuropneumoniae HlyX (FNR) regulon and identifi-
cation of iron-regulated protein B as an essential virulence
The authors thank to Acácio Rodrigues Salvador for helping with factor. Proteomics 2009;9:2383–98.
the figures. Chambers JR, Bender KS. The RNA chaperone Hfq is important
for growth and stress tolerance in Francisella novicida. PLoS
One 2011;6:e19797.
FUNDING Chiers K, De Waele T, Pasmans F et al. Virulence factors of Acti-
nobacillus pleuropneumoniae involved in colonization, persis-
The authors thank CNPq (201840/2011-1, 407849/2012-2, tence and induction of lesions in its porcine host. Vet Res
142495/2014-0 and 141328/2018), FAPEMIG (CBB-APQ-02732-15), 2010;41:65.
CAPES/PROEX (23038.019105/2016-86 and 23038.002486/2018- Cuccui J, Terra VS, Bossé JT et al. The N-linking glycosylation
26), FINEP (Núcleo de Microscopia e Microanálise—UFV), BBSRC system from Actinobacillus pleuropneumoniae is required for
(BB/K021109/1, BB/G019177/1, BB/M023052/1, BB/S020543/1, adhesion and has potential use in glycoengineering. Open Biol
BB/P001262/1, and BB/G018553) and CONFAP—the UK 2017;7:160212.
Academies (CBB- APQ-00689-16).
Cui M, Wang T, Xu J et al. Impact of Hfq on global gene expres-
Conflicts of Interest. None declared. sion and intracellular survival in Brucella melitensis. PLoS One
2013;8:e71933.
Dos Santos RF, Arraiano CM, Andrade JM. New molecular inter-
actions broaden the functions of the RNA chaperone Hfq.
REFERENCES
Curr Genet 2019;65:1313–9.
Auger E, Deslandes V, Ramjeet M et al. Host-pathogen interac- Fantappiè L, Metruccio MME, Seib KL et al. The RNA chaperone
tions of Actinobacillus pleuropneumoniae with porcine lung and Hfq is involved in stress response and virulence in Neisseria
tracheal epithelial cells. Infect Immun 2009;77:1426–41. meningitidis and is a pleiotropic regulator of protein expres-
Bai G, Golubov A, Smith EA et al. The importance of the small sion. Infect Immun 2009;77:1842–53.
RNA chaperone Hfq for growth of epidemic Yersinia pestis, but Feliciano JR, Grilo AM, Guerreiro SI et al. Hfq: a multi-
not Yersinia pseudotuberculosis, with implications for plague faceted RNA chaperone involved in virulence. Future Microbiol
biology. J Bacteriol 2010;192:4239–45. 2016;11:137–51.
Beddek AJ, Sheehan BJ, Bossé JT et al. Two TonB systems in Acti- Ferrières L, Hémery G, Nham T et al. Silent mischief: bacterio-
nobacillus pleuropneumoniae: their roles in iron acquisition and phage Mu insertions contaminate products of Escherichia coli
virulence. Infect Immun 2004;72:701–8. random mutagenesis performed using suicidal transposon
Behera P, Kutty VHM, Kumar A et al. Changing the codon usage of delivery plasmids mobilized by broad-host-range RP4 con-
hfq gene has profound effect on phenotype and pathogenic- jugative machinery. J Bacteriol 2010;192:6418–27.
ity of Salmonella Typhimurium. Curr Microbiol 2016;72:288–96. Franze de Fernandez MT, Hayward WS, August JT. Bacterial pro-
Blackall PJ, Klaasen H, Van Den Bosch H et al. Proposal of a teins required for replication of phage Q ribonucleic acid.
new serovar of Actinobacillus pleuropneumoniae: serovar 15. Vet Pruification and properties of host factor I, a ribonucleic acid-
Microbiol 2002;84:47–52. binding protein. J Biol Chem 1972;247:824–31.
12 Pathogens and Disease, 2020, Vol. 78, No. 9

Frey J. The role of RTX toxins in host specificity of animal Moen B, Røssvoll E, Måge I et al. Microbiota formed on attached
pathogenic Pasteurellaceae. Vet Microbiol 2011;153:51–58. stainless steel coupons correlates with the natural biofilm
Goel MK, Khanna P, Kishore J. Understanding survival analysis: of the sink surface in domestic kitchens. Can J Microbiol
Kaplan-Meier estimate. Int J Ayurveda Res 2010;1:274–8. 2015;62:148–60.
Hathroubi S, Hancock MA, Bossé JT et al. Surface polysaccharide Mégroz M, Kleifeld O, Wright A et al. The RNA-binding chaper-
mutants reveal that absence of O antigen reduces biofilm one Hfq is an important global regulator of gene expression
formation of Actinobacillus pleuropneumoniae. Infect Immun in Pasteurella multocida and plays a crucial role in production
2015;84:127–37. of a number of virulence factors, including hyaluronic acid

Downloaded from https://academic.oup.com/femspd/article/78/9/ftaa066/5936557 by Universidade Federal de Viçosa user on 25 November 2020


Hayes JA, Surmann K, Lamberti Y et al. Hfq modulates global pro- capsule. Infect Immun 2016;84:1361–70.
tein pattern and stress response in Bordetella pertussis. J Pro- O’Dwyer CA, Reddin K, Martin D et al. Expression of heterolo-
teomics 2020;211:1035592. gous antigens in commensal Neisseria spp.: preservation of
Hempel RJ, Morton DJ, Seale TW et al. The role of the RNA chap- conformational epitopes with vaccine potential. Infect Immun
erone Hfq in Haemophilus influenzae pathogenesis. BMC Micro- 2004;72:6511–8.
biol 2013;13:134. Paradis SÉ, Dubreuil D, Rioux S et al. High-molecular-mass
Izano EA, Sadovskaya I, Vinogradov E et al. Poly-N- lipopolysaccharides are involved in Actinobacillus pleuropneu-
acetylglucosamine mediates biofilm formation and antibi- moniae adherence to porcine respiratory tract cells. Infect
otic resistance in Actinobacillus pleuropneumoniae. Microb Immun 1994;62:3311–9.
Pathog 2007;43:1–9. Pereira MF, Rossi CC, de Queiroz MV et al. Galleria mellonella is
Kakoschke T, Kakoschke S, Magistro G et al. The RNA chaper- an effective model to study Actinobacillus pleuropneumoniae
one Hfq impacts growth, metabolism and production of viru- infection. Microbiology 2015;161:387–400.
lence factors in Yersinia enterocolitica. PLoS One 2014;9:e86113. Pereira MF, Rossi CC, Seide LE et al. Antimicrobial resistance,
Kaplan JB, Mulks MH. Biofilm formation is prevalent among biofilm formation and virulence reveal Actinobacillus pleu-
field isolates of Actinobacillus pleuropneumoniae. Vet Microbiol ropneumoniae strains’ pathogenicity complexity. Res Vet Sci
2005;108:89–94. 2018;118:498–501.
Kaplan JB, Velliyagounder K, Ragunath C et al. Genes involved Rioux S, Galarneau C, Harel J et al. Isolation and character-
in the synthesis and degradation of matrix polysaccharide ization of mini-Tn10 lipopolysaccharide mutants of Acti-
in Actinobacillus actinomycetemcomitans and Actinobacillus pleu- nobacillus pleuropneumoniae serotype 1. Can J Microbiol 1999;45:
ropneumoniae biofilms. J Bacteriol 2004;186:8213–20. 1017–26.
Kendall MM, Gruber CC, Rasko DA et al. Hfq virulence regulation Rogers RJ, Eaves LE, Blackall PJ et al. The comparative pathogenic-
in enterohemorrhagic Escherichia coli O157:H7 strain 86-24. J ity of four serovars of Actinobacillus (Haemo philus) pleuropneu-
Bacteriol 2011;193:6843–51. moniae. Aust Vet J 1990;67:9–12.
Kim S, Hwang H, Kim K-P et al. hfq plays important roles in vir- Sassu EL, Bossé JT, Tobias TJ et al. Update on Actinobacillus
ulence and stress adaptation in Cronobacter sakazakii ATCC pleuropneumoniae—knowledge, gaps and challenges. Trans-
29544. Infect Immun 2015;83:2089–98. bound Emerg Dis 2018;65:72–90.
Klitgaard K, Friis C, Jensen TK et al. Transcriptional por- Schilling D, Gerischer U. The Acinetobacter baylyi hfq gene
trait of Actinobacillus pleuropneumoniae during acute disease– encodes a large protein with an unusual C terminus. J Bac-
potential strategies for survival and persistence in the host. teriol 2009;191:5553–62.
PLoS One 2012;7:e35549. Sheehan BJ, Bossé JT, Beddek AJ et al. Identification of Acti-
Lai J, Tang D, Liang Y et al. The RNA chaperone Hfq is impor- nobacillus pleuropneumoniae genes important for survival dur-
tant for the virulence, motility and stress tolerance in the ing infection in its natural host. Infect Immun 2003;71:3960–70.
phytopathogen Xanthomonas campestris. Environ Microbiol Rep Sobrero P, Valverde C. The bacterial protein Hfq: much
2018;10:542–54. more than a mere RNA-binding factor. Crit Rev Microbiol
Li G, Xie F, Zhang Y et al. Role of (p)ppGpp in viability and 2012;38:276–99.
biofilm formation of Actinobacillus pleuropneumoniae S8. PLoS Solovyev VV, Shahmuradov IA, Salamov AA. Identification of
One 2015;10:e0141501. promoter regions and regulatory sites. Methods Mol Biol
Li H, Liu F, Peng W et al. The CpxA/CpxR two-component sys- 2010;674:57–83.
tem affects biofilm formation and virulence in Actinobacillus Subashchandrabose S, Leveque RM, Kirkwood RN et al. The
pleuropneumoniae. Front Cell Infect Microbiol 2018;8:2396. RNA chaperone Hfq promotes fitness of Actinobacillus pleu-
Li T, Xu Z, Zhang T et al. The genetic analysis of the flp locus ropneumoniae during porcine pleuropneumonia. Infect Immun
of Actinobacillus pleuropneumoniae. Arch Microbiol 2012;194: 2013;81:2952–61.
167–76. Tsui HC, Leung HC, Winkler ME. Characterization of broadly
Liu J, Cao Y, Gao L et al. Outer membrane lipoprotein Lip40 modu- pleiotropic phenotypes caused by an hfq insertion mutation
lates adherence, colonization, and virulence of Actinobacillus in Escherichia coli K-12. Mol Microbiol 1994;13:35–49.
pleuropneumoniae. Front Microbiol 2018;9:1426. Van Overbeke I, Chiers K, Charlier G et al. Characterization
Liu J, Hu L, Xu Z et al. Actinobacillus pleuropneumoniae two- of the in vitro adhesion of Actinobacillus pleuropneumoniae
component system QseB/QseC regulates the transcription of to swine alveolar epithelial cells. Vet Microbiol 2002;88:
PilM, an important determinant of bacterial adherence and 59–74.
virulence. Vet Microbiol 2015;177:184–92. Vogel J, Luisi BF. Hfq and its constellation of RNA. Nat Rev Micro-
Liu Y, Wu N, Dong J et al. Hfq is a global regulator that con- biol 2011;9:578–89.
trols the pathogenicity of Staphylococcus aureus. PLoS One Zhou L, Rycroft AN, Kroll JS et al. An Actinobacillus pleuropneu-
2010;5:e13069. moniae (APP) hfq mutant is attenuated for virulence. Proceed-
Mao F, Dam P, Chou J et al. DOOR: a database for prokaryotic ings of the International Pasteurellaceae Society, Sorrento, Italy
operons. Nucleic Acids Res 2009;37:D459–63. 2008.

You might also like