Boise State University Boise State University
ScholarWorks ScholarWorks
Biology Faculty Publications and Presentations Department of Biological Sciences
1-2020
Evaluation of the E=cacy of a Cholera Toxin-Based Evaluation of the E=cacy of a Cholera Toxin-Based
Staphylococcus aureus
Vaccine Against Bovine Intramammary Vaccine Against Bovine Intramammary
Challenge Challenge
Hussain A. Alabdullah
University of Idaho
Elise Overgaard
Boise State University
Danielle Scarbrough
Boise State University
Janet E. Williams
University of Idaho
Omid Mohammad Mousa
Boise State University
See next page for additional authors
Publication Information Publication Information
Alabdullah, Hussain A.; Overgaard, Elise; Scarborough, Danielle; Williams, Janet E.; Mousa, Omid
Mohammad; Dunn, Gary; . . . and Tinker, Juliette K. (2021). Evaluation of the E=cacy of a Cholera Toxin-
Based
Staphylococcus aureus
Vaccine Against Bovine Intramammary Challenge.
Vaccines, 9
(1), 6.
https://doi.org/10.3390/vaccines9010006
Authors Authors
Hussain A. Alabdullah, Elise Overgaard, Danielle Scarbrough, Janet E. Williams, Omid Mohammad Mousa,
Gary Dunn, Laura Bond, Mark A. McGuire, and Juliette K. Tinker
This article is available at ScholarWorks: https://scholarworks.boisestate.edu/bio_facpubs/697
Article
Evaluation of the Efficacy of a Cholera Toxin-Based
Staphylococcus aureus Vaccine against Bovine
Intramammary Challenge
Hussain A. Alabdullah
1,
, Elise Overgaard
2,
, Danielle Scarbrough
2
, Janet E. Williams
1
,
Omid Mohammad Mousa
3
, Gary Dunn
3
, Laura Bond
4
, Mark A. McGuire
1
and Juliette K. Tinker
2,3,
*

 
Citation: Alabdullah, H.A.; Overgaard,
E.; Scarbrough, D.; Williams, J.E.;
Mohammad Mousa, O.; Dunn, G.;
Bond, L.; McGuire, M.A.; Tinker, J.K.
Evaluation of the Efficacy of a Cholera
Toxin-Based Staphylococcus aureus
Vaccine against Bovine Intramammary
Challenge. Vaccines 2021, 9, 6. https://
dx.doi.org/10.3390/vaccines9010006
Received: 24 November 2020
Accepted: 18 December 2020
Published: 24 December 2020
Publishers Note: MDPI stays neu-
tral with regard to jurisdictional claims
in published maps and institutional
affiliations.
Copyright: © 2020 by the authors. Li-
censee MDPI, Basel, Switzerland. This
article is an open access article distributed
under the terms and conditions of the
Creative Commons Attribution (CC BY)
license (https://creativecommons.org/
licenses/by/4.0/).
1
Department of Animal and Veterinary Science, University of Idaho, Moscow, ID 83844, USA;
2
Biomolecular Sciences Graduate Program, Boise State University, Boise, ID 83725, USA;
eliseovergaar[email protected] (E.O.); [email protected] (D.S.)
3
Department of Biological Sciences, Boise State University, Boise, ID 83725, USA;
4
Biomolecular Research Center, Boise State University, Boise, ID 83725, USA; [email protected]
* Correspondence: [email protected]
The authors contribute equally.
Abstract:
Staphylococcus aureus (S. aureus) is a primary agent of bovine mastitis and a source of signifi-
cant economic loss for the dairy industry. We previously reported antigen-specific immune induction
in the milk and serum of dairy cows following vaccination with a cholera toxin A
2
and B subunit
(CTA
2
/B) based vaccine containing the iron-regulated surface determinant A (IsdA) and clumping
factor A (ClfA) antigens of S. aureus (IsdA + ClfA-CTA
2
/B). The goal of the current study was to
assess the efficacy of this vaccine to protect against S. aureus infection after intramammary chal-
lenge. Six mid-lactation heifers were randomized to vaccinated and control groups. On days 1 and
14 animals were inoculated intranasally with vaccine or vehicle control, and on day 20 animals were
challenged with S. aureus. Clinical outcome, milk quality, bacterial shedding, and somatic cell count
(SCC) were followed for ten days post-challenge. Vaccinated animals did not show signs of clinical
S. aureus mastitis and had lower SCCs compared to control animals during the challenge period.
Reductions in bacterial shedding were observed but were not significant between groups. Antibody
analysis of milk and serum indicated that, upon challenge, vaccinated animals produced enhanced
IsdA- and ClfA-CTA
2
/B specific immunoglobulin G (IgG) responses, while responses to CTA
2
/B
alone were not different between groups. Responses after challenge were largely IgG1 against the
IsdA antigen and mixed IgG1/IgG2 against the ClfA antigen. In addition, there was a significant
increase in interferon gamma (IFN-
γ
) expression from blood cells in vaccinated animals on day 20.
While preliminary, these findings support evidence of the induction of active immunity by IsdA +
ClfA-CTA
2
/B, and further assessment of this vaccine is warranted.
Keywords: Staphylococcus aureus; vaccine; bovine; mastitis
1. Introduction
Mastitis, or inflammation of the udder, is one of the most economically-significant
diseases affecting dairy cattle worldwide and is most often the result of a bacterial infection.
Staphylococcus aureus (S. aureus), a main etiological agent, is highly contagious and can
spread rapidly among herds. It is estimated that up to 70% of U.S. herds are positive
for S. aureus, and this bacterium caused the highest overall annual yield losses among
other mastitis pathogens in a recent Finnish study [
1
,
2
]. S. aureus infections are most
commonly transmitted during the milking process and can impact animal welfare as well
as milk yield and quality [
3
]. The ability of this bacterium to form biofilms and replicate
intracellularly can promote subclinical colonization of the mammary gland, often leading
Vaccines 2021, 9, 6. https://dx.doi.org/10.3390/vaccines9010006 https://www.mdpi.com/journal/vaccines
Vaccines 2021, 9, 6 2 of 17
to chronic infection, which is difficult to detect and is frequently the source of herd re-
infection [
4
6
]. S. aureus is also commonly resistant to antimicrobial treatment and has
a low expected cure rate during lactation [
7
]. While the impact of S. aureus infection is
difficult to quantify, clinical mastitis caused by Gram-positive pathogens is reported to cost
between $133 and $444 per case, or as much as USD 2 billion annually [
8
,
9
]. These costs
include many factors such as milk loss, veterinary expenses, diagnostic testing, and loss
of animals. Prevention of S. aureus mastitis with a cost-effective vaccine would improve
animal welfare, reduce antibiotic use, and positively impact the economics and efficiency
of milk production.
Previous approaches to S. aureus vaccination in cattle include whole-cell live and killed
vaccines as well as purified antigens. Currently, two whole-cell inactivated vaccines are
licensed for protection against S. aureus mastitis—Lysigin
®
(Boehringer Ingelheim, Duluth,
GA, USA) and Startvac
®
(Hipra, Girona, Spain). While efficacy studies are somewhat con-
flicting, these vaccines have reported moderate decreases in the incidence of new
S. aureus
intramammary infection but are not in widespread use [
10
15
]. Recent studies have fo-
cused on the use of multiple purified surface adhesins and secreted virulence factors to
develop a vaccine that offers more strain-to-strain cross-protection. Iron-regulated surface
determinant A (IsdA) is a fibrinogen- and fibronectin-binding adhesin that contributes to
iron sequestration and is a well-studied S. aureus vaccine candidate [
16
19
]. The presence
of isdA is conserved among bovine S. aureus, and IsdA is expressed from these strains in
milk [
18
,
20
23
]. The clumping factor A (ClfA) fibrinogen adhesin is also highly conserved,
expressed from bovine clinical isolates, and a recognized vaccine candidate against masti-
tis [
24
30
]. The conservation, surface exposure, and importance in multiple mechanisms of
pathogenesis supports the inclusion of the IsdA and ClfA antigens in a multivalent bovine
vaccine. However, a number of additional antigens have been characterized and may be
necessary to protect against multiple S. aureus serotypes.
While immune correlates of protection are not known, an understanding of immune
responses is needed to inform antigen selection. The induction of both humoral and cellular
immunity is essential to combating intracellular S. aureus infection [
31
33
]. Cellular sub-
populations that play a central role in defense against S. aureus include neutrophils, CD8
+
T lymphocytes, and CD4
+
Th17 lymphocytes [
34
,
35
]. Cholera toxin (CT), produced by the
bacterium Vibrio cholerae, and the homologous heat-labile toxin I (LTI), produced by the
bacterium Escherichia coli, are gold standard vaccine adjuvants that can stimulate systemic
immunity from mucosal and dermal sites (reviewed in [
36
]). The mechanism of adju-
vanticity of these toxins depends upon active binding subunit targeting of dendritic cells
and neutrophils, and has been attributed to enhanced antigen presentation, upregulation
of surface molecules, and promotion of B-cell isotype switching to antigen-specific im-
munoglobulin A (IgA) and immunoglobulin G (IgG) [
37
41
]. CT and its non-toxic binding
subunit (CTB) can also induce Th1, Th2, and Th17 responses [4244].
The toxic A subunit of CT (CTA) is subdivided into an enzymatically-active domain
(CTA
1
) and a linker domain (CTA
2
), which is non-covalently associated with the B subunit.
CTA
2
/B chimeras were first described as a mechanism to make stable human vaccines
with antigens coupled to the CTB subunit via the A
2
linker domain [
45
,
46
]. These non-toxic
molecules retain the adjuvanticity of CTB and possess additional advantages including
ease of purification, direct association of antigen to adjuvant, and a holotoxin-like structure
that retains binding and internalization motifs [
47
,
48
]. As reported previously, we have
incorporated S. aureus IsdA and ClfA into a CTA
2
/B vaccine platform (IsdA + ClfA-
CTA
2
/B). After two intranasal doses this vaccine was found to stimulate significant S.
aureus antigen-specific humoral and cellular immunity in bovine blood and milk [49].
For this study we hypothesized that intranasal IsdA + ClfA-CTA
2
/B would be effective
in reducing or eliminating S. aureus shedding and disease after intramammary challenge in
cattle. We describe a preliminary trial to determine the efficacy of this mucosal enterotoxin-
based vaccine to protect against acute S. aureus mastitis. While the vaccine did not prevent
bacterial shedding after challenge, results indicate that IsdA + ClfA-CTA
2
/B induces
Vaccines 2021, 9, 6 3 of 17
antigen-specific immune responses that may contribute to a reduction in clinical severity
and infiltration of leukocytes, or SCC, in infected animals.
2. Materials and Methods
2.1. Bacterial Strains, Plasmids, and Growth Conditions
S. aureus Newbould 305 was used for the cloning of isdA and clfA to construct IsdA +
ClfA-CTA
2
/B and was also used for bacterial challenge [
22
,
50
]. E. coli ClearColi
®
(Lucigen,
Madison, WI, USA) was used for protein expression (Table 1). The vector pARLDR19
expressing CTA
2
/B and containing a multiple cloning site was used to construct the
plasmids pLR001 for Isd-CTA
2
/B expression and pLR003 for ClfA-CTA
2
/B expression
(Figure 1A) as described previously [
51
]. For bacterial challenge, S. aureus Newbould 305
was prepared as described [
10
]. Briefly, Newbould 305 was grown at 37
C with shaking
to mid-log phase in brain–heart infusion and harvested by centrifugation at 3000
×
g for
15 min at 4
C. The cell pellet was washed with phosphate-buffered saline (1X PBS, pH 7.2)
and adjusted to an optical density (O.D.) of 0.2 at 600 nm. Serial dilutions were performed
in 1X PBS to reach a bacterial concentration of 400 CFU/mL, as determined by plating on
blood agar (BA).
Table 1. Bacterial strains, plasmids, and primers used in this study.
Bacterial Strains Genotype or Characteristics Source
E. coli ClearColi
®
BL21(DE3) Lucigen, Madison, WI
S. aureus Newbould 305 Bovine clinical isolate [50]
Plasmids Gene Vector Source
pLR001 isdA (Newbould) pARLDR19 [49]
pLR003 clfA (Newbould) pARLDR19 [49]
Bovine Cytokine qPCR Primers Gene Amplicon Source
FW 5
0
-GCATCGTGGAGGGACTTATGA-3
0
GAPDH 67
[52]
RV 5
0
-GGGCCATCCACAGTCTTCTG-3
0
FW 5
0
-CTTGTCGGAAATGATCCAGTTTT-3
0
IL-10 66
[53]
RV 5
0
-TCAGGCCCGTGGTTCTCA-3
0
FW 5
0
-CAGAAAGCGGAAGAGAAGTCAGA-3
0
IFN-γ
72
[52]
RV 5
0
-TGCAGGCAGGAGGACCAT-3
0
FW 5
0
-GGCTCCCATGATTGTGGTAGTT-3
0
IL-6 64
[53]
RV 5
0
-GCCCAGTGGACAGGTTTCTG-3
0
Vaccines 2021, 9, x 3 of 17
enterotoxin-based vaccine to protect against acute S. aureus mastitis. While the vaccine did
not prevent bacterial shedding after challenge, results indicate that IsdA + ClfA-CTA
2/B
induces antigen-specific immune responses that may contribute to a reduction in clinical
severity and infiltration of leukocytes, or SCC, in infected animals.
2. Materials and Methods
2.1. Bacterial Strains, Plasmids, and Growth Conditions
S. aureus Newbould 305 was used for the cloning of isdA and clfA to construct IsdA +
ClfA-CTA
2/B and was also used for bacterial challenge [22,50]. E. coli ClearColi
®
(Lucigen,
Madison, WI, USA) was used for protein expression (Table 1). The vector pARLDR19 ex-
pressing CTA
2/B and containing a multiple cloning site was used to construct the plasmids
pLR001 for Isd-CTA
2/B expression and pLR003 for ClfA-CTA2/B expression (Figure 1A)
as described previously [51]. For bacterial challenge, S. aureus Newbould 305 was pre-
pared as described [10]. Briefly, Newbould 305 was grown at 37 °C with shaking to mid-
log phase in brain–heart infusion and harvested by centrifugation at 3000× g for 15 min at
4 °C. The cell pellet was washed with phosphate-buffered saline (1X PBS, pH 7.2) and
adjusted to an optical density (O.D.) of 0.2 at 600 nm. Serial dilutions were performed in
1X PBS to reach a bacterial concentration of 400 CFU/mL, as determined by plating on
blood agar (BA).
Table 1. Bacterial strains, plasmids, and primers used in this study.
Bacterial Strains Genotype or Characteristics Source
E. coli ClearColi
®
BL21(DE3) Lucigen, Madison, WI
S. aureus Newbould 305 Bovine clinical isolate [50]
Plasmids Gene Vector Source
pLR001 isdA (Newbould) pARLDR19 [49]
pLR003 clfA (Newbould) pARLDR19 [49]
Bovine Cytokine qPCR Primers Gene Amplicon Source
FW 5-GCATCGTGGAGGGACTTATGA-3
GAPDH 67 [52]
RV 5-GGGCCATCCACAGTCTTCTG-3
FW 5-CTTGTCGGAAATGATCCAGTTTT-3
IL-10 66 [53]
RV 5-TCAGGCCCGTGGTTCTCA-3
FW 5-CAGAAAGCGGAAGAGAAGTCAGA-3
IFN-γ 72 [52]
RV 5-TGCAGGCAGGAGGACCAT-3
FW 5-GGCTCCCATGATTGTGGTAGTT-3
IL-6 64 [53]
RV 5-GCCCAGTGGACAGGTTTCTG-3
Figure 1. S. aureuscholera toxin A2/B (CTA2/B) chimeric mucosal vaccines. (A) pLR001 for expres-
sion of IsdA-CTA2/B, and pLR003 for expression of ClfA-CTA2/B, and (B) SDS-PAGE of purified
IsdA-CTA
2/B (1, IsdA-CTA2~38 kD, CTB~11 kD) and ClfA-CTA2/B (2, ClfA-CTA2~37 kD, CTB~11
kD).
Figure 1.
S. aureus cholera toxin A
2
/B (CTA
2
/B) chimeric mucosal vaccines. (
A
) pLR001 for expression
of IsdA-CTA
2
/B, and pLR003 for expression of ClfA-CTA
2
/B, and (
B
) SDS-PAGE of purified IsdA-
CTA
2
/B (1, IsdA-CTA
2
~38 kD, CTB~11 kD) and ClfA-CTA
2
/B (2, ClfA-CTA
2
~37 kD, CTB~11 kD).
2.2. Protein Expression and Purification
Chimeras were purified as previously described [
49
,
51
]. Briefly, to express IsdA-
CTA
2
/B and ClfA-CTA
2
/B, ClearColi
®
with pLR001 or pLR003 were grown at 37
C
to an O.D. of 0.9 at 600 nm and induced for 24 h with 0.2% L-arabinose. Proteins were
isolated from the periplasmic extract with 1 mg/mL polymyxin B and purified by affinity
Vaccines 2021, 9, 6 4 of 17
chromatography on immobilized D-galactose (Pierce
D-Galactose Agarose, Thermo
Fisher, Waltham, MA, USA). Vaccine proteins were dialyzed into sterile 5% glycerol +
1X PBS and concentrations were determined by bicinchoninic acid assay (BCA) (Pierce
BCA, Thermo Fisher, Waltham, MA, USA). Sizes and purities of the vaccine chimeras
were confirmed by sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-
PAGE) prior to mixing at a final protein concentration of 600
µ
g/5 mL for vaccination
(Figure 1B). Vaccines were tested to ensure endotoxin levels were below 0.05 EU/mL (LAL
Endpoint Chromogenic, Lonza, Allendale, NJ, USA), plated for sterility on tryptic soy agar,
and stored at 80
C until use.
2.3. Animals, Vaccination, Challenge, and Clinical Assessment
All animal protocols were pre-approved by the University of Idaho Animal Care
and Use Committee. Lactating healthy Holstein cows in the third or fourth lactation
were pre-screened for inclusion as being those with two consecutive SCC readings below
200
×
10
3
cells/mL and no clinical evidence of mastitis. Further enrollment criteria were
followed as described previously [
49
] and included: (1) no growth of S. aureus culture from
milk as determined by plating on mannitol salt agar (MSA) and PCR with
S. aureus nuc
and isdA primers, (2) low baseline anti-IsdA responses as determined by enzyme-linked
immunosorbent assay (ELISA) of milk and serum, and (3) no evidence of bovine leukemia
virus infection (Washington Animal Disease Diagnostic Lab, WADDL, Pullman, WA, USA).
Seven selected cows were ultimately randomized into vaccinated and control groups.
Figure 2 shows the summary of trial design. Four vaccinated animals received a 600
µ
g
intranasal dose of IsdA + ClfA-CTA
2
/B in 1X PBS + 5% glycerol on days 1 and 14 (or-
ange arrows, blue bar), and a control group of three animals of similar age and lactation
period received vehicle control (1X PBS + 5% glycerol) mock vaccination on days 1 and 14
(orange arrows, grey bar). The vaccine was delivered in 2.5 mL volumes into each nare
using a nasal cannula (Merck & Co., Kenilworth, NJ, USA). On day 20, all animals were
challenged in two quarters with 400 CFU in 1 mL of S. aureus Newbould 305 (yellow arrow).
Quarters were identified as left front (LF), left rear (LR), right front (RF), and right rear (RR).
The bacterial challenge was inoculated into two diagonal quarters of each vaccinated cow
using teat cannulae (Valley Vet Supply, Marysville, KS, USA). Animals were monitored
closely during the challenge period (days 20 to 30) and evaluated for the presence of clinical
mastitis by assessment of rectal temperature (Figure 3), milk quality (Figure S2), and udder
consistency (examination for edema, hardening, and/or swelling, Table S1) on days of
milk sampling during the trial (Figure 2) [
54
,
55
]. Enrolled animals that developed pain
and/or fever that exceeded 103
F were administered painkillers (Banamine
®
and aspirin)
as recommended by the attending veterinarian. Shortly after challenge (day 21), one vacci-
nated animal developed a severe Escherichia coli mastitis case in an unchallenged quarter
(2779 LF) with systemic illness including septicemia. Thereafter, the other three quarters
were involved and the animal developed clinical mastitis due to
Staphylococcus aureus
.
The animal was euthanized on day 5 post-challenge. Results from this animal are not
included in the data in this report and resulting sample size was n = 3 per group, as repre-
sented in
Figure 2
. On day 30 all other animals began treatment with Spectramast (Zoetis,
Parsippany, NJ, USA) until consecutive negative cultures were indicative of safe release to
herd as determined by the attending veterinarian.
Vaccines 2021, 9, 6 5 of 17
Vaccines 2021, 9, x 5 of 17
Figure 2. Trial design summary. Animals (n = 3 per group, #) were vaccinated intranasally on day 1 and boosted on day
14 with 5 mL of either phosphate-buffered saline (PBS) + 5% glycerol vehicle control or 600 µg IsdA + ClfA-CTA2/B vaccine
(orange arrows). On day 20 animals were challenged once with 400 colony-forming units (CFU) of S. aureus Newbould
305 in two quarters (yellow arrow) and on day 30, animals were treated (end of challenge period, green arrow). Samples
of blood were taken on days 1, 14, 20, and 30 (X). Samples of milk were taken on days 1 and 14 (X), and every day for ten
days over the challenge period (days 20–30, XX).
Figure 2.
Trial design summary. Animals (n = 3 per group, #) were vaccinated intranasally on day 1 and boosted on day 14
with 5 mL of either phosphate-buffered saline (PBS) + 5% glycerol vehicle control or 600
µ
g IsdA + ClfA-CTA
2
/B vaccine
(orange arrows). On day 20 animals were challenged once with 400 colony-forming units (CFU) of S. aureus Newbould 305
in two quarters (yellow arrow) and on day 30, animals were treated (end of challenge period, green arrow). Samples of
blood were taken on days 1, 14, 20, and 30 (X). Samples of milk were taken on days 1 and 14 (X), and every day for ten days
over the challenge period (days 20–30, XX).
2.4. Sample Collection and Milk Culture
Blood and milk were sampled on day
2 for screening and then on days 1, 14,
20, and 30, and milk was sampled twice daily during the challenge period (Figure 2).
Blood was collected from the tail vein and allowed to coagulate at room temperature (RT)
for 1 h prior to centrifugation and resuspension into 1:10 inhibitor solution (IS, 1X HALT
protease inhibitor and 5% glycerol in 1X PBS). On day 20, whole blood was also collected in
vacutainer tubes for peripheral blood mononuclear cell (PBMC) isolation (Becton Dickinson,
Franklin Lakes, NJ, USA). Milk was collected aseptically as 50 mL quarter samples after
washing teat ends with 70% ethanol and was aliquoted into three equal tubes for culture,
SCC, and ELISA. For SCC, milk was fixed prior to shipping and analysis was performed
using the California Mastitis Test (WADDL, Pullman, WA, USA). For ELISA, milk was
centrifuged at 700
×
g for 20 min at 4
C to remove fat. Skim milk was collected and
centrifuged at 20,000
×
g for 30 min at 4
C. Whey was collected and stored in 1:10 IS.
Equal volumes of diluted whey from each quarter were pooled and stored at
20
C prior
to analysis. For milk culture, 100
µ
L and 10
µ
L of tenfold serially-diluted quarter milk
was plated on MSA, BA, and MP2 agar (Udder Health Systems, Inc., Meridian, ID, USA)
to determine the number of colony-forming units per mL (CFU/mL). The presence of
larger yellow colonies with yellow zones on MSA, beta-hemolysis on BA, or small, white,
esculin-negative colonies on MP2 was considered presumptive S. aureus. These colonies
were isolated and confirmed by a positive coagulase test or a PCR test using nuc or isdA
primers [
23
]. CFU by quarter data, based upon final quantitation on MSA, was determined
once daily on days
2, 1, 14, and 20 prior to challenge and twice daily (AM/PM) during the
challenge period. Quarter data were combined and total CFU/mL by cow was reported
for six animals (n = 3 per group).
Vaccines 2021, 9, 6 6 of 17
Vaccines 2021, 9, x 6 of 17
Figure 3. Vaccination outcomes during the trial period. (A) Quantification of bacterial shedding by
cows during the challenge period. Log10 of CFU/mL of Staphylococcus aureus on mannitol salt agar
(MSA). Mean ± standard error, n = 3 per group, and analyzed using repeated measures analysis of
variance (ANOVA). No significance after false discovery rate (FDR) adjustment for multiple com-
parisons. (B) Somatic cell count (SCC) (×1000 cells/mL) by cow. Mean ± standard error, n = 3 per
group, and analyzed using repeated measures ANOVA. During the challenge period, control
cows uniformly had higher SCC than vaccinated cows (main model effect p = 0.002). (C) Rectal
temperature in degrees Fahrenheit (°F). Mean ± standard error, n = 3 per group, and analyzed us-
ing repeated measures ANOVA showing no significance between groups. Orange arrows indicate
Figure 3.
Vaccination outcomes during the trial period. (
A
) Quantification of bacterial shedding by
cows during the challenge period. Log10 of CFU/mL of Staphylococcus aureus on mannitol salt agar
(MSA). Mean
±
standard error, n = 3 per group, and analyzed using repeated measures analysis
of variance (ANOVA). No significance after false discovery rate (FDR) adjustment for multiple
comparisons. (
B
) Somatic cell count (SCC) (
×
1000 cells/mL) by cow. Mean
±
standard error, n = 3
per group, and analyzed using repeated measures ANOVA. During the challenge period, control cows
uniformly had higher SCC than vaccinated cows (main model effect p = 0.002). (
C
) Rectal temperature
in degrees Fahrenheit (
F). Mean
±
standard error, n = 3 per group, and analyzed using repeated
measures ANOVA showing no significance between groups. Orange arrows indicate day of booster
vaccination (14), yellow arrows indicate day of bacterial challenge (20) and green arrows indicates
the last day of challenge (30).
Vaccines 2021, 9, 6 7 of 17
2.5. IgG, IgG1, IgG2, and IgA Enzyme-Linked Immunosorbent Assay (ELISA)
IsdA- and ClfA-specific immune responses in serum and milk were detected using
ELISA as described [
51
]. Briefly, 96-well microtiter plates (Nunc, Thermo Fisher, Waltham,
MA, USA) were coated with 10
µ
g of either IsdA-CTA
2
/B, ClfA-CTA
2
/B, or CTA
2
/B in
1X PBS and incubated overnight at 4
C. Coated plates were blocked for 2 h at 37
C in
1% goat milk + 1X PBS. After washing, plates were incubated with two-fold dilutions of
either bovine serum (dilutions initiated at 1:200 concentration) or pooled quarter milk
(dilutions initiated at a 1:10 concentration). Plates were incubated at 4
C overnight.
After washing, plates were incubated with horseradish peroxidase (HRP)-conjugated anti-
bovine IgG, IgG1, IgG2, or IgA (1:10,000 Bethyl Laboratories, Montgomery, TX, USA)
at 37
C for 1 h. Plates were developed with tetramethylbenzidine (Promega
TM
TMB
One, Thermo Fisher, Waltham, MA, USA) and read at 370 nm per TMB manufacturer’s
instruction. ELISA results from serum or pooled quarter milk were reported by cow
(n = 3) and presented as the ratio of results (day X/day 1) of the O.D. (370 nm) from a
representative antibody dilution in the linear part of the curve (1:1600 serum, 1:160 milk).
Results are the average of three independent assays.
2.6. Peripheral Blood Mononuclear Cell (PBMC) Isolation and Cytokine qPCR
PBMCs were isolated from whole bovine blood on day 20 for cytokine analysis. PBMCs
were isolated using a density gradient established by layering whole blood diluted 1:2 with
1X PBS on Histopaque
®
-1077 (Sigma-Aldrich, St. Louis, MO, USA). Blood samples were cen-
trifuged at 800
×
g for 30 min at RT. The buffy coat was removed and washed three times by
centrifugation with Hank’s Balanced Salt Solution for 10 min at 400
×
g at RT, and cells were
counted with 0.2% trypan blue. For cytokine assays, total RNA from PBMCs from each cow
(n = 3 per group) was extracted (RNeasy, Qiagen, Germantown, MD, USA) with an addi-
tional Dnase I (Promega, Madison, WI, USA) digestion. cDNA was reverse transcribed per
manufacturer’s instructions (High-Capacity RNA-to-cDNA
Kit, Thermo Fisher, Waltham,
MA, USA). qRT-PCR was conducted using SYBR fast (Kapa Biosystems, Thermo Fisher,
Waltham, MA, USA) on interferon gamma (
IFN-γ
), interleukin-6 (IL-6), and interleukin-
10 (IL-10) primers, using bovine glyceraldehyde
3-phosphate
dehydrogenase (GAPDH)
as a reference gene (primers, Table 1). Results are presented as relative gene expression
2
Ct
[
56
]. All qRT-PCR experiments were performed in triplicate per cow PBMC sample.
2.7. Sample Size, Statistical Methods, and Analysis
Sample size was estimated prior to study by power analysis based upon predicted
SCC and CFU/mL in milk and using the assumption that quarters are independent,
as has been reported [
57
,
58
]. A sample size of 13 quarters per group was predicted
to provide, at a 95% level of confidence, 80% power to detect a difference in logged
SCC. Resulting quarter bacterial counts and SCC data from this study were analyzed
by (1) assuming independent quarters and (2) as the combined average of quarters by
cow. Outcomes were not different, thus results are reported as the average by cow and
assuming quarters are not independent. The log-base 10 values of CFU, SCC, temperature,
and serum and milk anti-IsdA, ClfA, and CTB antibodies were analyzed using repeated
measures analysis of variance (ANOVA) with time as the within-subjects variable and
group as the between-subjects variable. Within-subjects correlation was modeled with
either first-order autoregressive or compound symmetric structure, depending on Akaike’s
Information Criterion [
59
]. Comparisons of interest were identified prior to modeling
and were examined regardless of the significance of main effects or interaction. First,
we explicitly compared the outcome at each study time point. Second, we examined the
change in outcome within group, comparing days and adjusting the paired comparisons
using false discovery rate [
60
]. Cytokine analysis was performed using a two-group t-test
between vaccinated and control animals. Statistical analyses were conducted using JMP and
SAS software (Cary, NC). p-values are reported as p
0.05(*), p
0.01(**), or p
0.0001(****)
and reflect two-sided tests.
Vaccines 2021, 9, 6 8 of 17
3. Results
3.1. Bacterial Culture and Clinical Assessment
Quantification of S. aureus was determined after plating milk that had been sampled
once daily on days
2, 1, 14, and 20 prior to challenge and twice daily during the challenge
period (Figure 2). Prior to challenge no animals were found to be shedding S. aureus,
and immediately after challenge all animals shed high levels of S. aureus from infected
quarters (Figure 3A). Results revealed a rapid decline in bacterial shedding from all animals
within 24 h and then a slow decline beginning in the middle of the challenge period.
Between days 2 and 10 of the challenge period (days 22 and 30 of trial) control animals
shed a total of 1.08
×
10
6
CFU/mL and vaccinated animals shed 7.53
×
10
5
CFU/mL.
Differences between treatment groups were observed on days 21, 29, and 30 during the
challenge period (p = 0.029, 0.011, and 0.018, respectively), however, after adjusting for
multiple comparisons, these results are not significant. S. aureus was isolated from all
challenged quarters in both treatment groups, and all animals continued to shed S. aureus
throughout the trial. Animals did not shed from uninfected quarters. While one vaccinated
animal was culture negative at two time points late in the challenge period (day 29 AM and
day 30 AM), no animals were consistently sterile of S. aureus by the end of the challenge
period. Analysis of positive quarters indicated that there were more days showing a lower
percentage of infected quarters for vaccinated animals (Supplementary Figure S1).
SCC taken once daily before the challenge period and twice daily during challenge
is shown in Figure 3B. Results show a consistently reduced SCC from vaccinated ani-
mals beginning 48 h post-challenge (day 22). While individual days were not significant
after adjustment, across and after the challenge period (days 21 to 39) unvaccinated an-
imals had significantly-higher SCC than vaccinated animals (model main effect of treat-
ment group, p = 0.002). SCCs of individual cows throughout the trial are shown in
Supplementary Figure S3B.
The average rectal temperature per group during the challenge period is shown
in
Figure 3C
. Temperatures at 72 h post-challenge (day 23 AM) showed an average of
102.7
F for control and 101.5
F for vaccinated animals, however there was no statistically-
significant difference in temperature between groups on any day during this period. In ad-
dition, no differences in temperature between groups occurred within 24 h after vaccination
(days 1 and 14).
Clinical assessments indicated that animals did not show signs of systemic illness,
loss of appetite, or adverse local reactions due to the vaccine, and no animals had clinical
evidence of mastitis prior to challenge (day 20). Clinical results are summarized in Supple-
mentary Table S1. Clinical mastitis due to S. aureus was observed in challenged quarters of
control cows 2767 (LF and RF) and 2830 (LR) throughout the evaluation period. The latter
cow developed a persistent mastitis starting on day 23 with apparent milk changes that
included clots and flakes in the LR quarter. Clinical mastitis in this animal included persis-
tent udder swelling in addition to pain, heat, and sensation of the affected teat until the end
of the challenge period. Temporary enlargement of the supramammary lymph node was
noted in one of the vaccinated cows (2823) on day 24, and persistent enlargement observed
in one control animal (2830).
Milk quality assessments indicated that while the fat, protein, lactose, and solids-not-
fat (SNF) percentages were frequently higher in vaccinated animals, these differences were
not statistically significant (Supplementary Figure S2).
3.2. Vaccine-Specific Antibody Responses in Blood and Milk
Antigen-specific humoral responses were quantified by ELISA from blood and milk.
Anti-IgG responses in serum on days 14, 20 and 30, relative to day 1, are shown in
Figure 4A,C and E
. Vaccinated animals (blue bars) showed a significant IsdA-CTA
2
/B-
specific IgG response in serum after challenge on day 30 relative to days 14 and
20 (p
adj
= 0.008
for both) and on day 30 relative to control animals (p = 0.030 *) (
Figure 4A
). Vaccinated ani-
mals showed a similar, but non-significant, anti-ClfA-CTA
2
/B-specific IgG responses in
Vaccines 2021, 9, 6 9 of 17
serum on day 30 relative to days 14 and 20 (p
adj
= 0.120) as well as on day 30 relative
to control animals (p = 0.079) (Figure 4C). Anti-CTA
2
/B-specific IgG responses in serum
remained low and non-significant between groups throughout and after challenge (day
30) (Figure 4E).
Vaccines 2021, 9, x 10 of 17
Figure 4. Immunoglobulin G (IgG) antibody responses in serum and milk as determined by en-
zyme-linked immunosorbent assay (ELISA). Anti-IsdA-CTA
2/B IgG responses in (A) serum and
(B) milk, anti-ClfA-CTA2/B IgG responses in (C) serum and (D) milk, and anti-CTA2/B IgG re-
sponses in (E) serum and (F) milk. Serum was analyzed on days 14, 20, and 30 and milk on days
14, 20, 22, 24, 26, 28, and 30 during the trial period. Results are reported as ELISA ratios of day
X/day 1 at O.D. 370 at serum dilutions of 1:1600 and milk dilutions of 1:160. Shown are mean and
standard error by treatment with control (gray) and vaccinated (blue) (n = 3 per group). Significant
differences between groups are represented as p 0.05 (*). The log10 of the values were analyzed
using repeated measures analysis of variance (ANOVA) with a compound symmetric covariance
structure for cows across days. Model-based estimates were compared between groups within
days and adjusted for multiple comparisons.
Anti-IgG responses in milk on days 14, 22, 24, 26, 28, and 30, relative to day 1, are
shown in Figure 4D–F. IsdA-CTA
2/B-specific IgG responses in milk increased over the
challenge period in vaccinated animals, with values significantly higher on day 30 relative
to days 20 to 26 (adjusted p-values all <0.05) and on day 30 relative to control cows (p =
0.030 *) (Figure 4D). The anti-IsdA-CTA
2/B differences between days for control cows
were non-significant after day 20. Anti-ClfA-CTA
2/B-specific IgG responses in milk were
significant on day 30 relative to days 20–24 (adjusted p-values all <0.05) for the vaccinated
group and on day 30 relative to unvaccinated cows (p = 0.043 *) (Figure 4E). Milk anti-
CTA
2/B-specific IgG responses increased moderately during the challenge period in both
vaccinated and control animals with significant increases on day 30 relative to days 14
Figure 4.
Immunoglobulin G (IgG) antibody responses in serum and milk as determined by enzyme-
linked immunosorbent assay (ELISA). Anti-IsdA-CTA
2
/B IgG responses in (
A
) serum and (
B
) milk,
anti-ClfA-CTA
2
/B IgG responses in (
C
) serum and (
D
) milk, and anti-CTA
2
/B IgG responses in (
E
)
serum and (
F
) milk. Serum was analyzed on days 14, 20, and 30 and milk on days 14, 20, 22, 24,
26, 28, and 30 during the trial period. Results are reported as ELISA ratios of day X/day 1 at O.D.
370 at serum dilutions of 1:1600 and milk dilutions of 1:160. Shown are mean and standard error
by treatment with control (gray) and vaccinated (blue) (n = 3 per group). Significant differences
between groups are represented as p
0.05 (*). The log10 of the values were analyzed using repeated
measures analysis of variance (ANOVA) with a compound symmetric covariance structure for cows
across days. Model-based estimates were compared between groups within days and adjusted for
multiple comparisons.
Anti-IgG responses in milk on days 14, 22, 24, 26, 28, and 30, relative to day 1,
are shown in Figure 4B, D and F. IsdA-CTA
2
/B-specific IgG responses in milk increased
over the challenge period in vaccinated animals, with values significantly higher on day 30
relative to days 20 to 26 (adjusted p-values all <0.05) and on day 30 relative to control cows
(p = 0.030 *) (Figure 4B). The anti-IsdA-CTA
2
/B differences between days for control cows
Vaccines 2021, 9, 6 10 of 17
were non-significant after day 20. Anti-ClfA-CTA
2
/B-specific IgG responses in milk were
significant on day 30 relative to days 20–24 (adjusted p-values all <0.05) for the vaccinated
group and on day 30 relative to unvaccinated cows (p = 0.043 *) (Figure 4D). Milk anti-
CTA
2
/B-specific IgG responses increased moderately during the challenge period in both
vaccinated and control animals with significant increases on day 30 relative to days 14
and 20 in the vaccinated group and no change in the control group (adjusted p-values all
<0.05). The differences in anti-CTA
2
B responses between vaccine and control groups were
non-significant on all days tested (Figure 4F).
Serum IgG subtype (IgG1 and IgG2) responses were evaluated to further define
the T helper immune response (Figure 5A–D). Vaccinated animals exhibited increases
in IgG1 and IgG2 responses on day 30 for both the IsdA- and ClfA-CTA
2
/B antigens.
The serum anti-IsdA-CTA
2
/B IgG1 response on day 30 relative to days 14 and 20 was
significant for vaccinated animals (blue bars, p
adj
= 0.004 and p
adj
= 0.007, respectively),
and the difference between groups was significant on day 30 (p = 0.033 *) (Figure 5A).
For anti-IsdA-CTA
2
/B IgG2 responses, day 30 was higher than days 14 and 20 for both
vaccinated and control groups (p
adj
= 0.045 for both comparisons) with no significant
differences between groups on day 30 (Figure 5B). For serum anti-ClfA-CTA
2
/B IgG1,
vaccinated animals showed an increase on day 30 compared to day 14 (p
adj
= 0.023) and day
20 (p
adj
= 0.029), and the difference between groups was significant on day 30 (p = 0.029 *)
(
Figure 5C
). For anti-ClfA-CTA
2
/B IgG2 responses, day 30 was higher than days 14 and 20
for both groups as well (p
adj
= 0.015 for both comparisons), however the difference between
vaccinated and control groups on day 30 was non-significant after adjustment (p = 0.050)
(
Figure 5D
). Assessment of milk anti-IsdA-CTA
2
/B and anti-ClfA-CTA
2
/B IgG1, IgG2 and
IgA responses was also performed, and while results indicated an increase on day 30 for
both IgG1 and IgA, they were non-significant between vaccine and control groups on the
days (14, 20, and 30) tested (data not shown).
Combined, ELISA analysis shows an induction of antigen-specific humoral responses
in the milk and serum after intranasal IsdA + ClfA-CTA
2
/B vaccination, as evidenced by
a significant booster effect upon bacterial challenge. Antibody subtyping indicated that
both antigens stimulated a Th2-type response, with ClfA potentially inducing a mixed
Th1/Th2 response. Lastly, there was no significant antibody response to the CTA
2
/B
adjuvant vector alone.
3.3. Cytokine Assay
The stimulation of cellular cytokine responses was assessed by quantitative RT-PCR
using PBMCs isolated from vaccinated and control cows on day 20 (Figure 5E). IL-12,
TNF-
α
, and IL-4 levels were not significantly different between vaccinated and control
animals (data not shown). Vaccinated cows showed a slight but significant increase in
IFN-
γ
expression (p = 0.048 *) but no significant difference in IL-10 or IL-6 expression
(Figure 5E).
Vaccines 2021, 9, 6 11 of 17
Vaccines 2021, 9, x 11 of 17
and 20 in the vaccinated group and no change in the control group (adjusted p-values all
<0.05). The differences in anti-CTA
2B responses between vaccine and control groups were
non-significant on all days tested (Figure 4F).
Serum IgG subtype (IgG1 and IgG2) responses were evaluated to further define the
T helper immune response (Figure 5A–D). Vaccinated animals exhibited increases in IgG1
and IgG2 responses on day 30 for both the IsdA- and ClfA-CTA
2/B antigens. The serum
anti-IsdA-CTA
2/B IgG1 response on day 30 relative to days 14 and 20 was significant for
vaccinated animals (blue bars, p
adj = 0.004 and padj = 0.007, respectively), and the difference
between groups was significant on day 30 (p = 0.033 *) (Figure 5A). For anti-IsdA-CTA
2/B
IgG2 responses, day 30 was higher than days 14 and 20 for both vaccinated and control
groups (p
adj = 0.045 for both comparisons) with no significant differences between groups
on day 30 (Figure 5B). For serum anti-ClfA-CTA
2/B IgG1, vaccinated animals showed an
increase on day 30 compared to day 14 (p
adj = 0.023) and day 20 (padj = 0.029), and the dif-
ference between groups was significant on day 30 (p = 0.029 *) (Figure 5C). For anti-ClfA-
CTA
2/B IgG2 responses, day 30 was higher than days 14 and 20 for both groups as well
(p
adj = 0.015 for both comparisons), however the difference between vaccinated and control
groups on day 30 was non-significant after adjustment (p = 0.050) (Figure 5D). Assessment
of milk anti-IsdA-CTA
2/B and anti-ClfA-CTA2/B IgG1, IgG2 and IgA responses was also
performed, and while results indicated an increase on day 30 for both IgG1 and IgA, they
were non-significant between vaccine and control groups on the days (14, 20, and 30)
tested (data not shown).
Figure 5. Serum IgG1, IgG2, and cytokine expression analysis. (A) Anti-IsdA-CTA2/B IgG1, (B)
anti-IsdA-CTA2/B IgG2, (C) anti-ClfA-CTA2/B IgG1, and (D) anti-ClfA-CTA2/B IgG2 responses in
serum on days 14, 20, and 30. Results are reported as ELISA ratios of day X/day 1 at O.D. 370 at
serum dilutions of 1:1600. Shown are mean and standard error by treatment with control (gray)
and vaccinated (blue) (n = 3 per group). The log10 of the values were analyzed using analysis of
variance (ANOVA), with a compound symmetric covariance structure for cows across days.
Model-based estimates were compared between groups within days and adjusted for multiple
comparisons. (E) IL-10, IL-6, and IFN-γ expression as determined by quantitative RT-PCR of pe-
ripheral blood mononuclear cells (PBMCs) isolated from whole blood after boost on day 20. Re-
sults are shown as relative gene expression to GAPDH (2
−ΔΔCt
). Data are presented as mean and
standard error of control (gray) and vaccinated (blue) showing median and range (n = 3 per
Figure 5.
Serum IgG1, IgG2, and cytokine expression analysis. (
A
) Anti-IsdA-CTA
2
/B IgG1, (
B
) anti-IsdA-CTA
2
/B IgG2,
(
C
) anti-ClfA-CTA
2
/B IgG1, and (
D
) anti-ClfA-CTA
2
/B IgG2 responses in serum on days 14, 20, and 30. Results are reported
as ELISA ratios of day X/day 1 at O.D. 370 at serum dilutions of 1:1600. Shown are mean and standard error by treatment
with control (gray) and vaccinated (blue) (n = 3 per group). The log10 of the values were analyzed using analysis of variance
(ANOVA), with a compound symmetric covariance structure for cows across days. Model-based estimates were compared
between groups within days and adjusted for multiple comparisons. (
E
) IL-10, IL-6, and IFN-
γ
expression as determined
by quantitative RT-PCR of peripheral blood mononuclear cells (PBMCs) isolated from whole blood after boost on day 20.
Results are shown as relative gene expression to GAPDH (2
Ct
). Data are presented as mean and standard error of
control (gray) and vaccinated (blue) showing median and range (n = 3 per group). Data were analyzed using a two-group
t-test between vaccinated and control. Significant differences between groups are represented as p 0.05 (*).
4. Discussion
This report describes the outcomes of a small bovine challenge trial to assess the effi-
cacy of the IsdA + ClfA-CTA
2
/B mucosal S. aureus mastitis vaccine. We hypothesized that
vaccination would prevent or reduce bacterial shedding from the udder after intramam-
mary challenge and reduce disease outcomes. Animals were vaccinated intranasally during
milking and challenged in two quarters with the homologous S. aureus Newbould 305
vaccine strain. An averaged reduction in CFU/mL from combined quarters of vaccinated
compared to unvaccinated animals was observed beginning 24 h after challenge to the end
of the challenge period, however, this difference was not significant on specific days during
the challenge period. Analysis of bacteriology using independent quarters did not change
data interpretations, however, a lower percentage of infected quarters was observed on
multiple days after challenge. Analysis of SCC revealed that vaccinated animals had lower
numbers of cells on the majority of days during the challenge period of the trial, and this
decrease was significant between vaccinated and control animals during the whole of the
period. These results were also consistent with the evidence of reduced clinical mastitis in
vaccinated animals.
The assessment of humoral immune responses in milk and serum in this report showed
induction of IsdA- and ClfA-CTA
2
/B specific IgG antibodies in vaccinated animals after
S. aureus
challenge indicating that vaccination induced antigen-specific responses that were
amplified by bacterial challenge. In contrast to previous studies, no significant increase in
Vaccines 2021, 9, 6 12 of 17
antigen-specific humoral responses was detected in the serum directly after vaccination
and boost, despite the same vaccine dose and schedule [
49
].The lower sample size in this
trial compared to previous trials with IsdA + ClfA-CTA
2
/B may have contributed to this
outcome, and larger trials will be essential to advance this vaccine candidate. In addition,
animals were vaccinated during milking for this study instead of during dry-off, which is
a period of higher susceptibility to mastitis and changes in immune function that may
explain observed differences in immunogenicity. As with previous trials, antibody analysis
revealed that not all vaccinated animals responded well to the same vaccine preparation
and dosage. Variations in host genetics or inconsistencies in administration can cause
these disparities, and larger trials will help to exclude them. Other vaccination routes,
or alternate prime-boost strategies, may also promote vaccination consistency and efficacy.
These routes were not explored in this early study to enable a narrow focus on mucosal
delivery, but intramuscular, subcutaneous, and transdermal routes are all effective for
CT-adjuvanted vaccines and could be explored. Lastly, in this study we maintained a short
dosage interval of only 14 days to align with previous trials, however, a longer interval
between doses may improve responses and will be explored in the future.
Animals were vaccinated during milking to permit bacterial quantification and limit
the potential for systemic or chronic infection. Despite this, one vaccinated animal was
euthanized shortly after challenge due to an E. coli infection that rapidly became systemic.
While little has been reported about the effects of co-infection on the severity of E. coli
mastitis, the cow immune status is a key factor, and S. aureus is known for the production
of virulence factors that modulate the immune response [
61
]. Specifically the
S. aureus
superantigens (SAgs) can activate specific T-cell subsets, resulting in inflammation, tis-
sue damage, and potential T-cell anergy [
62
64
]. S. aureus Newbould 305 strain was
chosen for these studies because it induces mild and chronic mastitis, has been utilized
before in vaccine challenge trials, and contains a limited repertoire of SAgs [
50
,
65
,
66
]. It is
recognized, however, that immune dysregulation likely occurred upon challenge and,
despite vaccination, contributed to the enhanced spread and systemic infection in this
animal. The potential for co-infection and the ability of the vaccine to protect against
heterologous S. aureus isolates that may induce more severe disease will both need to be
addressed in future studies.
As described above, CT and its non-toxic B subunit can induce humoral and cellu-
lar immune responses to co-delivered antigens. CTA
2
/B molecules retain much of the
well-characterized adjuvanticity of CTB to induce both humoral and cellular responses.
The IgG1 and IgG2 profiles we observed in the serum of vaccinated animals on day 30 were
consistent with our previous studies indicating that CTA
2
/B chimeras promote a largely
Th2-type cellular response [
49
,
51
]. In the current study, however, the responses to IsdA
were more clearly polarized toward Th2, while the anti-ClfA responses are supportive of
a potential mixed Th1/Th2 response. Cytokine expression analysis in the current study,
performed on day 20 prior to challenge, showed no effect on IL-10 and IL-6, but an increase
in IFN-
γ
in vaccinated animals. Cytokine analysis from previous immunogenicity studies
using the IsdA + ClfA-CTA
2
/B vaccine largely supported a Th2-type response and did not
indicate IFN-
γ
upregulation [
49
,
51
]. This apparent contradiction may be due to differences
in the timing of analysis (6 days after vaccination in the current study versus 45 days after
vaccination in previous studies) and the methods used (unstimulated versus stimulated
PBMCs). Reports indicate that while CTB more commonly induces Th2-type responses,
it can induce a mixed Th2/Th1 response with enhanced IFN-
γ
secretion, depending upon
the antigen and route of delivery [
42
,
67
71
]. Similar to CTB, vaccination with CTA
2
/B
chimeras may promote early macrophage or dendritic cell activation and antigen presen-
tation through IFN-
γ
upregulation. In this study there was not a clear early effect on the
inflammatory and pro-inflammatory balance of serum IL-6 and IL-10, however, others have
reported anti-inflammatory properties in CT and its derivatives. These properties may be
advantageous for the prevention of S. aureus udder colonization and are consistent with
our observed reduction in SCC after challenge [7274].
Vaccines 2021, 9, 6 13 of 17
Lastly, in this study we determined if animals responded to the vaccine adjuvant alone
by producing anti-CTA
2
/B humoral responses. Results showed no significant differences
between vaccinated and control groups. While S. aureus challenge was not expected
to induce anti-CT antibodies, previous studies have reported the undesirable effect of
significant anti-CT antibodies after use of this adjuvant for mucosal vaccination [
75
].
The low adjuvant-specific antibody response observed here, combined with the reduced
recruitment of somatic cells, provides support for the utility of CTA
2
/B-based vaccines.
These studies indicate that IsdA + ClfA-CTA
2
/B may be effective in the reduction
of
S. aureus
colonization and clinical outcome, as evidenced by reduced SCC, but do not
provide evidence of complete protection or elimination. Both vaccinated and unvacci-
nated animals shed high levels of S. aureus Newbould 305 immediately after challenge,
and all animals in the study were found to shed the challenge strain during the entire
10-day
challenge period. This outcome may be the result of a high bacterial dose and
the artificial nature of intramammary challenge. The use of a lower challenge dose, a dif-
ferent method of challenge, and/or focus on natural transmission in a larger field trial
will better determine efficacy to prevent infection. In addition, studies are needed that
utilize heterologous isolates, compare outcomes with current vaccines, and assess alternate
routes of immunization. IsdA and ClfA are established and highly-conserved antigens
from bovine S. aureus, however, the incorporation of additional antigens, including toxins
and anti-immune factors, may also be necessary to promote strain cross-protection and
control immune modulation.
5. Conclusions
Results indicate vaccine efficacy in reducing SCC and improving clinical outcome and
support further exploration of the IsdA + ClfA-CTA
2
/B vaccine to prevent bovine mastitis.
The development of an effective vaccine to prevent mastitis caused by S. aureus would have
many positive impacts on animal health and food production and may decrease overall
antibiotic use in the industry. Needle-free vaccination of cattle would also be beneficial by
reducing the transmission of disease, inducing mucosal immunity, and promoting vaccine
distribution and use. This study provides important preliminary results of a cholera-
toxin-based intranasal vaccine in a mastitis challenge model and supports the continued
exploration of this antigen-adjuvant platform to prevent bovine disease.
Supplementary Materials:
The following are available online at https://www.mdpi.com/2076-3
93X/9/1/6/s1, Figure S1: Percent S. aureus Newbould 305 infected quarters from vaccinated and
control groups during the challenge period, Figure S2: Milk quality assessment during the trial period,
Figure S3: Individual cow bacterial shedding and SCC during trial, Table S1: Clinical outcomes after
challenge on day 20.
Author Contributions:
All authors were blinded throughout the trial with the exception of J.K.T.
H.A.A. performed trial coordination at the University of Idaho as well as vaccination, sampling,
and cow monitoring. D.S. performed whole-blood PBMC extraction and cytokine analysis as well
as milk DNA qPCR. E.O. performed milk and serum ELISA. J.E.W. performed sample processing,
trial coordination, and shipments. O.M.M. purified and quality tested vaccines. G.D. processed milk
and blood samples and performed colony counts at Boise State University. L.B. aided with study
design and post-trial statistical analysis. M.A.M. was co-lead investigator at the University of Idaho.
J.K.T. was co-lead investigator at Boise State. All authors have read and agreed to the published
version of the manuscript.
Funding:
This research was funded by a 2013 USDA AFRI standard grant (#2013-01189, PI-Tinker,
Co-PI McGuire
), and a faculty seed grant to J.K.T. from an Institutional Development Award (IDeA)
from the National Institute of General Medical Sciences of the National Institutes of Health
(#P20GM103408 and P20GM109095). We also acknowledge support from The Biomolecular Research
Center at Boise State with funding from the National Science Foundation, Grants # 0619793 and
#0923535, the MJ Murdock Charitable Trust, and the Idaho State Board of Education.
Vaccines 2021, 9, 6 14 of 17
Acknowledgments:
We would like to thank Neha Misra, Brian Mitchell, Laura Rogers, Jim Schroeder,
Edgar Ayala Tapia, and Aurora Thomson-Vogel, for technical support and discussion, as well as
Michael Jobling and Randall Holmes for kind donation of the pARLDR19 vector.
Conflicts of Interest:
J.K.T. holds an unlicensed patent for the use of cholera toxin chimera as a
staphylococcal vaccine (Tinker, U.S. Pat. No. 8,834,898).
References
1.
Heikkila, A.M.; Liski, E.; Pyorala, S.; Taponen, S. Pathogen-specific production losses in bovine mastitis. J. Dairy Sci
2018
,
101, 9493–9504. [CrossRef]
2.
APHIS, U. Dairy 2014. Milk Quality, Milking Procedures, and Mastitis on U.S. Dairies; United States Department of Agriculture:
Washington, DC, USA, 2014; Volume 2016.
3.
Roberson, J.R.; Fox, L.K.; Hancock, D.D.; Gay, J.M.; Besser, T.E. Sources of intramammary infections from Staphylococcus aureus in
dairy heifers at first parturition. J. Dairy Sci. 1998, 81, 687–693. [CrossRef]
4.
Hebert, A.; Sayasith, K.; Senechal, S.; Dubreuil, P.; Lagace, J. Demonstration of intracellular Staphylococcus aureus in bovine mastitis
alveolar cells and macrophages isolated from naturally infected cow milk. FEMS Microbiol. Lett. 2000, 193, 57–62. [CrossRef]
5.
Sacco, S.C.; Velazquez, N.S.; Renna, M.S.; Beccaria, C.; Baravalle, C.; Pereyra, E.A.L.; Monecke, S.; Calvinho, L.F.; Dallard, B.E.
Capacity of two Staphylococcus aureus strains with different adaptation genotypes to persist and induce damage in bovine
mammary epithelial cells and to activate macrophages. Microb. Pathog. 2020, 142, 104017. [CrossRef] [PubMed]
6.
Zaatout, N.; Ayachi, A.; Kecha, M. Staphylococcus aureus persistence properties associated with bovine mastitis and alternative
therapeutic modalities. J. Appl. Microbiol. 2020. [CrossRef]
7.
Sol, J.; Sampimon, O.C.; Snoep, J.J.; Schukken, Y.H. Factors associated with bacteriological cure during lactation after therapy for
subclinical mastitis caused by Staphylococcus aureus. J. Dairy Sci. 1997, 80, 2803–2808. [CrossRef]
8.
Rollin, E.; Dhuyvetter, K.C.; Overton, M.W. The cost of clinical mastitis in the first 30 days of lactation: An economic modeling
tool. Prev. Vet. Med. 2015, 122, 257–264. [CrossRef]
9.
Cha, E.; Bar, D.; Hertl, J.A.; Tauer, L.W.; Bennett, G.; Gonzalez, R.N.; Schukken, Y.H.; Welcome, F.L.; Grohn, Y.T. The cost
and management of different types of clinical mastitis in dairy cows estimated by dynamic programming. J. Dairy Sci.
2011
,
94, 4476–4487. [CrossRef]
10.
Middleton, J.R.; Ma, J.; Rinehart, C.L.; Taylor, V.N.; Luby, C.D.; Steevens, B.J. Efficacy of different Lysigin formulations in the
prevention of Staphylococcus aureus intramammary infection in dairy heifers. J. Dairy Res. 2006, 73, 10–19. [CrossRef]
11.
Schukken, Y.H.; Bronzo, V.; Locatelli, C.; Pollera, C.; Rota, N.; Casula, A.; Testa, F.; Scaccabarozzi, L.; March, R.; Zalduendo, D.; et al.
Efficacy of vaccination on Staphylococcus aureus and coagulase-negative staphylococci intramammary infection dynamics in
2 dairy herds. J. Dairy Sci. 2014, 97, 5250–5264. [CrossRef]
12.
Bradley, A.J.; Breen, J.E.; Payne, B.; White, V.; Green, M.J. An investigation of the efficacy of a polyvalent mastitis vaccine using
different vaccination regimens under field conditions in the United Kingdom. J. Dairy Sci.
2015
, 98, 1706–1720. [CrossRef] [PubMed]
13.
Piepers, S.; Prenafeta, A.; Verbeke, J.; De Visscher, A.; March, R.; De Vliegher, S. Immune response after an experimental
intramammary challenge with killed Staphylococcus aureus in cows and heifers vaccinated and not vaccinated with Startvac,
a polyvalent mastitis vaccine. J. Dairy Sci. 2017, 100, 769–782. [CrossRef] [PubMed]
14.
Landin, H.; Mork, M.J.; Larsson, M.; Waller, K.P. Vaccination against Staphylococcus aureus mastitis in two Swedish dairy herds.
Acta Vet. Scand. 2015, 57, 81. [CrossRef] [PubMed]
15.
Freick, M.; Frank, Y.; Steinert, K.; Hamedy, A.; Passarge, O.; Sobiraj, A. Mastitis vaccination using a commercial polyvalent
vaccine or a herd-specific Staphylococcus aureus vaccine. Results of a controlled field trial on a dairy farm. Tierarztl Prax Ausg G
Grosstiere Nutztiere 2016, 44, 219–229. [CrossRef]
16.
Kim, H.K.; DeDent, A.; Cheng, A.G.; McAdow, M.; Bagnoli, F.; Missiakas, D.M.; Schneewind, O. IsdA and IsdB antibodies protect
mice against Staphylococcus aureus abscess formation and lethal challenge. Vaccine 2010, 28, 6382–6392. [CrossRef]
17.
Clarke, S.R.; Andre, G.; Walsh, E.J.; Dufrene, Y.F.; Foster, T.J.; Foster, S.J. Iron-regulated surface determinant protein A mediates
adhesion of Staphylococcus aureus to human corneocyte envelope proteins. Infect. Immun. 2009, 77, 2408–2416. [CrossRef]
18.
Stapleton, M.; Wright, L.; Clarke, S.; Moseby, H.; Tarkowski, A.; Vendrengh, M.; Foster, S. Identification of Conserved Antigens
from Staphylococcal and Streptococcal Pathogens. J. Med. Microbiol. 2012. [CrossRef]
19.
Clarke, S.R.; Brummell, K.J.; Horsburgh, M.J.; McDowell, P.W.; Mohamad, S.A.; Stapleton, M.R.; Acevedo, J.; Read, R.C.; Day, N.P.;
Peacock, S.J.; et al. Identification of
in vivo
-expressed antigens of Staphylococcus aureus and their use in vaccinations for protection
against nasal carriage. J. Infect. Dis. 2006, 193, 1098–1108. [CrossRef]
20.
Wolf, C.; Kusch, H.; Monecke, S.; Albrecht, D.; Holtfreter, S.; von Eiff, C.; Petzl, W.; Rainard, P.; Broker, B.M.;
Engelmann, S
.
Genomic and proteomic characterization of Staphylococcus aureus mastitis isolates of bovine origin. Proteomics
2011
,
11, 2491–2502. [CrossRef]
21.
Herron-Olson, L.; Fitzgerald, J.R.; Musser, J.M.; Kapur, V. Molecular Correlates of Host Specialization in Staphylococcus aureus.
PLoS ONE 2007, 2, e1120. [CrossRef]
22.
Bouchard, D.; Peton, V.; Almeida, S.; Le Maréchal, C.; Miyoshi, A.; Azevedo, V.; Berkova, N.; Rault, L.; François, P.;
Schrenzel, J.; et al
. Genome sequence of Staphylococcus aureus Newbould 305, a strain associated with mild bovine mastitis.
J. Bacteriol. 2012, 194, 6292–6293. [CrossRef] [PubMed]
Vaccines 2021, 9, 6 15 of 17
23.
Misra, N.; Wines, T.F.; Knopp, C.L.; McGuire, M.A.; Tinker, J.K. Expression, immunogenicity and variation of iron-regulated
surface protein A from bovine isolates of Staphylococcus aureus. FEMS Microbiol. Lett. 2017, 364. [CrossRef] [PubMed]
24.
Xu, H.; Hu, C.; Gong, R.; Chen, Y.; Ren, N.; Xiao, G.; Xie, Q.; Zhang, M.; Liu, Q.; Guo, A.; et al. Evaluation of a novel
chimeric B cell epitope-based vaccine against mastitis induced by either Streptococcus agalactiae or Staphylococcus aureus in mice.
Clin. Vaccine Immunol. 2011, 18, 893–900. [CrossRef] [PubMed]
25.
Castagliuolo, I.; Piccinini, R.; Beggiao, E.; Palu, G.; Mengoli, C.; Ditadi, F.; Vicenzoni, G.; Zecconi, A. Mucosal genetic immunization
against four adhesins protects against Staphylococcus aureus-induced mastitis in mice. Vaccine 2006, 24, 4393–4402. [CrossRef]
26.
Gong, R.; Hu, C.; Xu, H.; Guo, A.; Chen, H.; Zhang, G.; Shi, L. Evaluation of clumping factor A binding region A in a subunit
vaccine against Staphylococcus aureus-induced mastitis in mice. Clin. Vaccine Immunol. 2010, 17, 1746–1752. [CrossRef]
27.
Maira-Litran, T.; Bentancor, L.V.; Bozkurt-Guzel, C.; O’Malley, J.M.; Cywes-Bentley, C.; Pier, G.B. Synthesis and Evaluation of a
Conjugate Vaccine Composed of Staphylococcus aureus Poly-N-Acetyl-Glucosamine and Clumping Factor A. PLoS ONE
2012
,
7, e43813. [CrossRef]
28.
Hawkins, J.; Kodali, S.; Matsuka, Y.V.; McNeil, L.K.; Mininni, T.; Scully, I.L.; Vernachio, J.H.; Severina, E.; Girgenti, D.;
Jansen, K.U.; et al. A recombinant Clumping factor A containing vaccine induces functional antibodies to Staphylococcus aureus
that are not observed after natural exposure. Clin. Vaccine Immunol. 2012. [CrossRef]
29. Creech, C.B.; Frenck, R.W.; Fiquet, A.; Feldman, R.; Kankam, M.K.; Pathirana, S.; Baber, J.; Radley, D.; Cooper, D.; Eiden, J.; et al.
Persistence of Immune Responses Through 36 Months in Healthy Adults After Vaccination with a Novel. Open Forum Infect. Dis.
2020, 7, ofz532. [CrossRef]
30.
Fluit, A.C.; Terlingen, A.M.; Andriessen, L.; Ikawaty, R.; van Mansfeld, R.; Top, J.; Cohen Stuart, J.W.; Leverstein-van Hall, M.A.;
Boel, C.H. Evaluation of the DiversiLab system for detection of hospital outbreaks of infections by different bacterial species.
J. Clin. Microbiol. 2010, 48, 3979–3989. [CrossRef]
31.
Gomez, M.I.; Sordelli, D.O.; Buzzola, F.R.; Garcia, V.E. Induction of cell-mediated immunity to Staphylococcus aureus in the mouse
mammary gland by local immunization with a live attenuated mutant. Infect. Immun. 2002, 70, 4254–4260. [CrossRef]
32.
Lee, J.W.; O’Brien, C.N.; Guidry, A.J.; Paape, M.J.; Shafer-Weaver, K.A.; Zhao, X. Effect of a trivalent vaccine against Staphylo-
coccus aureus mastitis lymphocyte subpopulations, antibody production, and neutrophil phagocytosis. Can. J. Vet. Res.
2005
,
69, 11–18. [PubMed]
33.
Camussone, C.M.; Veaute, C.M.; Porporatto, C.; Morein, B.; Marcipar, I.S.; Calvinho, L.F. Immune response of heifers against a
Staphylococcus aureus CP5 whole cell vaccine formulated with ISCOMATRIX adjuvant. J. Dairy Res. 2012, 1–9. [CrossRef]
34.
Riollet, C.; Rainard, P.; Poutrel, B. Cell subpopulations and cytokine expression in cow milk in response to chronic Staphylococ-
cus aureus infection. J. Dairy Sci 2001, 84, 1077–1084. [CrossRef]
35.
Lin, L.; Ibrahim, A.S.; Xu, X.; Farber, J.M.; Avanesian, V.; Baquir, B.; Fu, Y.; French, S.W.; Edwards, J.E., Jr.; Spellberg, B. Th1-Th17
cells mediate protective adaptive immunity against Staphylococcus aureus and Candida albicans infection in mice. PLoS Pathog.
2009, 5, e1000703. [CrossRef]
36.
Snider, D.P. The Mucosal Adjuvant Activities of ADP-Ribosylating Bacterial Enterotoxins. Crit Rev. Immunol.
2017
, 37, 499–530.
[CrossRef] [PubMed]
37.
George-Chandy, A.; Eriksson, K.; Lebens, M.; Nordstrom, I.; Schon, E.; Holmgren, J. Cholera toxin B subunit as a carrier molecule
promotes antigen presentation and increases CD40 and CD86 expression on antigen-presenting cells. Infect. Immun.
2001
,
69, 5716–5725. [CrossRef]
38.
Schnitzler, A.C.; Burke, J.M.; Wetzler, L.M. Induction of cell signaling events by the cholera toxin B subunit in antigen-presenting
cells. Infect. Immun. 2007, 75, 3150–3159. [CrossRef]
39.
Nashar, T.O.; Hirst, T.R.; Williams, N.A. Modulation of B-cell activation by the B subunit of Escherichia coli enterotoxin: Receptor
interaction up-regulates MHC class II, B7, CD40, CD25 and ICAM-1. Immunology 1997, 91, 572–578. [CrossRef]
40.
Bromander, A.K.; Kjerrulf, M.; Holmgren, J.; Lycke, N. Cholera toxin enhances antigen presentation. Adv. Exp. Med. Biol.
1995
,
371B, 1501–1506.
41.
Cong, Y.; Weaver, C.T.; Elson, C.O. The mucosal adjuvanticity of cholera toxin involves enhancement of costimulatory activity by
selective up-regulation of B7.2 expression. J. Immunol. 1997, 159, 5301–5308.
42.
Eriksson, K.; Fredriksson, M.; Nordstrom, I.; Holmgren, J. Cholera toxin and its B subunit promote dendritic cell vaccination with
different influences on Th1 and Th2 development. Infect. Immun. 2003, 71, 1740–1747. [CrossRef] [PubMed]
43.
Xu-Amano, J.; Jackson, R.J.; Fujihashi, K.; Kiyono, H.; Staats, H.F.; McGhee, J.R. Helper Th1 and Th2 cell responses following
mucosal or systemic immunization with cholera toxin. Vaccine 1994, 12, 903–911. [CrossRef]
44.
Mattsson, J.; Schon, K.; Ekman, L.; Fahlen-Yrlid, L.; Yrlid, U.; Lycke, N.Y. Cholera toxin adjuvant promotes a balanced
Th1/Th2/Th17 response independently of IL-12 and IL-17 by acting on Gsalpha in CD11b(+) DCs. Mucosal Immunol.
2015
,
8, 815–827. [CrossRef]
45.
Jobling, M.G.; Holmes, R.K. Fusion proteins containing the A2 domain of cholera toxin assemble with B polypeptides of cholera
toxin to form immunoreactive and functional holotoxin-like chimeras. Infect. Immun.
1992
, 60, 4915–4924. [CrossRef] [PubMed]
46.
Hajishengallis, G.; Hollingshead, S.K.; Koga, T.; Russell, M.W. Mucosal immunization with a bacterial protein antigen genetically
coupled to cholera toxin A2/B subunits. J. Immunol. 1995, 154, 4322–4332.
Vaccines 2021, 9, 6 16 of 17
47.
Martin, M.; Hajishengallis, G.; Metzger, D.J.; Michalek, S.M.; Connell, T.D.; Russell, M.W. Recombinant antigen-enterotoxin A2/B
chimeric mucosal immunogens differentially enhance antibody responses and B7-dependent costimulation of CD4(+) T cells.
Infect. Immun. 2001, 69, 252–261. [CrossRef]
48.
Sultan, F.; Jin, L.L.; Jobling, M.G.; Holmes, R.K.; Stanley, S.L., Jr. Mucosal immunogenicity of a holotoxin-like molecule
containing the serine-rich Entamoeba histolytica protein (SREHP) fused to the A2 domain of cholera toxin. Infect. Immun.
1998
,
66, 462–468. [CrossRef]
49.
Misra, N.; Wines, T.F.; Knopp, C.L.; Hermann, R.; Bond, L.; Mitchell, B.; McGuire, M.A.; Tinker, J.K. Immunogenicity of a
Staphylococcus aureus-cholera toxin A. Vaccine 2018, 36, 3513–3521. [CrossRef]
50.
Prasad, L.B.; Newbould, F.H. Inoculation of the bovine teat duct with Staph. aureus: The relationship of teat duct lenght, milk yield
and milking rate to development of intramammary infection. Can. Vet. J. 1968, 9, 107–115.
51.
Arlian, B.M.; Tinker, J.K. Mucosal immunization with a Staphylococcus aureus IsdA-cholera toxin A2/B chimera induces antigen-
specific Th2-type responses in mice. Clin. Vaccine Immunol. 2011, 18, 1543–1551. [CrossRef]
52.
Gonzales, V.K.; de Mulder, E.L.; de Boer, T.; Hannink, G.; van Tienen, T.G.; van Heerde, W.L.; Buma, P. Platelet-rich plasma can
replace fetal bovine serum in human meniscus cell cultures. Tissue Eng. Part. C Methods
2013
, 19, 892–899. [CrossRef] [PubMed]
53.
Coussens, P.M.; Verman, N.; Coussens, M.A.; Elftman, M.D.; McNulty, A.M. Cytokine gene expression in peripheral blood
mononuclear cells and tissues of cattle infected with Mycobacterium avium subsp. paratuberculosis: Evidence for an inherent
proinflammatory gene expression pattern. Infect. Immun. 2004, 72, 1409–1422. [CrossRef] [PubMed]
54.
Wenz, J.R.; Garry, F.B.; Barrington, G.M. Comparison of disease severity scoring systems for dairy cattle with acute coliform
mastitis. J. Am. Vet. Med. Assoc. 2006, 229, 259–262. [CrossRef] [PubMed]
55.
Atalla, H.; Gyles, C.; Wilkie, B.; Leslie, K.; Mallard, B. Somatic cell scores and clinical signs following experimental intramammary
infection of dairy cows with a Staphylococcus aureus small colony variant (S. aureus SCV) in comparison to other bovine strains.
Vet. Microbiol. 2009, 137, 326–334. [CrossRef]
56.
Livak, K.J.; Schmittgen, T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta
C(T)) Method. Methods 2001, 25, 402–408. [CrossRef]
57. Shkreta, L.; Talbot, B.G.; Diarra, M.S.; Lacasse, P. Immune responses to a DNA/protein vaccination strategy against Staphylococ-
cus aureus induced mastitis in dairy cows. Vaccine 2004, 23, 114–126. [CrossRef]
58.
Leitner, G.; Lubashevsky, E.; Glickman, A.; Winkler, M.; Saran, A.; Trainin, Z. Development of a Staphylococcus aureus vaccine
against mastitis in dairy cows. I. Challenge trials. Vet. Immunol. Immunopathol. 2003, 93, 31–38. [CrossRef]
59.
Burnham, K.P.; Anderson, D.R. Model Selection and Multimodel Inference: A Practical Information-Theoretic Approach, 2nd ed.;
Springer: New York, NY, USA, 2002; p. 488.
60. Benjamini, Y.; Hochberg, Y. Controlling the False Discovery Rate—A Practical and Powerful Approach to Multiple Testing. J. R.
Stat. Soc. Ser. B Stat. Methodol. 1995, 57, 289–300. [CrossRef]
61.
Burvenich, C.; Van Merris, V.; Mehrzad, J.; Diez-Fraile, A.; Duchateau, L. Severity of E. coli mastitis is mainly determined by cow
factors. Vet. Res. 2003, 34, 521–564. [CrossRef]
62. Proctor, R.A. Immunity to Staphylococcus aureus: Implications for Vaccine Development. Microbiol. Spectr. 2019, 7. [CrossRef]
63.
Wilson, G.J.; Tuffs, S.W.; Wee, B.A.; Seo, K.S.; Park, N.; Connelley, T.; Guinane, C.M.; Morrison, W.I.; Fitzgerald, J.R. Bovine
Staphylococcus aureus Superantigens Stimulate the Entire T Cell Repertoire of Cattle. Infect. Immun.
2018
, 86. [CrossRef] [PubMed]
64.
Gunther, J.; Petzl, W.; Bauer, I.; Ponsuksili, S.; Zerbe, H.; Schuberth, H.J.; Brunner, R.M.; Seyfert, H.M. Differentiating Staphylococ-
cus aureus from Escherichia coli mastitis: S. aureus triggers unbalanced immune-dampening and host cell invasion immediately
after udder infection. Sci Rep. 2017, 7, 4811. [CrossRef] [PubMed]
65.
Schukken, Y.H.; Mallard, B.A.; Dekkers, J.C.; Leslie, K.E.; Stear, M.J. Genetic impact on the risk of intramammary infection
following Staphylococcus aureus challenge. J. Dairy Sci. 1994, 77, 639–647. [CrossRef]
66.
Kim, Y.; Atalla, H.; Mallard, B.; Robert, C.; Karrow, N. Changes in Holstein cow milk and serum proteins during intramammary
infection with three different strains of Staphylococcus aureus. BMC Vet. Res. 2011, 7, 51. [CrossRef] [PubMed]
67.
Liu, T.; Wei, Y.; Liu, G.; Shi, B.; Giovanni, S.; Peterson, J.W.; Chopra, A.K. A mutated cholera toxin without the ADP-
ribosyltransferase activity induces cytokine production and inhibits apoptosis of splenocytes in mice possibly via toll-like
receptor-4 signaling. Mol. Immunol. 2016, 75, 21–27. [CrossRef] [PubMed]
68.
Wiedinger, K.; Pinho, D.; Bitsaktsis, C. Utilization of cholera toxin B as a mucosal adjuvant elicits antibody-mediated protection
against S. pneumoniae infection in mice. Ther. Adv. Vaccines 2017, 5, 15–24. [CrossRef] [PubMed]
69.
Anjuere, F.; George-Chandy, A.; Audant, F.; Rousseau, D.; Holmgren, J.; Czerkinsky, C. Transcutaneous immunization with
cholera toxin B subunit adjuvant suppresses IgE antibody responses via selective induction of Th1 immune responses. J. Immunol.
2003, 170, 1586–1592. [CrossRef]
70.
Albu, D.I.; Jones-Trower, A.; Woron, A.M.; Stellrecht, K.; Broder, C.C.; Metzger, D.W. Intranasal vaccination using interleukin-12
and cholera toxin subunit B as adjuvants to enhance mucosal and systemic immunity to human immunodeficiency virus type 1
glycoproteins. J. Virol. 2003, 77, 5589–5597. [CrossRef]
71.
Baldauf, K.J.; Royal, J.M.; Hamorsky, K.T.; Matoba, N. Cholera toxin B: One subunit with many pharmaceutical applications.
Toxins 2015, 7, 974–996. [CrossRef]
72.
Holmgren, J.; Harandi, A.M.; Czerkinsky, C. Mucosal adjuvants and anti-infection and anti-immunopathology vaccines based on
cholera toxin, cholera toxin B subunit and CpG DNA. Expert Rev. Vaccines 2003, 2, 205–217. [CrossRef]
Vaccines 2021, 9, 6 17 of 17
73.
Royal, J.M.; Matoba, N. Therapeutic Potential of Cholera Toxin B Subunit for the Treatment of Inflammatory Diseases of the
Mucosa. Toxins 2017, 9. [CrossRef] [PubMed]
74.
Zhang, L.; Huang, Y.; Lin, Y.; Shan, Y.; Tan, S.; Cai, W.; Li, H.; Zhang, B.; Men, X.; Lu, Z. Anti-inflammatory effect of cholera toxin
B subunit in experimental stroke. J. Neuroinflammation 2016, 13, 147. [CrossRef] [PubMed]
75.
Russell, M.W.; Moldoveanu, Z.; White, P.L.; Sibert, G.J.; Mestecky, J.; Michalek, S.M. Salivary, nasal, genital, and systemic antibody
responses in monkeys immunized intranasally with a bacterial protein antigen and the Cholera toxin B subunit. Infect. Immun.
1996, 64, 1272–1283. [CrossRef] [PubMed]