ArticlePDF Available

Enhanced expression of codon optimized Mycobacterium avium subsp. paratuberculosis antigens in Lactobacillus salivarius

Frontiers
Frontiers in Cellular and Infection Microbiology
Authors:

Abstract and Figures

It is well documented that open reading frames containing high GC content show poor expression in A+T rich hosts. Specifically, G+C-rich codon usage is a limiting factor in heterologous expression of Mycobacterium avium subsp. paratuberculosis (MAP) proteins using Lactobacillus salivarius. However, re-engineering opening reading frames through synonymous substitutions can offset codon bias and greatly enhance MAP protein production in this host. In this report, we demonstrate that codon-usage manipulation of MAP2121c can enhance the heterologous expression of the major membrane protein (MMP), analogous to the form in which it is produced natively by MAP bacilli. When heterologously over-expressed, antigenic determinants were preserved in synthetic MMP proteins as shown by monoclonal antibody mediated ELISA. Moreover, MMP is a membrane protein in MAP, which is also targeted to the cellular surface of recombinant L. salivarius at levels comparable to MAP. Additionally, we previously engineered MAP3733c (encoding MptD) and show herein that MptD displays the tendency to associate with the cytoplasmic membrane boundary under confocal microscopy and the intracellularly accumulated protein selectively adheres to the MptD-specific bacteriophage fMptD. This work demonstrates there is potential for L. salivarius as a viable antigen delivery vehicle for MAP, which may provide an effective mucosal vaccine against Johne's disease.
Content may be subject to copyright.
ORIGINAL RESEARCH ARTICLE
published: 04 September 2014
doi: 10.3389/fcimb.2014.00120
Enhanced expression of codon optimized Mycobacterium
avium subsp. paratuberculosis antigens in Lactobacillus
salivarius
Christopher D. Johnston1, John P. Bannantine2, Rodney Govender1, Lorraine Endersen1,
Daniel Pletzer 3, Helge Weingart 3, Aidan Coffey1, Jim O’Mahony 1*and Roy D. Sleator1
1Biological Sciences Department, Cork Institute of Technology, Cork, Ireland
2United States Department of Agriculture - Agricultural Research Service, National Animal Disease Center, Ames, IA, USA
3School of Engineering and Science, Jacobs University Bremen, Bremen, Germany
Edited by:
Adel M. Talaat, University of
Wisconsin–Madison, USA
Reviewed by:
Martin S. Pavelka, University of
Rochester, USA
Jan Peter Van Pijkeren, University of
Wisconsin–Madison, USA
*Correspondence:
Jim O’Mahony, Department of
Biological Sciences, Cork Institute of
Technology, Rossa Avenue,
Bishopstown, Cork, Ireland
e-mail: jim.omahony@cit.ie
It is well documented that open reading frames containing high GC content show poor
expression in A+T rich hosts. Specifically, G+C-rich codon usage is a limiting factor in
heterologous expression of Mycobacterium avium subsp. paratuberculosis (MAP) proteins
using Lactobacillus salivarius. However, re-engineering opening reading frames through
synonymous substitutions can offset codon bias and greatly enhance MAP protein
production in this host. In this report, we demonstrate that codon-usage manipulation
of MAP2121c can enhance the heterologous expression of the major membrane protein
(MMP), analogous to the form in which it is produced natively by MAP bacilli. When
heterologously over-expressed, antigenic determinants were preserved in synthetic MMP
proteins as shown by monoclonal antibody mediated ELISA. Moreover, MMP is a
membrane protein in MAP, which is also targeted to the cellular surface of recombinant
L. salivarius at levels comparable to MAP. Additionally, we previously engineered
MAP3733c (encoding MptD) and show herein that MptD displays the tendency to
associate with the cytoplasmic membrane boundary under confocal microscopy and the
intracellularly accumulated protein selectively adheres to the MptD-specific bacteriophage
fMptD. This work demonstrates there is potential for L. salivarius as a viable antigen
delivery vehicle for MAP, which may provide an effective mucosal vaccine against Johne’s
disease.
Keywords: MAP antigens, MptD, MMP, codon optimization, expression host, paratuberculosis, MAP vaccine,
Johne’s disease
INTRODUCTION
Mycobacterium avium subsp. paratuberculosis (MAP) is an
intracellular pathogen and the etiological agent of Johne’s dis-
ease, a chronic inflammatory disorder of the gastrointestinal tract
which affects multiple ruminant species including cattle (Chacon
et al., 2004). Live attenuated vaccine formulations against Johne’s
disease do not appear to offer adequate protection against MAP
infection in goats (Hines et al., 2014) and while heat-killed whole
cell vaccines that are commercially available do show some effi-
cacy, these also fail to provide full protection against MAP in
models of infection (Rosseels and Huygen, 2008). Moreover,
issues relating to interference with diagnostic assays for bovine
tuberculosis have further hindered their widespread development
and application (Buddle et al., 2010; Bastida and Juste, 2011;
Coad et al., 2013). These limitations combined with the avail-
ability of complete genome sequences for MAP (Li et al., 2005;
Bannantine et al., 2014) have focused recent attention on experi-
mental subunit based vaccine strategies against MAP (Bull et al.,
2007; Rosseels and Huygen, 2008). One particularly promising
subunit vaccine for MAP is the 70 kDa heat shock protein termed
Hsp70 (Koets et al., 2006), which activates B cells (Vrie lin g et al.,
2013).
Lactobacillus are interesting candidates for the development of
novel oral vaccine vectors due to certain strains widespread use
in the food industry and GRAS (generally regarded as safe) status
(Yu et al., 2013). Specific members of this genus are also an attrac-
tive alternative to using attenuated pathogens for mucosal deliv-
ery strategies because they can survive the upper gastrointestinal
tract (GI) and colonize the lower GI tract (Bermudez-Humaran
et al., 2011). Numerous studies have substantiated the potential
of specific Lactobacilli strains to serve as live vaccine delivery vehi-
cles against a broad spectrum of mucosal pathogens including
Bacillus anthracis,Streptococcus pneumonia, Clostridium difficile
and the Avian Influenza Virus H5N1 (Campos et al., 2008;
Sleator and Hill, 2008; Mohamadzadeh et al., 2010; Wang et al.,
2012). MAP is particularly well-suited to Lactobacillus vector
delivery because it is a pathogen transmitted by ingestion of
contaminated feces or milk and passes through the GI tract
where it infects the intestinal mucosa (Bannantine and Bermudez,
2013).
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |1
CELLULA R AND INFECTION MICROBIOLOG
Y
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
However, despite the obvious advantages of Lactobacillus based
mucosal immunization; encompassing the inherent ability of
specific strains to survive gastric transit, adhere to the intesti-
nal epithelium (Messaoudi et al., 2012), and stimulate both
mucosal and systemic immune responses (Mohamadzadeh et al.,
2009), there are known difficulties in expression of G+Crich
coding sequences in the A+TrichLactobacillus host (Johnston
et al., 2013). Indeed, we recently determined that the significantly
divergent genomic G+C content of MAP and Lactobacillus sali-
varius (69 and 33% respectively) leads to a disparity in codon
usage, which significantly impedes recombinant MAP protein
synthesis. To alleviate this translation inefficiency likely due to
ribosomal pausing at rare codons (Buchan and Stansfield, 2007),
codon optimization of a MAP gene (MAP3733c) was performed
by introducing a series of synonymous mutations; modifying
the coding region to better suit the codon bias of L. salivar-
ius. It was shown that synthesis of a MAP specific membrane
protein within L. salivarius could be markedly enhanced (>37-
fold) owing to codon optimization, resulting in the abundance
of MAP-GFP protein fusion fluorescence in recombinant cells
(Johnston et al., 2013). However, that protein was never con-
firmed to truly represent the native protein as no monoclonal
antibodies or other specific detection reagents were developed
against it.
Because MAP surface proteins likely play the dominant role
in the initial interactions with bovine intestinal cells (Bannantine
et al., 2003; He and De Buck, 2010; Gurung et al., 2012), we
here focused on two important MAP membrane proteins, MMP
(MAP2121c) and the previously studied MptD (MAP3733c).
MMP, encoded by MAP2121c, is a 33.5 kDa surface protein
which shares homology to a Mycobacterium leprae 35-kDa major
membrane protein-1 (MMP-1) (Winter et al., 1995), previ-
ously identified as a potent immunodominant antigen capable
of inducing T-lymphocyte responses in paucibacillary leprosy
patients (Triccas et al., 1996). MptD is a MAP specific, virulence
associated membrane protein which is surface expressed during
natural infection, warranting its further investigation as a pro-
phylactic antigen (Stratmann et al., 2004; Shin et al., 2006; Cossu
et al., 2011).
A significant limitation of current whole cell MAP vaccine
strategies is interference with cellular immune assays and tuber-
culin skin tests for bovine tuberculosis (M. bovis), which restricts
their widespread application in many countries (Kohler e t al .,
2001; Scandurra et al., 2010). It is notable that although MMP
is not specific to MAP, with homologs existing in other mycobac-
terial species including M. avium (MAC) and M. leprae, previous
genetic and serological evidence suggests that the protein is absent
from M. tuberculosis and M. bovis (Triccas et al., 1998; Banasure
et al., 2001). In addition, a DNA vaccine incorporating the
M. leprae MMP-1 protein in isolation demonstrated significant
levels of protective efficacy against M. leprae footpad infection in
outbred Swiss Albino mice (Martin et al., 2001). As such, if an
MMP-based vaccine against Johne’s disease were to display strong
prophylactic efficacy, as it has done against Leprosy, it leads to
the exciting prospect of a vaccine formulation that could over-
come issues with interference of bovine tuberculosis screening
assays.
Herein, we analyzed MAP proteins recoded using synony-
mous substitutions and expressed in L. salivarius. Faithful
expression and antigenicity was examined using a combina-
tion of fluorescence microscopy, monoclonal antibody- and
bacteriophage-based ELISA.
MATERIALS AND METHODS
STRAINS, PLASMIDS, BACTERIOPHAGE, AND GROWTH CONDITIONS
The strains and plasmids used in this study are listed in
Table 1.Mycobacterium avium subsp. paratuberculosis strain K-
10 (ATCC BAA968) were propagated in Middlebrook 7H9
broth supplemented with OADC (BD Biosciences), 2μg/ml
of Mycobactin J and 0.2% glycerol and incubated at 37C
for 6–10 weeks. Escherichia coli DH5αelectrocompetent cells
(Invitrogen) were used as intermediate cloning hosts for all
vector constructs within this study and grown at 37Cin
Luria-Bertani (LB) media. Lactobacillus salivarius NRRL B-
30514 (kindly donated by Dr. Norman Stern, USDA), was used
throughout this study as the host for individual vector con-
structs and was grown aerobically at 37CinMRSmedia
(Fluka).
Plasmids pNZ9530 and pNZ8048 were originally obtained
from the University College Cork culture collection, while
pUC57 vectors containing codon-optimized synthetic GFP and
MptDsynth genes were obtained from the Cork Institute of
Technology culture collection (Johnston et al., 2013). Cultures
of E. coli harboring individual vectors were supplemented
with Erythromycin (Ery, 200 μg/ml), Chloramphenicol (Cm,
12.5 μg/ml), or Ampicillin (Amp, 200 μg/ml) for pNZ9530,
pNZ8048, and pUC57 containing cells respectively. Recombinant
L. salivarius cells were subcultured from 40% v/v glycerol
stocks at 20C and grown using antibiotic selection. L. sali-
varius (pNZ9530) cultures were supplemented with 5 μg/ml
Ery, while L. salivarius harboring both pNZ9530 and pNZ8048
constructsweregrowninthepresenceof5μg/ml Ery and
10 μg/ml Cm.
The M13 phage fMptD, originally isolated from the Ph.D.12
phage display library (New England Biolabs), was obtained from
the laboratory of Gerald F. Gerlach (University of Veterinary
Medicine, Hanover, Germany). Bacteriophage fMptD was prop-
agated using standard methods previously described (Chappel
et al., 1998). Phage titers were assessed by a standard plaque assay
test (Sambrook et al., 1990). Purified high titer phage solutions
(1010 pfu/ml) were stored at 4C until required.
NUCLEIC ACID ISOLATION
Mycobacterial DNA was prepared as described previously by
Douarre et al. (2010). Plasmid DNA was isolated from E. coli
DH5αusing a plasmid extraction kit as per the manufac-
turer’s instructions (Sigma Aldrich). Plasmid isolation from
Lactobacillus strains was also carried out using this kit, after ini-
tial incubation (30 min at 37C) in protoplast buffer (20 mM
Tris-HCl, 5 mM EDTA, 0.75 M Sucrose, 10 mg/ml lyzozyme
and 50 U/ml mutanolysin pH 7.5). Quantitative analysis was
carried out using microspectrophotometry (Nanodrop, De,
USA) and plasmid DNA concentration was normalized to
250 ng/μl.
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |2
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
Table 1 | Strains, Phage, and Plasmids used in this study.
Plasmid, phage, and strains Relevant genotype or characteristics Source or References
PLASMIDS
pNZ9530 N.I.C.E system helper plasmid. Eryr,nisRK cloned in pIL252, expression
of nisRK driven by rep read through, low copy number
Pavan et al., 2000
pNZ8048 N.I.C.E system expression plasmid. Cmr, carries the nisin-inducible
promoter PnisA
Pavan et al., 2000
pNZ:MAP3733c pNZ8048 with MptD fused to GFP gene under control of PnisA promoter This work
pNZ:MAP3733synth pNZ8048 with codon optimized MptDsynth gene under control of PnisA
promoter
This work
pNZ:MAP3733c-GFP pNZ8048 with MptD fused to GFP gene under control of PnisA promoter Johnston et al., 2013
pNZ:MAP3733synth-GFP pNZ8048 with codon optimized MptDsynth fused to GFP gene under
control of PnisA promoter
Johnston et al., 2013
pNZ:MAP2121c pNZ8048 with MMP fused to GFP gene under control of PnisA promoter This work
pNZ:MAP2121synth pNZ8048 with codon optimized MMPsynth gene under control of PnisA
promoter
This work
pNZ:MAP2121c-GFP pNZ8048 with MMP fused to GFP gene under control of PnisA promoter This work
pNZ:MAP2121synth-GFP pNZ8048 with codon optimized MMPsynth fused to GFP gene under
control of PnisA promoter
This work
pNZ:GFP pNZ8048 with L. salivarius codon optimized GFP gene under control of
PnisA promoter
Johnston et al., 2013
PHAGE
Phage fMptD Phage isolated from the Ph.D.-12 phage display library, with a
dodecapeptide sequence (GKNHHHQHHRPQ) fused to the N-terminus
of its minor coat protein (pIII)
Stratmann et al., 2004
STRAINS
Mycobacterium avium subsp. paratuberculosis
MAP K-10 American bovine virulent isolate and sequencing project reference strain Li et al., 2005
Escherichia coli
DH5αIntermediate cloning host Invitrogen
Lactobacillus salivarius
NRRL B-30514 Host strain, originally isolated from cecal contents of broiler chicken.
Aerobic
Stern et al., 2006
L. salivarius pNZ9530 Host strain harboring pNZ9530 helper plasmid, EryrJohnston et al., 2013
L. salivarius pNZ8048 Harbors pNZ9530 plasmid and pNZ8048 expression plasmid lacking
insert. Eryrand Cmr
Johnston et al., 2013
L. salivarius MAP3733c Harbors pNZ9530 and pNZ:MAP3733c plasmids. Eryrand CmrThis work
L. salivarius MAP3733synth Harbors pNZ9530 and pNZ:MAP3733synth plasmids. Eryrand CmrThis work
L. salivarius MAP3733c-GFP Harbors pNZ9530 and pNZ:MAP3733c-GFP plasmids. Eryrand CmrJohnston et al., 2013
L. salivarius MAP3733synth-GFP Harbors pNZ9530 and pNZ:MAP3733synth-GFP plasmids. Eryrand CmrJohnston et al., 2013
L. salivarius MAP2121c Harbors pNZ9530 and pNZ:MAP2121c plasmids. Eryrand CmrThis work
L. salivarius MAP2121synth Harbors pNZ9530 and pNZ:MAP2121synth plasmids. Eryrand CmrThis work
L. salivarius MAP2121c-GFP Harbors pNZ9530 and pNZ:MAP2121c-GFP plasmids. Eryrand CmrThis work
L. salivarius MAP2121synth-GFP Harbors pNZ9530 and pNZ:MAP2121synth-GFP plasmids. Eryrand CmrThis work
L. salivarius GFP Harbors pNZ9530 and pNZ:GFP plasmids. Eryrand CmrJohnston et al., 2013
CODON OPTIMIZATION OF MAP2121c
MAP2121c encodes a major membrane protein (MMP) in MAP.
Because this protein has been implicated in the early events
of infection in the bovine intestinal musoca, it is an ideal
candidate for testing expression in L. salivarius as a poten-
tial mucosal vaccine. We have previously described a strategy
for the codon optimization of the MAP3733c gene for expres-
sion in L. salivarius (Johnston et al., 2013). Here we used the
same strategy for MAP2121c; briefly, a bioinformatics analysis
was performed to identify codons from MAP2121c that could
be modified at the third base position without a change in
the resulting amino acid (termed a synonymous substitution).
Coding sequences were synthesized by GenScript USA Inc.
(Piscataway, NJ). Constructs were cloned as described below and
confirmed by DNA sequencing. Final sequences for each gene
are available from GenBank (Accession numbers: KC854397 and
KC517484). All modifications to MAP2121c are summarized in
Table 2.
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |3
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
Table 2 | Modification and codon optimization of MAP genes.
Gene Length G+C Unfavorable Modified Source
(bp) content codons*codons
MAP2121c 924 66.5% 276/307 0/307 This work
MAP2121synth 924 32.8% 7/307 279/307 This work
*Codons were deemed unfavorable due to the presence of a guanine or cytosine
within the 3rd base of the triplet.
PCR AMPLIFICATIONS AND MODIFICATIONS
PCR primers are listed in Supplementary Table S1. Primers
were designed for the native and codon optimized MAP genes
(MAP2121c, MAP3733c, MAP2121synth, and MAP3733synth)
based on either MAP strain K-10 sequence data available from
the NCBI database (NC_002944) or using the sequence from
GenScript synthesized genes. All conventional PCR reactions
were carried out using high fidelity Velocity DNA polymerase
Kit (Bioline) in accordance with the manufacturer’s instructions.
Restriction enzymes and T4 DNA ligase were purchased from
Roche Diagnostics (Mannheim, Germany) and New England
Biolabs (Beverly, MA, USA) and used as per manufacturer’s rec-
ommendations. Ligation reaction mixtures were purified using
the High Pure PCR product purification kit (Roche).
MAP GENE AND FUSION CONSTRUCTS
Individual MAP gene constructs (native and synthetic) were
cloned into the E. coli-L. lactis shuttle vector pNZ8048, a deriva-
tive of pNZ124 that allows expression of proteins under the
control of the nisin-inducible promoter PnisA,partofthe
NIsin Controlled Expression (NICE) system (Pavan et al., 2000).
Nisin induced expression was achieved in L. salivarius via co-
transformation of a dual plasmid system; pNZ8048 with insert
to be expressed downstream of PnisA and pNZ9530 providing the
necessary nisRK regulatory genes in trans (de Ruyter et al., 1996).
Two native MAP genes, MAP2121c and MAP3733c (des-
ignated “c” to indicate that each is originally transcribed on
the complimentary strand of the MAP K-10 genome) and two
synthetic codon optimized counterparts of these, designated
MAP2121synth and MAP3733synth, were initially cloned form-
ing pNZ:2121c, pNZ:3733c, pNZ:2121synth, and pNZ:3733synth.
To facilitate fluorometric analysis of each of these genes during
expression and monitor subcellular localization of proteins within
the host cell, a C-terminus GFP gene was translationally fused
to each gene construct forming pNZ:2121c-GFP, pNZ:3733c-
GFP, pNZ:2121synth-GFP, and pNZ:3733synth-GFP. The fusion
of GFP and individual MAP genes was performed using Splicing
by Overlap Extension (SOEing) as previously described (Johnston
et al., 2013).
The GFP coding region used for fusions throughout this
study was codon optimized for use within L. salivarius. The GFP
gene sequence cloned into pNZ8048 (Johnston et al., 2013)was
used to provide a comparative control for subsequent assays and
designated pNZ:GFP.
TRANSFORMATION AND INDUCTION OF L. SALIVARIUS CONSTRUCTS
Competent L. salivarius (pNZ9530) were transformed
individually with each of the MAP gene constructs (Ta b l e 1 )as
described previously (Johnston et al., 2013). Overnight cultures
of recombinant L. salivarius strains were subcultured (1:100
dilution) into fresh MRS broth (Cm 8 μg/ml, Ery 3.5 μg/ml) and
incubated with agitation (100 rpm) at 37C. At an optical density
(OD600nm) of 0.35, nisin was added at a final concentration
of 10 ng/ml and cultures were incubated statically at 37C
for 2 h. 10 ml aliquots of each culture were then harvested by
centrifugation (6000 rpm for 5min) for subsequent analysis.
FLUORESCENCE MICROSCOPY
To facilitate visualization of cells using fluorescence microscopy,
induced L. salivarius strains (L.sal GFP, L.sal MAP2121c-
GFP, L.sal MAP2121synth-GFP, L.sal MAP3733c-GFP, L.sal
MAP3733synth-GFP) were fixed using 3.7% formaldehyde,
washed with PBS (pH 7.2), subsequently resuspended in 1 ml
PBS and stored at 4C until visualized. For detection of GFP
fusion peptides, L. salivarius cell images were taken using a Zeiss
LSM 510 META laser-scanning microscope equipped with Argon
and Helium-Neon lasers (Carl Zeiss, Oberkochen, Germany) at
a resolution of 2048 ×2048 pixels, using LSM 5 software (ver-
sion 3.2; Carl Zeiss). Equal settings were used for detection
of green fluorescence among different strains (Amplifier Offset:
0.05, Amplifier Gain: 1, Gain: 820).
PREPARATION OF WHOLE CELL L. SALIVARIUS FOR ELISA
To determine if recombinant MMP and MMPsynth peptides
were displayed on the surface of whole cell L. salivarius, nisin-
induced strains were harvested, washed and resuspended in PBS
and coated directly to the wells of a Nunc Maxisorp plate (1 ×
109cells/ml). To determine if these peptides were accumulating
within the cytoplasm of L. salivarius, 500 μl of the harvested cells
were transferred to 2 ml screw cap tubes with 0.3 g glass beads
(Sigma, 150–212 μm, acid washed) and lysed (4000 rpm for 45 s)
using a MagNA Lyser Instrument (Roche). Subsequently, 100 μl
aliquots of crude cell lysate were added to individual wells of the
same Maxisorp plate.
In the analysis of MptD proteins, similar techniques were
applied for whole-cell and cell-lysate preparations, however,
use of the alternative bacteriophage (fMptD) detection method
necessitated the substitution of PBS for TBS in all washing and
subsequent steps. MAP strain K-10, processed in the same man-
ner, was included in all assays to provide a comparative control.
MONOCLONAL ANTIBODY BASED ELISA OF MMP
Maxisorp plates containing recombinant L. salivarius (pNZ,
MMP, MMPsynth, MMP-GFP, and MMPsynth-GFP), as well
as MAP K-10, whole cells and cell lysate were incubated at
37C for 1 h and then blocked with a 5% (w/v) solution of
dry skimmed milk powder in PBS. A 100-μl aliquot of puri-
fied monoclonal antibody 13E1 or 8G2, appropriately diluted
in PBS plus 0.1% tween 20 (PBS/T) was added to each test
well. Samples were incubated at 37C for 1 h on a rocking plat-
form. The wells were washed and 100 μl of secondary antibody
(Peroxidase-labeled, Anti-Mouse IgG detection antibody) diluted
in PBS/T containing 1% (w/v) milk powder was added. After a 1 h
incubation at 37C the wells were washed and 100 μlof3,3
,5,5-
Tetramethylbenzidine Liquid Substrate System for ELISA (Sigma)
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |4
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
was added per well. The substrate was left to develop in the dark
at room temperature for 30 min after which the reaction was
stopped by addition of 50 μl of a 10% HCl solution. Absorbance
readings were read at 450 nm using a microplate reader. The bind-
ing response values against each respective recombinant cell type
were normalized by dividing the absorbance level obtained in
test wells by that obtained in parallel control wells treated with
diluent buffer without the addition of the primary monoclonal
antibody.
BACTERIOPHAGE-fMptD BASED ELISA OF MptD
Due to a lack of monoclonal antibodies available for the MptD
protein, the fMptD bacteriophage (Stratmann et al., 2006)was
used in lieu of the primary binding antibody within MptD
ELISA. Maxisorp plates containing recombinant L. salivarius
(pNZ, MptD, MptDsynth, MptD –GFP, and MptDsynth-GFP),
as well as MAP K-10, whole cells and cell lysate were incubated
at 37C for 1 h and then blocked with a 5% (w/v) solution
of dry skimmed milk powder in TBS. Plates were washed with
TBS/T (TBS +0.1% [v/v] Tween-20) and 100 μloffMptD
(109pfu/ml) diluted in TBS/T was added to each test well after
which samples were incubated at 37C for 1 h on a rocking
platform. Individual wells were subsequently washed and 100 μl
of the detection antibody, HRP Conjugated anti-M13 mon-
oclonal antibody (GE Healthcare), diluted in TBS/T contain-
ing 1% (w/v) milk powder was added. After 1 h incubation at
37C the wells were washed, 100 μl of TMB Liquid Substrate
(Sigma) was added per well and color developed as already
described.
STATISTICAL ANALYSIS
Statistical analysis was carried out using GraphPad Prism (ver-
sion 4.03; GraphPad Software, San Diego, CA). Means with
standard error (s.e.m.) are presented in each graph. Differences
between two groups were calculated using unpaired Student’s
t-test. Differences were considered significant at P<0.05.
RESULTS
DISTINCT LOCALIZATION OF MAP-GFP FUSION PEPTIDES
In our previous work we confirmed poor fluorescence for the
native MptD-GFP fusion protein expressed in L. salivarius,and
showed markedly improved expression through codon opti-
mization of the synthetic gene (MptDsynth-GFP; Figures 1E,F)
(Johnston et al., 2013). Here, consistent with our previous find-
ings for MptD, induction and confocal microscopy of the native
MMP-GFP fusion resulted in no fluorescence (Figure 1C) simi-
lar to the L. salivarius wild type control (Figure 1A). In contrast,
improved levels of fluorescence were noted for codon optimized
MMPsynth-GFP after induction under identical conditions as the
native variant (Figure 1D).
While an even distribution of strong fluorescence was
observed within pNZ:GFP-containing bacilli (Figure 1B), both
engineered MAP fusion displayed different fluorescence pat-
terns when expressed within L. salivarius (Figures 1D,F).
MMPsynth-GFP was aggregated (Figure 1D) whereas MptD was
more uniformly distributed around the periphery of the cells
(Figure 1F).
FIGURE 1 | Representative phase contrast (upper panel) and
fluorescent microscopy (lower panel) images of recombinant
L. salivarius cells. (A) Control cells pNZ:8048. (B) L. salivarius GFP cells
displayed fluorescence observed throughout individual bacilli. (C) Negligible
levels of fusion fluorescence were observed from MMP-GFP cells,
(D) while fluorescent foci, suggestive of aggregation, were observed
toward the polar regions of individual cells for the codon optimized variant
MMPsynth-GFP. (E) Low levels of fluorescence prevented determination of
the subcellular localization of native MptD-GFP fusions within L. salivarius
cells, while (F) codon optimized MptDsynth-GFP fusion proteins
demonstrated the tendency to localize toward the cellular membrane
periphery. All culture assays were performed in triplicate, multiple images
were taken from each sample and representative pictures were chosen.
Bars represent 5 μm.
MptD-GFP (MAP3733c/synth)
In silico TMHMM analysis of MptD suggests the presence of six
transmembrane segments (TMSs) within the 208 amino acids
and a large external loop at positions 147–175, with both N and
C termini being cytoplasmic associated. In cases where trans-
membrane proteins have their native C-terminus located in the
cytoplasm, fusion of a GFP tag is particularly useful for anal-
ysis of protein localization. If the fusion is expressed at the
cytoplasmic membrane the GFP peptide may fold correctly and
fluoresce allowing visualization of protein localized at this mem-
brane, however if it aggregates and forms inclusion bodies the
downstream GFP might not fold correctly and therefore not flu-
oresce (Drew et al., 2005), although this will be largely protein
dependant.
The MptDsynth-GFP fusion peptides demonstrated a ten-
dency to localize at the periphery of individual cells (Figure 1F),
suggestive of membrane domain insertion and conformation.
This result extends our initial findings with MptD (Johnston et al.,
2013).
MMP-GFP (MAP2121c/synth)
Confocal microscopy of L. salivarius MMP-GFP resulted in
undetectable fluorescence from individual bacilli (Figure 1C),
further supporting our previous hypothesis that native MAP
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |5
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
genes are poorly translated within L. salivarius.However,flu-
orescent foci were observed within the re-engineered sequence
(MMPsynth-GFP) suggesting the presence of aggregated GFP
fusion proteins within the cytoplasm of L. salivarius (Figure 1D).
This result was not expected since MMP was predicted to be more
soluble than MptD using SOLpro1.0 software (Magnan et al.,
2009).
RECOMBINANT MptD ACCUMULATES WITHIN THE CYTOPLASM OF
L. SALIVARIUS
To investigate whether the L. salivarius expressed MptD pro-
tein was analogous to the native MAP protein, we utilized a
modified version of the fMptD bacteriophage mediated ELISA
protocol outlined by Rosu et al. (2009), to probe both whole-
cellandcell-lysatesofrecombinantL. salivarius transformed with
both MptD-GFP gene constructs. Two constructs lacking the GFP
fusion were also included to control for potential interference
from the tag (Figures 2A,B).
Investigation of the crude cell lysates (Figure 2A)fromrecom-
binant L. salivarius revealed MptD phage binding to both native
and codon optimized MptD constructs lacking a GFP fusion
tag with a 2.2-fold increase in signal observed for the codon
optimized variant over native (P=0.0008), and 2-fold when
compared to MAP K-10 (P=0.0007). Interestingly, in a manner
similar to MMP analysis, the addition of a GFP fusion to each
of these constructs appears to have effectively inhibited phage
binding, possibly by masking the MptD epitope with GFP or
prevention of a conformational structure from forming.
Based on the results from confocal microscopy (Figure 1F),
we also examined intact cells (Figure 2B) to determine if the
tendency of MptD to localize toward the periphery of L. salivarius
represented true surface exposure. However, despite abundant
concentrations of MptD detected within the cytoplasm, no sig-
nal (P=<0.05 vs. control) could be detected from unbro-
ken cells expressing either native or re-engineered MptD in
the presence or absence of a GFP tag. MptD was detected
at similar levels in MAP regardless of the cell preparation
(Figures 2A,B).
RE-ENGINEERED MMP DISPLAYS TWO DISTINCT EPITOPES
The aggregation observed during confocal microscopy of
MMPsynth-GFP prompted further investigation of MMP fold-
ing within the L. salivarius host. To examine the possibility
that the C-terminus GFP tag was itself associating with the
MMP peptide, potentially leading to the fluorescent aggre-
gates observed (Figure 1D): two additional constructs, L. sali-
varius with native and codon optimized MMP genes lacking
a GFP fusion (pNZ:MAP2121c and pNZ:MAP2121synth), were
included in the assays.
Antigenic determinants or epitopes which are recognized on a
target protein by an antibody can exist in multiple forms rang-
ing from linear, present on both native or misfolded peptides, to
discontinuous or conformationally complex epitopes which are
displayed through the native folding of a protein (Brown et al.,
2011). In this study, ELISA analysis was performed using two
monoclonal antibodies specific to the MMP protein (8G2 and
13E1), which detected two distinct epitopes (Bannantine et al.,
2007).
ELISA analysis with mAb 8G2 (linear epitope)
The 8G2 antibody is reported to associate with a linear epitope
present within a 77-amino acid sequence near the N-terminus
FIGURE 2 | Comparative fMptD-mediated ELISA analysis of
recombinant L. salivarius cell lysate and whole-cell against MAP
K-10 (A,B). Standard deviation of triplicate results is indicated by error
bars. Statistically significant difference was observed at P<0.05.
Horizontal dashed lines indicate the negative control threshold for each
assay.
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |6
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
of MMP (Bannantine et al., 2007). ELISA analysis with 8G2 on
crude cell lysates of recombinant L. salivarius-MMP constructs
revealed that the linear epitope could be detected in all
MMP constructs (Figure 3A), with a significant 6.4-fold sig-
nal increase noted for the engineered MMPsynth compared to
native MMP.
While microscopy data indicate that heterologously expressed
MMP largely accumulates in the cytosol (Figure 1D), intrigu-
ingly, the engineered MMP was also detected during whole-cell
ELISA using the 8G2 antibody (Figure 3B). Although these levels
were low, the signal detected was approximate to that of the MAP
K-10 control strain (Figure 3B). Notably, as MMP expression is
enhanced in L. salivarius, more protein is localized on the surface
(Figure 3B).
ELISA analysis with mAb 13E1
Analysis of L. salivarius cellular lysates with 13E1 indicated that
the epitope recognized by this mAb is blocked when MMP
is fused to GFP fusions (Figure 3C). However, this epitope
appears to be restored by removal of the GFP tag, with 4-
fold higher levels of MMP detected from codon optimized
MMPsynth when compared to the native gene (P=0.0009),
and 1.5-fold when compared to the MAP K-10 control strain
(P=0.003).
However, most intriguingly is that MMP protein could also be
detected from whole cell L. salivarius MMPsynth using the 13E1
antibody, which was comparable to that in MAP K-10, albeit both
were present at low levels (Figure 3D). While it has previously
been indicated that MMP contains a 30 amino acid hydrophobic
FIGURE 3 | Comparative ELISA analysis of recombinant L. salivarius
cell-lysate and whole-cell against MAP K-10, using two monoclonal
antibodies directed against MMP. The 8G2 antibody detects MMP (A,B).
The mAb13E1 antibody detects a discontinuous epitope on MMP (C,D).The
average val ue of L. sal MMP-GFP and L. sal MMP are below the negative
control threshold. Standard deviation of triplicate results is indicated by error
bars. Statistically significant difference was observed at P<0.05. Horizontal
dashed lines indicate the negative control threshold for each assay.
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |7
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
domain near the C-terminal end suggestive of a membrane pro-
tein, the protein lacks a discernible N-terminal signal sequence
(Bannantine et al., 2003). Moreover, it has already been exper-
imentally demonstrated that MMP is both surface exposed and
actively shed from live MAP bacilli (Yakes et al., 2008), suggest-
ing that MMP is translocated in a non-classical manner similar
to other mycobacterial proteins (Pallen, 2002; Bendtsen et al.,
2005).
DISCUSSION
There have been several efforts at heterologous expression of
MAP proteins. E. coli expression of an impressive collection of
over 650 MAP proteins has been constructed (Bannantine et al.,
2010) and these proteins have been incorporated into a protein
array to monitor antibody response in different disease stages
(Bannantine et al., 2008).Effortshavealsobeendevotedtoin vitro
transcription-translation (Li et al., 2007), but those efforts did not
yield much protein to work with. Interestingly MMP was a test
proteininthatstudyaswell.Morerecently,MAPproteinshave
been expressed in a Salmonella vaccine delivery strategy (Faisal
et al., 2013). Our group is the first to use the probiotic genus
Lactobacillus as a vaccine vector for MAP. We have overcome the
limitations of low expression yields through re-engineering GC-
richcodingsequences.NoeffortshavebeenmadetoexpressMAP
proteins in a faster growing species of Mycobacterium such as
M. smegmatis.
We show that MptD, which contains six transmembrane
domains, was targeted to the cell periphery, likely the cytoplas-
mic membrane, while MMP formed aggregates in the cytoplasm
of L. salivarius. In general, proteins which are located within
the cytoplasmic membrane must be targeted to a translocation
site prior to their insertion and/or translocation (Fekkes and
Driessen, 1999). However, in the case of MptD, such integral
membrane proteins generally do not contain a signal sequence.
Instead, their hydrophobic TMSs function as an internal signal
for targeting and insertion which needs to be recognized early
in the translocation pathway to prevent their aggregation in the
cytoplasm (Fekkes and Driessen, 1999).
It is known that addition of any fusion tag to the N- or C-
termini of a peptide can modify specific structural characteris-
tics either by sterically interfering with protein interactions or
disrupting conformational folding (Snapp, 2009). The lack of
13E1 antibody recognition formed by MMP-GFP fusion led us to
propose that the fluorescent foci within L. salivarius MMPsynth-
GFP were related to a masking effect of the MMP epitope with
GFP and not representative of translational errors brought about
through codon optimization. The GFP tag does not appear to
impede localization of MMP at the cell surface, suggesting that
this effect is steric at the C-terminus only and does not provide
clues on secondary folding that might be required for localiza-
tion. In originally isolating the 13E1 antibody, Bannantine and
coworkers immunized mice with an N-terminal maltose-binding
protein fusion with MMP (Bannantine et al., 2007). This suggests
that the addition of a large (40 kDa) protein tag at the N- termi-
nus does not structurally impede the epitope of MMP. However,
in our study GFP was at the C-terminus and while it is possi-
ble that the GFP tag might mask the 13E1 epitope irrespective
of its fused location, it is also possible that the C-terminus is
more heavily involved than the N-terminal region in producing
theepitope.Insupportofthis,Li et al. (2007) expressed the MMP
protein with a six-histidine tag at the C-terminus, noting that the
recombinant antigen could not be detected by MAP antibodies in
pooled positive serum samples from cattle shedding MAP bacilli
(Li et al., 2007).
Detailed studies have demonstrated that MMP is a surface-
located virulence factor involved in mediating the invasion of
bovine epithelial cells and is transcriptionally upregulated in oxy-
gen limiting and solute stress conditions similar to those encoun-
tered within the intestine (Bannantine et al., 2003; Wu et al.,
2007). Based upon in silico analysis MMP is structurally dissimi-
lar to MptD and analysis lacks polytopic transmembrane domains
or highly hydrophobic stretches. The aggregates observed were
initially considered to be indicative of translational disorder
through synonymous codon modification, since MMP expres-
sion in native MAP appears throughout the bacilli by electron
microscopy (Bannantine et al., 2003). Alternatively, aggregation
could be a consequence of L. salivarius reacting to over expres-
sion by inducing vesicle formation to compensate for inefficient
processing or secretion of a non-host protein. Unfortunately,
the negligible fluorescence for native MMP-GFP prevented direct
comparison to determine if these foci also occurred in the absence
of synonymous mutations necessitating use of the monoclonal
antibody based ELISA.
The detection of a non-classically exported protein, such
as MMP, from the extracellular surface of L. salivarius could
indeed be attributed to cell lysis during experimental handling.
However, this would presumably have also occurred for those
L. salivarius cells expressing recombinant MptD, yet this was not
observed (Figure 2). Nevertheless, we acknowledge the behav-
ior of the MMP recombinant strains may be different than
the MptD recombinants and despite the lack of fMptD phage
binding in whole cells, we cannot rule out cell lysis in the
whole cell experiments with the MMP strains. As such, we have
yet to determine the mechanism by which MMP was effec-
tively presented in such a manner from our heterologous host
strain.
Generally, there are three strategies available for the subcellu-
lar distribution of recombinant antigens from Lactobacillus based
vaccine hosts and while cell surface display and secretion are
favored, these can result in degradation of recombinant peptides
due to exposure to proteolytic enzymes associated with gastric
and pancreatic fluids (Kajikawa et al., 2011). Cytoplasmic expres-
sion on the other hand, subverts this and protects heterologously
expressed peptides from degradation by encapsulation within the
cytosol, as well as facilitating the accumulation of high concen-
trations of the antigenic component intracellularly (de Ruyter
et al., 1996). As such, our L. salivarius MMPsynth host may pro-
vide an interesting combination of both high concentrations of
cytoplasmically accumulated MMP, with the added advantage of
superficial surface exposure.
Additionally, these results further demonstrate that heterol-
ogously expressed codon optimized MptD retains fMptD epi-
tope presentation in L. salivarius. However, ELISA data also
suggests that MptD, or at least the epitope recognized by the
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |8
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
fMptD phage, is not exposed on the surface of L. salivarius.
It has already been shown that MptD expression on the cell
surface of a recombinant host can be achieved using fMptD
for both M. smegmatis and M. bovis BCG (Stratmann et al.,
2004; Heinzmann et al., 2008); which could be due to the pres-
ence of mycobacterium specific chaperones facilitating appro-
priate presentation of the MptD protein (Goldstone et al.,
2008). However, mptD is naturally present on a six gene operon
(mptA-F) transcribed as a single polycistronic mRNA molecule
[60] and in both the aforementioned studies, the recombi-
nant hosts were not transformed with the mptD gene in iso-
lation. The M. smegmatis harbored a vector including mptC-F
genes of the mpt operon (Stratmann et al., 2004) while the
M. bovis BCG host contained the entire operon integrated into
the chromosome (Heinzmann et al., 2008). Consequently, it
is possible that for effective MptD cell surface display, some
ancillary Mpt proteins may also be required. The MptE pro-
tein (with five predicted TMSs) encoded by MAP3732c, located
immediately downstream and overlapping the MAP3733c gene
on the mpt operon, is a credible candidate in this respect
owing to its association with the C-terminus of the MptD
protein.
In future studies it may be interesting to ascertain if the
sequential addition of this and other auxiliary mpt genes to
L. salivarius enables native surface display of the MptD protein.
Nevertheless, the lack of cell surface display may not be entirely
disadvantageous in the context of a MAP antigen delivery host.
The association of the MptD protein with the L. salivarius mem-
brane, while incomplete, likely sequestered hydrophobic domains
thus preventing undesirable aggregation and allowing higher lev-
els of intracellular MptD to accumulate. Moreover it is clear from
the analysis of cellular lysate that abundant MptD epitope could
be detected within the cytoplasm, indicating that the synthetic
gene and the L. salivarius host can effectively express and present
MptD antigens.
CONCLUSION
In conclusion, we have demonstrated that the synonymous
mutation of 279 rare or unfavorable codons within the MMP
coding region facilitates improved protein synthesis within L. sali-
varius. Furthermore, both synthetic MMP and MptD proteins
retain their epitopes or structural characteristics allowing them
to effectively mimic the MAP expressed protein. Importantly,
we also noted that while codon optimization enhances heterol-
ogous overexpression, the addition of a C-terminus GFP tag to
both proteins may obstruct some conformational structure from
forming.
Nevertheless, in the absence of a GFP tag and any extrinsic
signaling peptide, both proteins displayed slight, yet noteworthy,
tendencies to associate in the intended location within L. sali-
varius; MMP being detected on the cell surface, while the multi-
TMSs containing protein MptD associated with the cytoplasmic
membrane boundary. This work underscores the potential of
Lactobacillus salivarius to be used within a subunit vaccine devel-
opment against MAP, as additional antigens are optimized for
L. salivarius expression, the next step will require in-vivo testing
to demonstrate true efficacy.
FUNDING INFORMATION
The author’s acknowledge the financial assistance of the Irish
Government through funding under the Food Institutional
Research Measure (FIRM) grant 08RDCIT617 as well as the
USDA-Agricultural Research Service.
ACKNOWLEDGMENTS
The authors thank Norman Stern for providing the Lactobacillus
salivarius NRRL B-30514 strain, Gerald F. Gerlach for provid-
ing the fMptD bacteriophage, and Alan Lucid for his assistance
with bioinformatic analysis. The technical assistance of Janis K.
Hansen at the USDA’s National Animal Disease Center is grate-
fully acknowledged.
SUPPLEMENTARY MATERIAL
The Supplementary Material for this article can be found
online at: http://www.frontiersin.org/journal/10.3389/fcimb.
2014.00120/abstract
REFERENCES
Banasure, K. D., Basagoudanavar, S. H., Chaudhury, P., Tiwari, V., Parihar, N. S.,
and Goswami, P. P. (2001). Identification and characterization of a gene encod-
ing a 35-kDa protein from Mycobacterium avium subspecies paratuberculosis.
FEMS Microbiol. Lett. 196, 195–199. doi: 10.1111/j.1574-6968.2001.tb10564.x
Bannantine, J. P., and Bermudez, L. E. (2013). No holes barred: invasion of
the intestinal mucosa by Mycobacterium av ium subsp. paratuberculosis. Infect
Immun. 81, 3960–3965. doi: 10.1128/IAI.00575-13
Bannantine, J. P., Huntley, J. F., Miltner, E., Stabel, J. R., and Bermudez, L. E.
(2003). The Mycobacterium avium subsp. paratuberculosis 35 kDa protein plays
a role in invasion of bovine epithelial cells. Microbiology 149, 2061–2069. doi:
10.1099/mic.0.26323-0
Bannantine, J. P., Li, L., Mwangi, M., Cote, R., Raygoza Garay, J. A., and
Kapur, V. (2014). Complete genome sequence of Mycobacterium avium subsp.
paratuberculosis, isolated from human breast milk. Genome Announc.2:e01252-
13. doi: 10.1128/genomeA.01252-13
Bannantine, J. P., Paustian, M. L., Waters, W. R., Stabel, J. R., Palmer, M. V., Li,
L., et al. (2008). Profiling bovine antibody responses to Mycobacterium avium
subsp. paratuberculosis infection by using protein arrays. Infect Immun. 76,
739–749. doi: 10.1128/IAI.00915-07
Bannantine, J. P., Radosevich, T. J., Stabel, J. R., Berger, S., Griffin, J. F.,
and Paustian, M. L. (2007). Production and characterization of mono-
clonal antibodies against a major membrane protein of Mycobac terium avium
subsp. paratuberculosis. Clin. Vaccine Immunol. 14, 312–317. doi: 10.1128/CVI.
00353-06
Bannantine, J. P., Stabel, J. R., Bayles, D. O., and Geisbrecht, B. V. (2010).
Characteristics of an extensive Mycobacterium av ium subspecies paratu-
berculosis recombinant protein set. Protein Expr. Purif. 72, 223–233. doi:
10.1016/j.pep.2010.03.019
Bastida, F., and Juste, R. A. (2011). Parat uberculosis control: a review with a
focus on vaccination. J. Immune Based Ther. Vaccines 9:8. doi: 10.1186/1476-
8518-9-8
Bendtsen, J. D., Kiemer, L., Fausboll, A., and Brunak, S. (2005). Non-classical
protein secretion in bacteria. BMC Microbiol. 5:58. doi: 10.1186/1471-2180-5-58
Bermudez-Humaran, L. G., Kharrat, P., Chatel, J. M., and Langella, P. (2011).
Lactococci and lactobacilli as mucosal delivery vectors for therapeutic proteins
and DNA vaccines. Microb. Cell Fact. 10(Suppl. 1), S4. doi: 10.1186/1475-2859-
10-S1-S4
Brown, M. C., Joaquim, T. R., Chambers, R., Onisk, D. V., Yin, F., Moriango, J. M.,
et al. (2011). Impact of immunization technology and assay application on anti-
body performance–a systematic comparative evaluation. PLoS ONE 6:e28718.
doi: 10.1371/journal.pone.0028718
Buchan, J. R., and Stansfield, I. (2007). Halting a cellular production line:
responses to ribosomal pausing during translation. Biol. Cell 99, 475–487. doi:
10.1042/BC20070037
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |9
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
Buddle, B. M., Wilson, T., Denis, M., Greenwald, R., Esfandiari, J., Lyashchenko, K.
P., et al. (2010). Sensitivity, specificity, and confounding factors of novel sero-
logical tests used for the rapid diagnosis of bovine tuberculosis in farmed red
deer (Cervus elaphus). Clin. Vaccine Immunol. 17, 626–630. doi: 10.1128/CVI.
00010-10
Bull, T. J., Gilbert, S. C., Sridhar, S., Linedale, R., Dierkes, N., Sidi-Boumedine,
K., et al. (2007). A novel multi-antigen virally vectored vaccine against
Mycobacterium avium subspecies paratuberculosis.PLoS ONE 2:e1229. doi:
10.1371/journal.pone.0001229
Campos, I. B., Darrieux, M., Ferreira, D. M., Miyaji, E. N., Silva, D. A., Areas, A.
P., et al. (2008). Nasal immunization of mice with Lactobacillus casei express-
ing the Pneumococcal Surface Protein A: induction of antibodies, complement
deposition and partial protection against Streptococcus pneumoniae challenge.
Microbes Infect. 10, 481–488. doi: 10.1016/j.micinf.2008.01.007
Chacon, O., Bermudez, L. E., and Barletta, R. G. (2004). Johne’s disease, inflamma-
tory bowel disease, and Mycobacterium paratuberculosis.Annu. Rev. Microbiol.
58, 329–363. doi: 10.1146/annurev.micro.58.030603.123726
Chappel, J. A., He, M., and Kang, A. S. (1998). Modulation of antibody display on
M13 filamentous phage. J. Immunol. Methods 221, 25–34. doi: 10.1016/S0022-
1759(98)00094-5
Coad, M., Clifford, D. J., Vordermeier, H. M., and Whelan, A. O. (2013). The con-
sequences of vaccination with the Johne’s disease vaccine, Gudair, on diagnosis
of bovine tuberculosis. Vet. R e c. 172, 266. doi: 10.1136/vr.101201
Cossu, A., Rosu, V., Paccagnini, D., Cossu, D., Pacifico, A., and Sechi, L. A. (2011).
MAP3738c and MptD are specific tags of Mycobacteriu m avium subsp. paratu-
berculosis infection in type I diabetes mellitus. Clin. Immunol. 141, 49–57. doi:
10.1016/j.clim.2011.05.002
de Ruyter, P. G., Kuipers, O. P., and De Vos, W. M. (1996). Controlled gene expres-
sion systems for Lactococcus lactis with the food-grade inducer nisin. Appl.
Environ. Microbiol. 62, 3662–3667.
Douarre, P. E., Cashman, W., Buckley, J., Coffey, A., and O’mahony, J. M. (2010).
Isolation and detection of Mycobacterium avium subsp. paratuberculosis (MAP)
from cattle in Ireland using both traditional culture and molecular based
methods. Gut Pathog. 2:11. doi: 10.1186/1757-4749-2-11
Drew, D., Slotboom, D. J., Friso, G., Reda, T., Genevaux, P., Rapp, M., et al.
(2005). A scalable, GFP-based pipeline for membrane protein overexpression
screening and purification. Protein Sci. 14, 2011–2017. doi: 10.1110/ps.0514
66205
Faisal, S. M., Yan, F., Chen, T. T., Useh, N. M., Guo, S., Yan, W., et al. (2013).
Evaluation of a Salmonella vectored vaccine expressing Mycobacter ium avium
subsp. paratuberculosis antigens against challenge in a goat model. PLoS ONE
8:e70171. doi: 10.1371/journal.pone.0070171
Fekkes, P., and Driessen, A. J. (1999). Protein targeting to the bacterial cytoplasmic
membrane. Microbiol. Mol. Biol. Rev. 63, 161–173.
Goldstone, R. M., Moreland, N. J., Bashiri, G., Baker, E. N., and Shaun Lott, J.
(2008). A new Gateway vector and expression protocol for fast and efficient
recombinant protein expression in Mycobacterium smegmatis.Protein Expr.
Purif. 57, 81–87. doi: 10.1016/j.pep.2007.08.015
Gurung, R. B., Purdie, A. C., Begg, D. J., and Whittington, R. J. (2012). In
silico screened Mycobac terium avium subsp. paratuberculosis (MAP) recom-
binant proteins upregulated under stress conditions are immunogenic in
sheep. Vet. Immunol. Immunopathol. 149, 186–196. doi: 10.1016/j.vetimm.2012.
06.026
He, Z., and De Buck, J. (2010). Localization of proteins in the cell wall of
Mycobacterium avium subsp. paratuberculosis K10 by proteomic analysis.
Proteome Sci. 8:21. doi: 10.1186/1477-5956-8-21
Heinzmann, J., Wilkens, M., Dohmann, K., and Gerlach, G. F. (2008).
Mycobacterium avium subsp. paratuberculosis-specific mpt operon expressed
in M. bovis BCG as vaccine candidate. Vet. Microbiol. 130, 330–337. doi:
10.1016/j.vetmic.2008.01.014
Hines, M. E., Turnquist, S. E., Ilha, M. R., Rajeev, S., Jones, A. L., Whittington,
L., et al. (2014). Evaluation of novel oral vaccine candidates and valida-
tion of a caprine model of Johne’s disease. Front. Microbiol. 4:26. doi:
10.3389/fcimb.2014.00026
Johnston, C., Douarre, P. E., Soulimane, T., Pletzer, D., Weingart, H.,
Macsharry, J., et al. (2013). Codon optimisation to improve expression of
aMycobacterium avium ssp. paratuberculosis-specific membrane-associated
antigen by Lactobacillus salivarius. Pathog. Dis. 68, 27–38. doi: 10.1111/2049-
632X.12040
Kajikawa, A., Nordone, S. K., Zhang, L., Stoeker, L. L., Lavoy, A. S.,
Klaenhammer, T. R., et al. (2011). Dissimilar properties of two recombi-
nant Lactobacillus acidophilus strains displaying Salmonella FliC with different
anchoring motifs. Appl. Environ. Microbiol. 77, 6587–6596. doi: 10.1128/AEM.
05153-11
Koets, A., Hoek, A., Langelaar, M., Overdijk, M., Santema, W., Franken, P.,
et al. (2006). Mycobacterial 70 kD heat-shock protein is an effective sub-
unit vaccine against bovine paratuberculosis.Vaccine 24, 2550–2559. doi:
10.1016/j.vaccine.2005.12.019
Kohler, H., Gyra, H., Zimmer, K., Drager, K. G., Burkert, B., Lemser, B., et al.
(2001). Immune reactions in cattle after immunization with a Mycobacterium
paratuberculosis vaccine and implications for the diagnosis of M. paratubercu-
losis and M. bovis infections. J. Vet. Med. B Infect. Dis. Vet. Public Health 48,
185–195. doi: 10.1046/j.1439-0450.2001.00443.x
Li, L., Bannantine, J. P., Zhang, Q., Amonsin, A., May, B. J., Alt, D., et al.
(2005). The complete genome sequence of Mycobacterium avium sub-
species paratuberculosis.Proc. Natl. Acad. Sci. U.S.A. 102, 12344–12349. doi:
10.1073/pnas.0505662102
Li, L., Munir, S., Bannantine, J. P., Sreevatsan, S., Kanjilal, S., and Kapur, V. (2007).
Rapid expression of Mycobac terium avium subsp. paratuberculosis recombi-
nant proteins for antigen discovery. Clin. Vaccine Immunol. 14, 102–105. doi:
10.1128/CVI.00138-06
Magnan, C. N., Randall, A., and Baldi, P. (2009). SOLpro: accurate sequence-
based prediction of protein solubility. Bioinformatics 25, 2200–2207. doi:
10.1093/bioinformatics/btp386
Martin, E., Roche, P. W., Triccas, J. A., and Britton, W. J. (2001). DNA encoding
a single mycobacterial antigen protects against leprosy infection. Vaccine 19,
1391–1396. doi: 10.1016/S0264-410X(00)00374-1
Messaoudi, S., Madi, A., Prevost, H., Feuilloley, M., Manai, M., Dousset, X.,
et al. (2012). In vitro evaluation of the probiotic potential of Lactobacillus
salivarius SMXD51. Anaerobe 18, 584–589. doi: 10.1016/j.anaerobe.2012.
10.004
Mohamadzadeh, M., Duong, T., Sandwick, S. J., Hoover, T., and Klaenhammer,
T. R. (2009). Dendritic cell targeting of Bacillus anthracis protective anti-
gen expressed by Lactobacillus acidophilus protects mice from lethal chal-
lenge. Proc. Natl. Acad. Sci. U.S.A. 106, 4331–4336. doi: 10.1073/pnas.09000
29106
Mohamadzadeh, M., Durmaz, E., Zadeh, M., Pakanati, K. C., Gramarossa, M.,
Cohran, V., et al. (2010). Targeted expression of anthrax protective antigen by
Lactobacillus gasseri as an anthrax vaccine. Future Microbiol. 5, 1289–1296. doi:
10.2217/fmb.10.78
Pallen, M. J. (2002). The ESAT-6/WXG100 superfamily—and a new Gram-
positive secretion system? Trends Microbiol. 10, 209–212. doi: 10.1016/S0966-
842X(02)02345-4
Pavan, S., Hols, P., Delcour, J., Geoffroy, M. C., Grangette, C., Kleerebezem,
M., et al. (2000). Adaptation of the nisin-controlled expression system
in Lactobacillus plantarum: a tool to study in vivo biological effects.
Appl. Environ. Microbiol. 66, 4427–4432. doi: 10.1128/AEM.66.10.4427-44
32.2000
Rosseels, V., and Huygen, K. (2008). Vaccination against paratuber-
culosis.Expert Rev. Vaccines 7, 817–832. doi: 10.1586/14760584.
7.6.817
Rosu, V., Ahmed, N., Paccagnini, D., Gerlach, G., Fadda, G., Hasnain, S. E., et al.
(2009). Specific immunoassays confirm association of Mycobacterium avium
Subsp. paratuberculosis with type-1 but not type-2 diabetes mellitus. PLoS ONE
4:e4386. doi: 10.1371/journal.pone.0004386
Sambrook, J., Fritsch, E. F., and Maniatis, T. (1990). Molecular-cloning–a labora-
tory manual, 2nd Edition. Nature 343, 604–660.
Scandurra, G. M., De Lisle, G. W., Cavaignac, S. M., Young, M., Kawakami, R. P.,
and Collins, D. M. (2010). Assessment of live candidate vaccines for paratuber-
culosis in animal models and macrophages. Infect. Immun. 78, 1383–1389. doi:
10.1128/IAI.01020-09
Shin, S. J., Wu, C. W., Steinberg, H., and Talaat, A. M. (2006). Identification of
novel virulence determinants in Mycobacterium paratuberculosis by screening a
library of insertional mutants. Infect. Immun. 74, 3825–3833. doi: 10.1128/IAI.
01742-05
Sleator, R. D., and Hill, C. (2008). Designer probiotics: a potential therapeutic
for Clostridium difficile?J. Med. Microbiol. 57, 793–794. doi: 10.1099/jmm.0.
47697-0
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |10
Johnston et al. Enhanced M. paratuberulosis antigen expression in L. salivarius
Snapp, E. L. (2009). Fluorescent proteins: a cell biologist’s user guide. Tre nd s Cel l
Biol. 19, 649–655. doi: 10.1016/j.tcb.2009.08.002
Stern, N. J., Svetoch, E. A., Eruslanov, B. V., Perelygin, V. V., Mitsevich, E. V.,
Mitsevich, I. P., et al. (2006). Isolation of a Lactobacillus salivarius strain and
purification of its bacteriocin, which is inhibitory to Campylobacter jejuni in the
chicken gastrointestinal system. Antimicrob. Agents Chemother. 50, 3111–3116.
doi: 10.1128/AAC.00259-06
Stratmann, J., Dohmann, K., Heinzmann, J., and Gerlach, G. F. (2006). Peptide
aMptD-mediated capture PCR for detection of Mycobacterium avium subsp.
paratuberculosis in bulk milk samples. Appl. Environ. Microbiol. 72, 5150–5158.
doi: 10.1128/AEM.00590-06
Stratmann, J., Strommenger, B., Goethe, R., Dohmann, K., Gerlach, G. F.,
Stevenson, K., et al. (2004). A 38-kilobase pathogenicity island specific for
Mycobacterium avium subsp. paratuberculosis encodes cell surface proteins
expressed in the host. Infect Immun. 72, 1265–1274. doi: 10.1128/IAI.72.3.1265-
1274.2004
Triccas, J. A., Roche, P. W., Winter, N., Feng, C. G., Butlin, C. R., and Britton,
W. J. (1996). A 35-kilodalton protein is a major target of the human immune
response to Mycobacterium leprae.Infect. Immun. 64, 5171–5177.
Triccas, J. A., Winter, N., Roche, P. W., Gilpin, A., Kendrick, K. E., and Britton, W.
J. (1998). Molecular and immunological analyses of the Mycobacterium avium
homolog of the immunodominant Mycobacterium leprae 35-kilodalton protein.
Infect. Immun. 66, 2684–2690.
Vrieling, M., Santema, W., Vordermeier, M., Rutten, V., and Koets, A. (2013).
Hsp70 vaccination-induced primary immune responses in efferent lymph of
the draining lymph node. Vaccine 31, 4720–4727. doi: 10.1016/j.vaccine.2013.
08.021
Wang, Z., Yu, Q., Gao, J., and Yang, Q. (2012). Mucosal and systemic immune
responses induced by recombinant Lactobacillus spp. expressing the hemagglu-
tinin of the avian influenza virus H5N1. Clin. Vaccine Immunol. 19, 174–179.
doi: 10.1128/CVI.05618-11
Winter, N., Triccas, J. A., Rivoire, B., Pessolani, M. C., Eiglmeier, K., Lim, E.
M., et al. (1995). Characterization of the gene encoding the immunodomi-
nant 35 kDa protein of Mycobacterium leprae.Mol. Microbiol. 16, 865–876. doi:
10.1111/j.1365-2958.1995.tb02314.x
Wu, C. W., Schmoller, S. K., Shin, S. J., and Talaat, A. M. (2007). Defining
the stressome of Mycobacterium avium subsp. paratuberculosis in vitro and
in naturally infected cows. J. Bacteriol. 189, 7877–7886. doi: 10.1128/JB.
00780-07
Yakes, B. J., Lipert, R. J., Bannantine, J. P., and Porter, M. D. (2008).
Impact of protein shedding on detection of Mycobacterium av ium subsp.
paratuberculosis by a whole-cell immunoassay incorporating surface-enhanced
Raman scattering. Clin. Vaccine Immunol. 15, 235–242. doi: 10.1128/CVI.
00335-07
Yu, Q., Zhu, L., Kang, H., and Yang, Q. (2013). Mucosal Lactobacillus vec-
tored vaccines. Hum. Vaccin. Immunother. 9, 805–807. doi: 10.4161/
hv.23302
Conflict of Interest Statement: The Associate Editor Dr. Adel Talaat declares
that despite having collaborated with the author Dr. John Bananntine, the
review process was handled objectively and no conflict of interest exists. The
authors declare that the research was conducted in the absence of any commer-
cial or financial relationships that could be construed as a potential conflict of
interest.
Received: 24 April 2014; accepted: 15 August 2014; published online: 04 September
2014.
Citation: Johnston CD, Bannantine JP, Govender R, Endersen L, Pletzer D, Weingart
H, Coffey A, O’Mahony J and Sleator RD (2014) Enhanced expression of codon
optimized Mycobacterium avium subsp. paratuberculosis antigens in Lactobacillus
salivarius. Front. Cell. Infect. Microbiol. 4:120. doi: 10.3389/fcimb.2014.00120
This article was submitted to the journal Frontiers in Cellular and Infection
Microbiology.
Copyright © 2014 Johnston, Bannantine, Govender, Endersen, Pletzer, Weingart,
Coffey, O’Mahony and Sleator. This is an open-access article distributed under the
terms of the Creative Commons Attribution License (CC BY). The use, distribution or
reproduction in other forums is permitted, provided the original author(s) or licensor
are credited and that the original publication in this journal is cited, in accordance w ith
accepted academic practice. No use, distribution or reproduction is permitted which
does not comply with these terms.
Frontiers in Cellular and Infection Microbiology www.frontiersin.org September 2014 | Volume 4 | Article 120 |11
... A frequently occurring problem is the over-expression of genes derived from G+C rich coding sequences in an A+T rich Lactobacillus host. Johnston et al. (2014) demonstrated that codon optimization by synonymous substitutions improves expression levels of Mycobacterium avium subsp. paratuberculosis antigens in L. salivarius ( Johnston et al., 2014). ...
... Johnston et al. (2014) demonstrated that codon optimization by synonymous substitutions improves expression levels of Mycobacterium avium subsp. paratuberculosis antigens in L. salivarius ( Johnston et al., 2014). ...
... Although prokaryotic expression systems (mainly based on Escherichia coli) are very efficient for the production of several subunit vaccines (Johnston et al. 2015), they have some limitations, such as the need of purification of the recombinant antigen due to the toxic compounds from the host and limitations for the synthesis of complex antigens (Mamat et al. 2015). In this context, plant-made veterinary vaccines have advantages compared to prokaryotic systems including (1) lower contamination risks, (2) lower cost, (3) easier scaling-up, and (4) the possibility of oral administration (Shahid and Daniell 2016;Rosales-Mendoza et al. 2017). ...
Article
Full-text available
Transient expression systems for plants have several advantages for the development of innovative plant-made vaccines. This technology has the potential to render veterinary vaccines against relevant diseases, such as paratuberculosis, a chronic intestinal infectious disease affecting livestock. This study showed that Agrobacterium-mediated transient transformation of maize seedlings allows the production of the Ag85B antigen from Mycobacterium avium subsp. paratuberculosis (MAP) at levels up to 11.35 µg g−1 dry weight. Moreover, BALB/c mice orally and subcutaneously immunized with the maize-made Ag85B vaccine produced specific IgG and IgA antibodies. Therefore, this platform is proposed as a rapid approach to produce the functional MAPAg85B antigen at sufficient yields to induce specific humoral immune responses in animal testing, which constitutes the first step in the development of a plant-made oral vaccine against paratuberculosis.
... In addition to the typical classification of the central nervous system (CNS) as an immune privileged area, increasing evidence points to the reciprocal communication between the periphery and brain in neurophysiological features, including mood and behavior (Bercik et al., 2011;Johnston et al., 2014). In particular, the gut microbiota is increasingly considered one of the most important actors in the bidirectional interactions between the intestine and brain, in both physiological and pathological conditions (Scriven et al., 2018). ...
Article
Recent evidence points to the gut microbiota as a regulator of brain and behavior, although it remains to be determined if gut bacteria play a role in chronic pain. The endocannabinoid system is implicated in inflammation and chronic pain processing at both the gut and central nervous system (CNS) levels. In the present study, we used low Vitamin D dietary intake in mice and evaluated possible changes in gut microbiota, pain processing and endocannabinoid system signaling. Vitamin D deficiency induced a lower microbial diversity characterized by an increase in Firmicutes and a decrease in Verrucomicrobia and Bacteroidetes. Concurrently, vitamin D deficient mice showed tactile allodynia associated with neuronal hyperexcitability and alterations of endocannabinoid system members (endogenous mediators and their receptors) at the spinal cord level. Changes in endocannabinoid (anandamide and 2-arachidonoylglycerol) levels were also observed in the duodenum and colon. Remarkably, the anti-inflammatory anandamide congener, palmitoylethanolamide, counteracted both the pain behaviour and spinal biochemical changes in vitamin D deficient mice, whilst increasing the levels of Akkermansia, Eubacterium and Enterobacteriaceae, as compared with vehicle-treated mice. Finally, induction of spared nerve injury in normal or vitamin D deficient mice was not accompanied by changes in gut microbiota composition. Our data suggest the existence of a link between Vitamin D deficiency – with related changes in gut bacterial composition – and altered nociception, possibly via molecular mechanisms involving the endocannabinoid and related mediator signaling systems.
... Delivery of expressed MAP antigens through attenuated strains like Salmonella and Lactobacillus salivarius was shown to be an alternate way to stimulate protective mucosal immune responses [12,13].This approach is, however in primitive stage and needs further studies to prove its usefulness in the development of subunit vaccines. Combination of viral vectors with MAP antigens was also tried to develop DNA subunit vaccines. ...
... Alternatively, the generation of libraries of synthetic constitutive promoters displaying a wide range of strength (Jensen and Hammer 1998;Rud et al. 2006) appears to be a potent tool for mimicking native cellulase systems, in which the highest synergism is obtained for non-equimolar expression of different enzymes (Mazzoli, Lamberti and Pessione 2012). Additional tools to increase cellulase/hemicellulose expression in LAB include improvement of mRNA stability (Narita, Ishida and Okano 2006;Okano et al. 2010a) or increase of translation efficiency through design of synthetic genes with optimised codon usage (Johnston et al. 2014;Dong et al. 2015;Li et al. 2016). The most challenging factor in heterologous expression of cellulases consists in finding an efficient secretion strategy (Mazzoli, Lamberti and Pessione 2012). ...
Article
Lactic acid bacteria (LAB) have a long history in industrial processes as food starters and biocontrol agents, and also as producers of high-value compounds. Lactic acid, their main product, is among the most requested chemicals because of its multiple applications, including the synthesis of biodegradable plastic polymers. Moreover, LAB are attractive candidates for the production of ethanol, polyhydroalkanoates, sweeteners and exopolysaccharides. LAB generally have complex nutritional requirements. Furthermore, they cannot directly ferment inexpensive feedstocks such as lignocellulose. This significantly increases the cost of LAB fermentation and hinders its application in the production of high volumes of low-cost chemicals. Different strategies have been explored to extend LAB fermentation to lignocellulosic biomass. Fermentation of lignocellulose hydrolysates by LAB has been frequently reported and is the most mature technology. However, current economic constraints of this strategy have driven research for alternative approaches. Co-cultivation of LAB with native cellulolytic microorganisms may reduce the high cost of exogenous cellulase supplementation. Special attention is given in this review to the construction of recombinant cellulolytic LAB by metabolic engineering, which may generate strains able to directly ferment plant biomass. The state of the art of these strategies is illustrated along with perspectives of their applications to industrial second generation biorefinery processes.
... In the context of vectored subunit vaccines, success has been achieved in the expression of MAP antigens in Lactobacillus salivarius using a codon optimization strategy (151,152). Multi-antigen virally vectored vaccines were also engineered and tested in mice and cattle. In this context, the vaccine designated HAV is a fusion of two secreted and two cell surface MAP proteins whose DNA coding sequence was inserted into the human adenovirus 5 and Modified Vaccinia Ankara delivery vectors. ...
Article
Full-text available
Mycobacterium avium subsp. paratuberculosis (MAP) is the etiologic agent of Johne’s disease in ruminants causing chronic diarrhea, malnutrition, and muscular wasting. Neonates and young animals are infected primarily by the fecal–oral route. MAP attaches to, translocates via the intestinal mucosa, and is phagocytosed by macrophages. The ensuing host cellular immune response leads to granulomatous enteritis characterized by a thick and corrugated intestinal wall. We review various tissue culture systems, ileal loops, and mice, goats, and cattle used to study MAP pathogenesis. MAP can be detected in clinical samples by microscopy, culturing, PCR, and an enzyme-linked immunosorbent assay. There are commercial vaccines that reduce clinical disease and shedding, unfortunately, their efficacies are limited and may not engender long-term protective immunity. Moreover, the potential linkage with Crohn’s disease and other human diseases makes MAP a concern as a zoonotic pathogen. Potential therapies with anti-mycobacterial agents are also discussed. The completion of the MAP K-10 genome sequence has greatly improved our understanding of MAP pathogenesis. The analysis of this sequence has identified a wide range of gene functions involved in virulence, lipid metabolism, transcriptional regulation, and main metabolic pathways. We also review the transposons utilized to generate random transposon mutant libraries and the recent advances in the post-genomic era. This includes the generation and characterization of allelic exchange mutants, transcriptomic analysis, transposon mutant banks analysis, new efforts to generate comprehensive mutant libraries, and the application of transposon site hybridization mutagenesis and transposon sequencing for global analysis of the MAP genome. Further analysis of candidate vaccine strains development is also provided with critical discussions on their benefits and shortcomings, and strategies to develop a highly efficacious live-attenuated vaccine capable of differentiating infected from vaccinated animals.
... Subunit vaccines have been reported to provide incomplete protection in murine models (Stabel, Barnhill, Bannantine, Chang, & Osman, 2012) or ruminant models (calves and goats) of infection (Kathaperumal et al., 2008(Kathaperumal et al., , 2009Koets et al., 2006). In addition, other subunit vaccines (Facciuolo & Mutharia, 2014;Gurung, Begg, Purdie, & Whittington, 2013;Johnston et al., 2014) are being proposed as having potential to prevent infection. Most vaccines have been tested in a preventive pre-exposure setting. ...
Article
In the last decades, many regional and country-wide control programmes for Johne's disease (JD) were developed due to associated economic losses, or because of a possible association with Crohn's disease. These control programmes were often not successful, partly because management protocols were not followed, including the introduction of infected replacement cattle, because tests to identify infected animals were unreliable, and uptake by farmers was not high enough because of a perceived low return on investment. In the absence of a cure or effective commercial vaccines, control of JD is currently primarily based on herd management strategies to avoid infection of cattle and restrict within-farm and farm-to-farm transmission. Although JD control programmes have been implemented in most developed countries, lessons learned from JD prevention and control programmes are underreported. Also, JD control programmes are typically evaluated in a limited number of herds and the duration of the study is less than 5 year, making it difficult to adequately assess the efficacy of control programmes. In this manuscript, we identify the most important gaps in knowledge hampering JD prevention and control programmes, including vaccination and diagnostics. Secondly, we discuss directions that research should take to address those knowledge gaps.
... Indeed, the treatment schedules and the pharmacokinetic properties of the agents can affect bacterial organisation and, in turn, (positively or negatively) the behaviour (Bercik et al., 2011;Johnston et al., 2014). Here, we used a broad-spectrum activity antibiotics cocktail (Lamouse-Smith et al., 2011) in order to generate an overall microflora perturbation and identify possible biomolecular mechanisms responsible for behavioural modifications. ...
Article
The microbiota-gut-brain axis (MGBA) regulates the reciprocal interaction between chronic inflammatory bowel and psychiatric disorders. This interaction involves multiple pathways that are highly debated. We examined the behavioural, biochemical and electrophysiological alterations, as well as gut microbiota composition in a model of antibiotic-induced experimental dysbiosis. Inflammation of the small intestine was also assessed. Mice were exposed to a mixture of antimicrobials for 2 weeks. Afterwards, they received Lactobacillus casei DG (LCDG) or a vehicle for up to 7 days via oral gavage. Perturbation of microbiota was accompanied by a general inflammatory state and alteration of some endocannabinoidome members in the gut. Behavioural changes, including increased immobility in the tail suspension test and reduced social recognition were observed, and were associated with altered BDNF/TrkB signalling, TRPV1 phosphorylation and neuronal firing in the hippocampus. Moreover, morphological rearrangements of non-neuronal cells in brain areas controlling emotional behaviour were detected. Subsequent probiotic administration, compared with vehicle, counteracted most of these gut inflammatory, behavioural, biochemical and functional alterations. Interestingly, levels of Lachnospiraceae were found to significantly correlate with the behavioural changes observed in dysbiotic mice. Our findings clarify some of the biomolecular and functional modifications leading to the development of affective disorders associated with gut microbiota alterations.
Chapter
Current developments in microbial research and synthetic biology have motivated the investigators to develop a diverse range of engineered microbes expressing valuable biotherapeutics targeting a range of human clinical applications. Probiotics, developed through a recombinant approach, represent the new era of whole cell-mediated biotherapeutics. These engineered probiotics are target specific and have high endurance capability, which makes them a perfect vaccine or drug carrying vehicle in the host system. This chapter outlines the diverse capabilities of biotherapeutics expressed through engineered microbes as a potent engineered probiotic, for the treatment of different chronic human pathologies including metabolic disorder, malignancy, chronic inflammation bowel disease concerned with the gastrointestinal tract and autoimmune disorders, respectively. This chapter also entails the significant advancement in the area of reprogrammed microbes used as an effective bio-scavenger, for the remediation of toxic heavy metals from the environment. The sustainable production of target specific biotherapeutics from a recombinant microbiome will enable us to overcome the burden of expensive drugs with severe side effects. This modern concept of engineered biotherapeutics will narrow down many technical and economic barriers associated with drug development globally. The modern era on microbial biotherapeutics will ensure healthy lives and promote the well-being of the global population.
Article
Full-text available
Lactic acid bacteria (LAB) have been utilized since the 1990s for therapeutic heterologous gene expression. The ability of LAB to elicit an immune response against expressed foreign antigens has led to their exploration as potential mucosal vaccine candidates. LAB vaccine vectors offer many attractive advantages: simple, noninvasive administration (usually oral or intranasal), the acceptance and stability of genetic modifications, relatively low cost, and the highest level of safety possible. Experimentation using LAB of the genus Lactobacillus has become popular in recent years due to their ability to elicit strong systemic and mucosal immune responses. This article reviews Lactobacillus vaccine constructs, including Lactobacillus species, antigen expression, model organisms, and in vivo immune responses, with a primary focus on viral and bacterial antigens.
Article
Full-text available
Johne's disease (JD) caused by Mycobacterium avium subspecies paratuberculosis (MAP) is a major threat to the dairy industry and possibly some cases of Crohn's disease in humans. A MAP vaccine that reduced of clinical disease and/or reduced fecal shedding would aid in the control of JD. The objectives of this study were (1) to evaluate the efficacy of 5 attenuated strains of MAP as vaccine candidates compared to a commercial control vaccine using the protocol proposed by the Johne's Disease Integrated Program (JDIP) Animal Model Standardization Committee (AMSC), and (2) to validate the AMSC Johne's disease goat challenge model. Eighty goat kids were vaccinated orally twice at 8 and 10 weeks of age with an experimental vaccine or once subcutaneously at 8 weeks with Silirum® (Zoetis), or a sham control oral vaccine at 8 and 10 weeks. Kids were challenged orally with a total of approximately 1.44 × 10⁹ CFU divided in two consecutive daily doses using MAP ATCC-700535 (K10-like bovine isolate). All kids were necropsied at 13 months post-challenge. Results indicated that the AMSC goat challenge model is a highly efficient and valid model for JD challenge studies. None of the experimental or control vaccines evaluated prevented MAP infection or eliminated fecal shedding, although the 329 vaccine lowered the incidence of infection, fecal shedding, tissue colonization and reduced lesion scores, but less than the control vaccine. Based on our results the relative performance ranking of the experimental live-attenuated vaccines evaluated, the 329 vaccine was the best performer, followed by the 318 vaccine, then 316 vaccine, 315 vaccine and finally the 319 vaccine was the worst performer. The subcutaneously injected control vaccine outperformed the orally-delivered mutant vaccine candidates. Two vaccines (329 and 318) do reduce presence of JD gross and microscopic lesions, slow progression of disease, and one vaccine (329) reduced fecal shedding and tissue colonization.
Article
Full-text available
Mycobacterium avium subsp. paratuberculosis causes Johne’s disease (JD) in cattle, sheep, goats, and other ruminant animals. JD presents as a chronic granulomatous intestinal infection with a worldwide distribution and imposes a significant economic toll on livestock industries (1). M. avium subsp. paratuberculosis has a complex cell wall structure containing mycolic acids and several lipids similar to those of other members of this genus, yet it is the most slowly growing member. This bacterium often requires 8 to 16 weeks before colonies are visible in culture, which is a major hurdle in diagnostics and therefore in the implementation of optimal JD control measures. Although a well-established domestic and wild animal pathogen, it has also been implicated as a causative agent in human Crohn’s disease (2), and even though this link is controversial (3), M. avium subsp. paratuberculosis isolates have been obtained from humans. For instance, M. avium subsp. paratuberculosis 4, the isolate whose sequence we report here, was originally isolated from the breast milk of a Crohn’s disease patient in 2000 (4).
Article
Full-text available
Johnes disease (JD), caused by Mycobacterium avium subsp paratuberculosis (MAP), occurs worldwide as chronic granulomatous enteritis of domestic and wild ruminants. To develop a cost effective vaccine, in a previous study we constructed an attenuated Salmonella strain that expressed a fusion product made up of partial fragments of MAP antigens (Ag85A, Ag85B and SOD) that imparted protection against challenge in a mouse model. In the current study we evaluated the differential immune response and protective efficacy of the Sal-Ag vaccine against challenge in a goat model as compared to the live attenuated vaccine MAP316F. PBMCs from goats vaccinated with Sal-Ag and challenged with MAP generated significantly lower levels of IFN-γ, following in vitro stimulation with either Antigen-mix or PPD jhonin, than PBMC from MAP316F vaccinated animals. Flow cytometric analysis showed the increase in IFN-γ correlated with a significantly higher level of proliferation of CD4, CD8 and γδT cells and an increased expression of CD25 and CD45R0 in MAP316F vaccinated animals as compared to control animals. Evaluation of a range of cytokines involved in Th1, Th2, Treg, and Th17 immune responses by quantitative PCR showed low levels of expression of Th1 (IFN-γ, IL-2, IL-12) and proinflammatory cytokines (IL-6, IL-8, IL-18, TNF-α) in the Sal-Ag immunized group. Significant levels of Th2 and anti-inflammatory cytokines transcripts (IL-4, IL-10, IL-13, TGF-β) were expressed but their level was low and with a pattern similar to the control group. Over all, Sal-Ag vaccine imparted partial protection that limited colonization in tissues of some animals upon challenge with wild type MAP but not to the level achieved with MAP316F. In conclusion, the data indicates that Sal-Ag vaccine induced only a low level of protective immunity that failed to limit the colonization of MAP in infected animals. Hence the Sal-Ag vaccine needs further refinement to increase its efficacy.
Article
Full-text available
The infection biology of Mycobacterium avium subsp. paratuberculosis has recently crystallized, with added details surrounding intestinal invasion. The involvement of pathogen-derived effector proteins such as the major membrane protein, oxidoreductase, and fibronectin attachment proteins have been uncovered. Mutations constructed in this pathogen have also shed light on genes needed for invasion. The host cell types that are susceptible to invasion have been defined, along with their transcriptional response. Recent details have given a new appreciation for the dynamic interplay between the host and bacterium that occurs at the outset of infection. An initial look at the global expression pathways of the host has shown a circumvention of the cell communication pathway by M. avium subsp. paratuberculosis, which loosens the integrity of the tight junctions. We now know that M. avium subsp. paratuberculosis activates the epithelial layer and also actively recruits macrophages to the site of infection. These notable findings are summarized along with added mechanistic details of the early infection model. We conclude by proposing critical next steps to further elucidate the process of M. avium subsp. paratuberculosis invasion.
Article
Full-text available
Not Available Bibtex entry for this abstract Preferred format for this abstract (see Preferences) Find Similar Abstracts: Use: Authors Title Return: Query Results Return items starting with number Query Form Database: Astronomy Physics arXiv e-prints
Article
Bovine paratuberculosis is a highly prevalent chronic infection of the small intestine in cattle, caused by Mycobacterium avium subspecies paratuberculosis (MAP). In earlier studies we showed the protective effect of Hsp70/DDA subunit vaccination against paratuberculosis. In the current study we set out to measure primary immune responses generated at the site of Hsp70 vaccination. Lymph vessel cannulation was performed to obtain efferent lymph from the prescapular lymph node draining the neck area where the vaccine was applied. Hsp70 vaccination induced a significant increase of CD21(+) B cells in efferent lymph, accounting for up to 40% of efferent cells post-vaccination. Proliferation (Ki67(+)) within the CD21(+) B cell and CD4(+) T cell populations peaked between day 3 and day 5 post-vaccination. From day 7, Hsp70-specific antibody secreting cells (ASCs) could be detected in efferent lymph. Hsp70-specific antibodies, mainly of the IgG1 isotype, were also detected from this time point onwards. However, post-vaccination IFN-γ production in efferent lymph was non-sustained. In conclusion, Hsp70-vaccination induces only limited Th1 type immune responsiveness as reflected in efferent lymph draining the vaccination site. This is in line with our previous observations in peripheral blood. The main primary immunological outcome of the Hsp70/DDA subunit vaccination is B cell activation and abundant Hsp70-specific IgG1 production. This warrants the question whether Hsp70-specific antibodies contribute to the observed protective effect of Hsp70 vaccination in calves.