You are on page 1of 22

Trained immunity is not universal: oral heat-

inactivated Mycobacterium bovis confers no


protection against the non-enveloped Porcine
Circovirus 2
Elisa Ferreras-Colino 
Instituto de Investigación en Recursos cinegéticos IREC (UCLM-CSIC). Ciudad Real
Jose A. Barasona 
Universidad Complutense de Madrid
Marina Sibila 
Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA),
Campus de la Universitat Autònoma de Barcelona (UAB)
María Mazariegos 
Universidad Complutense de Madrid
Rita Vaz-Rodrigues 
Instituto de Investigación en Recursos cinegéticos IREC (UCLM-CSIC). Ciudad Real
Fátima Cruz 
Universidad Complutense de Madrid
Marinela Contreras 
Instituto de Investigación en Recursos cinegéticos IREC (UCLM-CSIC). Ciudad Real
Joseba M. Garrido 
NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario
Joaquim Segalés 
Unitat Mixta d'Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA),
Campus de la Universitat Autònoma de Barcelona (UAB)
José Fuente 
Instituto de Investigación en Recursos cinegéticos IREC (UCLM-CSIC). Ciudad Real
Lucas Domínguez 
Universidad Complutense de Madrid
Christian Gortázar 
(

christian.gortazar@uclm.es
)
Instituto de Investigación en Recursos cinegéticos IREC (UCLM-CSIC). Ciudad Real
Maria A. Risalde 
Universidad de Córdoba

Page 1/22
Research Article

Keywords: heat-inactivated Mycobacterium bovis, porcine circovirus type 2, trained immunity, α-Gal

Posted Date: May 3rd, 2023

DOI: https://doi.org/10.21203/rs.3.rs-2865092/v1

License:


This work is licensed under a Creative Commons Attribution 4.0 International
License.
 
Read Full License

Page 2/22
Abstract
Background

Trained immunity, the enhanced response of innate cells leading to an improved innate immune
response, and antibodies against the glycan galactose-α-1,3-galactose (α-Gal), produced by animals
unable to synthesize α-Gal epitopes, have been suggested to provide the host certain advantage in
infections with enveloped viruses. Conversely, the evidence of protection against non-enveloped viruses
attributed to the referred mechanisms remains scarce. Aiming to evaluate whether a heat-inactivated
Mycobacterium bovis (HIMB) immunostimulant, which had proven to protect against related and non-
related pathogens, confers an advantage against non-enveloped viruses, we performed an immunization
and challenge experiment with porcine circovirus 2 (PCV-2) in swine. Sixteen piglets were randomly
assigned to the immunized group (n = 8), which received two oral doses of HIMB with an interval of three
weeks, or to the control group (n = 8). All animals were infected by intranasal inoculation with PCV-2 21
days later and euthanized at day 21 post-challenge.

Results

No differences in body weight and body temperature, viremia and viral burden in target tissues, antibody
production and histopathological changes in target tissues were observed between the immunized and
the control group. Overall, oral immunization with HIMB did not protect pigs against PCV-2 infection.

Conclusions

Our study suggests that HIMB confers no advantage against pathogens lacking α-Gal, mainly non-
enveloped viruses such as PCV-2, in α-Gal-producing hosts, such as the swine.

Background
The term “trained immunity” was proposed by Netea et al. (2011) [1] to refer to an adaptative response
elicited by the innate immune system towards a subsequent encounter not only with the same pathogen
but also with non-related ones. Trained immunity or “innate immune memory” occurs in a T and B cell-
independent and non-specific manner and, thus, the innate immunity may provide cross-protection
against a wide range of pathogens even in the absence of an adaptative response [2]. Trained immunity
involves epigenetic modifications and metabolic changes functioning interdependently within innate
cells, mainly natural killer (NK) cells and monocytes/macrophages [3–5]. 

The Bacille Calmette-Guérin (BCG) vaccine, a live attenuated Mycobacterium bovis strain, beyond being
the only commercialized vaccine to prevent human tuberculosis, has emerged as one of the most widely
explored inducers of trained immunity [6]. Indeed, numerous in vitro and in vivo experiences have
demonstrated that BCG boosts proinflammatory cytokines production in NK cells and

Page 3/22
monocytes/macrophages upon restimulation with non-related microorganisms, thus leading to non-
specific and adaptative-independent protection in the host [6].

Since 2020, worldwide labelled as the year of the pandemic of coronavirus disease 19 (COVID- 19)
caused by severe acute respiratory syndrome coronavirus 2 (SARS- CoV- 2), the scientific interest
regarding the role of “trained immunity” in viral infections has risen (over 60 entries per year in 2020,
2021 and 2022 versus 20 or less in previous years in the Scopus database). Nevertheless, the protection
against viral infection attributed to trained immunity is still unclear (Table 1). Preliminary experiences of
immunization with BCG or β-glucans evidenced protection against diverse viruses. Further assessment of
the effect of both immunostimulants in viral infections confirmed the reduction in viral burden, mortality,
and morbidity, as well as a shift in the cytokine profile (Table 1). However, certain experimental studies
reported failure of the heterologous protection expected from trained immunity inducers. For instance, no
protective effect of prior BCG vaccination was observed regarding influenza infection [7], and no
protective effect of β-glucans in zebrafish infected with spring viremia carp virus [8] nor in turbots
infected with viral hemorrhagic septicemia virus [9]. In addition, BCG does not reduce lesions, morbidity,
mortality or viral burden in the SARS-CoV-2 infection model in mice [10] or primates [11]. Most of the
literature refers to a beneficial effect of trained immunity against viruses that are enveloped, while
evidence of protection against non-enveloped viruses is limited (for non-enveloped viruses see: [12–18]).

Primates, including humans, as well as fish, amphibians, reptiles, and birds, evolved with the inability to
synthesize the glycan galactose-α-1,3-galactose (α-Gal). Therefore, they naturally produce antibodies in
response to this modification present in glycoproteins, glycolipids and other biomolecules present in other
species’ cells, including gastrointestinal microbiota, and viruses [19,20]. Thus, antibodies against α-Gal
have been suggested to confer non-specific protection against enveloped viruses containing α-Gal
epitopes in its glycoproteins [19,21,22]. Anti-α-Gal antibodies bound to viruses may activate the
complement system for virus lysis or uptake by dendritic cells and macrophages and be presented to T
cells to trigger adaptative responses [19]. Trained immunity has been also proposed to be associated with
α-Gal [22,23]. Accordingly, several experimental studies have demonstrated that anti-α-Gal antibodies
contribute to neutralization of enveloped viruses [24,25]; thus, efficacy of enveloped virus vaccines can be
increased by expression of α-Gal epitopes in the viral surface [26–29]. 

Although it has been more extensively explored in rodents [30–32] and humans [14,15,33], the novel
phenomenon of training the innate immunity arouses growing interest in domestic animals (reviewed
in Angulo and Angulo, 2022). In particular, piglets that had been administered Saccharomyces
cerevisae β-glucan orally prior to being challenged with the enveloped swine influenza virus displayed
milder clinical signs and lung lesions, as well as higher interferon (IFN)-γ and nitric oxide (NO) levels [35].
Likewise, priming with β-glucan impaired cell invasion and replication of the enveloped African Swine
Fever virus and boosted IFNα and interleukin (IL)-6 levels in porcine alveolar macrophages [36]. Recently,
Byrne et al. (2020) [37] provided the first evidence of trained immunity induced by BCG in pigs, as they
observed that stimulating porcine monocytes with BCG in vitro enhanced IL-1β and tumor necrosis factor
(TNF)-α gene expression and protein production upon LPS re-stimulation. In line with this finding, pigs
Page 4/22
that had received an immunostimulant composed of heat-inactivated M. bovis (HIMB) via the oral route
prior to being challenged with Salmonella enterica serovar Choleraesuis displayed increased
proinflammatory cytokines production and antioxidant activity, together with reduced pulmonary lesions,
compared with non-immunized pigs [38]. Moreover, HIMB also elicits partial immunity against
protozoal [39] and arthropod (ticks; [40] infections in different animal models, although its cross-
protection against viruses has not been evaluated to date. 

Porcine circovirus 2 (PCV-2), a small non-enveloped virus, is considered one of the most relevant
pathogens that affect swine worldwide, mainly due to the substantial economic losses it causes to the
pig industry [41]. PCV-2 infection has been associated with various disease syndromes in both domestic
pigs and wild boar, such as PCV-2-systemic disease (PCV-2-SD, formerly known as postweaning
multisystemic wasting syndrome), PCV-2-reproductive disease, PCV-2-subclinical infection and porcine
dermatitis and nephropathy syndrome (PDNS). Importantly, PCV-2-SD affected pigs suffer from
immunosuppression [41–44]. Currently, all commercialized PCV-2 vaccines are only indicated for the
parenteral route [41], which is a limitation to target wild boar and free-range pigs  [45,46]. Consequently,
although several attempts to develop oral vaccines composed of recombinant bacteria [47,48] or
yeasts [49–52] expressing PCV-2 Cap protein have revealed promising results, there is still a long way to
go before achieving an orally delivered preparation that is feasible to protect swine against PCV-2.

Thus, the objective of the present study was to evaluate the effect of the oral HIMB immunostimulant in
the porcine model of infection with the non-enveloped virus PCV-2. 

Results
Clinical signs

No clinical sings were observed throughout the experiment. No significant effect of the immunization was
observed on either body temperature (Figure 2A) or weight gain (Figure 2B).

PCV-2 load

Viremia and viral load were estimated weekly by detecting PCV-2 DNA load in blood and the progression
was similar in both groups: steadily increased after infection (viremia: HIMB = 3/8, control = 3/8; viral
load: HIMB = 2.97 x 104 copies/ml, control = 3.38 x 104 copies/ml), reaching a peak 14 dpi (viremia: HIMB
= 8/8, control = 8/8; viral load: HIMB = 5.75 x 104 copies/ml, control = 5.59 x 104 copies/ml), and then
decreased until the end of the experiment (viremia: HIMB = 3/8, control = 5/8; viral load: HIMB = 2.20 x
104 copies/ml, control = 2.40 x 104 copies/ml) (Figure 3A). No differences in percentage of viremic
animals and mean viral loads between groups were detected at any timepoint. PCV-2 antigen in target
tissues was evaluated after necropsy, detecting low amount of antigen in both groups (mean total score:
HIMB = 0.0892; control = 0.0892). Although the immunized group showed an apparently higher
proportion of animals with one or more PCV-2-immunolabeled tissues (HIMB: 4/8 (50%), control: 3/8
(37.5%)), the differences between groups were not significant (P = 0.72).
Page 5/22
Humoral response

Humoral response was measured through the production of anti-PCV-2 antibodies. As expected, antibody
titers started raising upon infection, reaching maximum levels twenty-one days after infection (Figure 3B).
No differences in antibody S/P ratios between groups were detected. No anti-P22 antibodies were
detected.

Pathological findings                    

To assess protection against PCV-2, macroscopic and microscopic lesions were examined at the end of
the experiment. All individuals showed a good general condition at necropsy. Among the macroscopic
lesions, the main findings were moderate congestion and lymphadenomegaly in several LNs, especially in
the mandibular, tracheobronquial, mediastinal, mesenteric and inguinal ones (Figure 4A). The proportion
of animals with lymphadenomegaly in the pulmonary LNs (tracheobronquial and mediastinal) was
significantly higher in the HIMB group (7/8, 87.5%) compared with the control group (1/8, 12.5%)
(P<0.04). Microscopically, mandibular and pulmonary LNs presented mild lymphocyte depletion and low
number of histiocytes and multinucleated giant cells infiltrate (Figure 4B, and Figures 5A, B), occasionally
associated to low amount of PCV-2 antigen (Figure 4B inset and Figures 5A, B). Scarce presence of PCV-2
antigen also was observed in the tonsil of some animals, along with light infiltrating of histiocytes and
multinucleated giant cells, and lymphocyte depletion (Figure 5C). Moreover, mild to moderate interstitial
pneumonia was frequently observed in both groups, composed by infiltrate of mononuclear cells in the
pulmonary parenchymal (Figures 4C, D and Figure 5D). In addition, one individual from each group
showed mild interstitial nephritis (data not shown). The appearance and severity of microscopic lesions
was similar between groups and no significant differences were detected (Figure 5). 

α-Gal content in PCV-2

The presence of α-Gal in PCV-2 was assessed by a direct ELISA of the infective viral inoculum, with an
uninfected PK15 cell culture as control. As expected for non-enveloped viruses such as PCV-2, α-Gal
content was lower in purified viruses (0.20 ng/μg) than in cultured cells from an α-Gal producing animal
such as the pig (0.43 ng/μg).

Discussion
Stimulating the immune system with trained immunity inducers, such as mycobacterial or fungal derived
compounds, constitutes a strategy to elicit non-specific protection against a wide range of pathogens,
including certain viruses [5]. However, oral immunization with HIMB, an immunostimulant that had
previously demonstrated protective effect attributed to trained immunity in heterologous infections [38,
39, 61] failed to exert protective effects in the PCV-2 infection model.

A paramount concern about PCV-2-SD in the pig industry is the detriment in the average daily weigh gain
[62]. In the present study, oral HIMB immunization did not influence weigh gain or body temperature in

Page 6/22
pigs infected with PCV-2. Although [63] did not observe a beneficial effect on growth performance after
immunization, most field trials showed better weigh gain in pigs vaccinated with a commercial vaccine
[64, 65].

Other hallmarks of PCV-2-SD in pigs are lymphocyte depletion together with granulomatous inflammation
of lymphoid tissues and interstitial pneumonia [66]. In agreement with previous studies [57, 67, 68],
interstitial pneumonia and enlarged LNs were our predominant findings, with occasional presence of
lymphocyte depletion and histiocytic infiltrate with multinucleated giant cells, pigs orally immunized with
HIMB not showing lower lesion scores the compared to the control animals. The capacity of
commercialized vaccines for alleviating pathology in naturally PCV-2 infected pigs has been widely
demonstrated in the field [69, 70], and the beneficial effect has also been evident under experimental
conditions. For instance, Seo et al. (2014) [71] observed lower lymphoid depletion and granulomatous
replacement in pigs vaccinated with either Fostera PCV/Suvaxyn PCV-2 one dose (Zoetis), Circovac
(Merial), Circoflex (Boehringer Ingelheim Vetmedica) and Porcilis PCV (Merck, Sharp and Dohme Animal
Health) and infected with PCV-2 compared with non-vaccinated pigs.

Oral immunization with HIMB did not reduce PCV-2 load in pig, illustrated by viremia and viral load in
tissues, and did not stimulate a specific humoral response. Conversely, the efficacy of commercialized
specific vaccines to reduce viremia and viral burden in tissues in pigs infected with PCV-2, associated to a
strong antibody response, has been confirmed under experimental and field conditions [63–66, 71, 72].

The present study challenges the prevailing paradigm of trained immunity being universal and suggests
that it might not be extendable to all pathogens, for instance, non-enveloped viruses such as PCV-2.
Similarly, the lack of effect of β-glucan or BCG in experimental or natural infections with enveloped
viruses had been previously reported [7–11, 73, 74] However, to the best of our knowledge, only a few
studies have explored the protective efficacy of the compounds of interest against non-enveloped viruses
[12–18]. Xue et al. (2017) [16] assessed the effect of Astragalus spp. polysaccharides on PCV-2 infection
in an in vitro model using porcine PK15 cells and in an in vivo model using mice, and concluded that
Astragalus spp. polysaccharide suppressed PCV-2 infection by inhibiting endoplasmic reticulum stress.
Still, the latter results should be adressed carefully and further research is needed to extrapolate them to
PCV-2 infection in its natural host, the pig.

Ultimately, we confirmed that, as expected from a non-enveloped virus [19, 21], PCV-2 does not contain α-
Gal. Unlike for enveloped viruses (because the carbohydrate chains of the envelope glycoproteins are
adquired from the host golgi apparatus during viral replication), activation of the innate components,
such as the complement system and antigen presenting cells, mediated by natural anti-α-Gal antibodies
[19, 21] would not contribute in the host immunity against non-enveloped viruses including PCV-2.
Furthermore, non-specific immune antibody-mediated and non-antibody-mediated mechanisms in
response to α-Gal may not be activated in α-Gal-positive hosts.

Conclusions
Page 7/22
In conclusion, our results demonstrate that oral immunization with HIMB does not protect swine against
challenge with PCV-2, a non-enveloped DNA virus. Considering the absence of α-Gal in PCV-2, our study
suggests that HIMB confers no advantage against pathogens lacking α-Gal in hosts producing this
molecule.

Methods
Animals and experimental design

Sixteen 21 days-old Landrace x Large White hybrid female piglets with homogeneous weights and tested
as PCV-2 PCR negative with low anti-PCV2 antibody titers were obtained from a commercial pig
production farm. Animals were housed in class III biocontainment animal facilities (BSL-3) located at
VISAVET Health Surveillance Centre (Madrid, Spain) for acclimatization one week before the experiment.
All animals received continuous access to water, non-medicated pig feed and veterinary monitorization.
Individuals were randomly assigned to either the immunized group (n=8), which received two oral doses
of HIMB with an interval of three weeks (-42 and -21 days pre-infection), or the control group (n=8), which
received PBS instead of immunostimulant. Twenty-one days after the second immunization dose (day 0),
all animals from both groups were infected by intranasal inoculation with 105 TCID 50 of PCV-2 per
animal (Figure 1). All pigs were confirmed as PCV-2 PCR negative and PCV-2 ELISA negative before
challenge. Animals were handled on 0, 7, 14 and 21 days post-infection (dpi) for blood sampling, rectal
temperature and weight measurements. 

All piglets were euthanized 21 dpi by captive bolt after sedation with xylazine (Xilagesic 2%, Laboratories
Calier, Barcelona, Spain) and were subjected to necropsy to assess PCV-2 gross lesions in palatine tonsil,
as well as mandibular, tracheobronchial and mediastinal lymph nodes (LNs), lung (cranial and caudal
lobes) and kidney. Typical PCV-2 gross lesions such as lymphadenomegaly, interstitial pneumonia and
nephritis were evaluated by experienced pathologists. Lesion severity was graded with a lesion score (0
= absent, 1 = mild, 2 = moderate or 3 = severe).

Samples from palatine tonsil, lung (cranial and caudal lobes), as well as mandibular, tracheobronchial
and mediastinal LNs were fixed by immersion in 10% neutral buffered formalin during 24 hours at room
temperature (RT), dehydrated in a graded series of ethanol, immersed in xylol, and embedded in paraffin
wax using an automatic processor for further histopathology and immunohistochemical studies. 

Heat-inactivated Mycobacterium bovis (HIMB) immunostimulant 

The oral immunostimulant consisted of 2 mL of sterile PBS containing approximately 107 heat-
inactivated colony forming units (CFU)/mL of a field isolate (Strain 1403; spoligotype SB0339) originally
obtained from a naturally infected wild boar. HIMB was prepared at Neiker-Tecnicalia (Derio, Spain)
following the protocol described by Garrido et al. (2011) [53], except for an extended inactivation step at
83 °C for 45 minutes (min). Bacterial concentration was determined prior to inactivation by measuring
Turbidity in a VITEK® DensiCHEK® (BioMerieux) and by plating a serially diluted aliquot onto agar-
Page 8/22
solidified Middlebrook 7H9 with glycerol (0.2% v/v) and OADC (10% v/v) (Becton Dickinson, Franklin
Lakes, NJ, USA).

Porcine circovirus 2

PCV-2 genotype b (Sp-10-7-54-13 strain at 14th passage) was used as the inoculum [54]. This strain was
isolated from the lymphoid tissues of a field case of PCV-2-SD in 2006 in Spain. For this study, the PCV-2
strain was propagated in PCV-free PK15 cells in minimal essential medium (MEM) [55]  to a titer of 105
TCID 50/mL. 

Humoral response

Blood was extracted through puncture of the sinus ophthalmicus at intervals of one week upon infection.
Blood samples were centrifuged at 400 g for 10 min to extract sera which was further stored at – 20 °C. 

Sera were tested in duplicate by means of Ingezim CIRCO IgG ELISA (R.11.PCV.K1; Eurofins Technologies,
SA; Madrid, Spain) to detect antibodies against PCV-2 VP2 protein, following manufacturer’s instructions.

Serum samples were also analyzed by ELISA to detect antibodies against Mycobacterium tuberculosis
complex using P22 as antigen in an indirect in-house ELISA previously described by Thomas et al.
(2019) [56]. The estimated sensitivity and specificity of this ELISA in swine was 84.1% and 98.4%,
respectively.

Detection of porcine circovirus 2 infection by real time quantitative PCR (qPCR)

DNA was extracted from 200 μL of serum by using the MagMAX™ Pathogen RNA/DNA Kit (Thermo
Fischer Scientific Baltics. Vilnius, Lithuania) following the manufacturer’s instructions. The DNA obtained
was suspended in 90 μL of elution solution. The commercial QPCR kit VetMAX™ Porcine PCV2 Quant Kit
(Applied Biosystems, Lisseu, France) was used to detect and quantify PCV-2 DNA in serum samples. PCV-
2 qPCR results were expressed as PCV-2 copies/mL of serum. A cutoff value of 104 PCV-2 DNA
copies/mL was considered the quantification limit of the PCV-2 qPCR. Viremia represented the proportion
of positive animals (quantifiable and non-quantifiable) per group, and viral load represented the mean of
the quantifiable individuals per group.

Detection of porcine circovirus 2 infection by immunohistochemistry

Immunohistochemistry was performed to detect PCV-2 as previously described in Chianini et al.


(2003) [57]. Briefly, tissue sections were deparaffinized with xylene and rehydrated through graded
alcohols. Endogenous peroxidase activity was blocked by incubation with hydrogen peroxide 3% in
distilled water for 30 min. Afterwards, antigen retrieval was performed using 0.1% Protease XIV (Sigma,
ref. P5147) in PBS for 8 min at 37ºC. A monoclonal PCV-2 antibody (Ingenasa, M.11.PCV.I36A9) able to
recognize the Cap protein was used at 1/1000 dilution in 0.1 M Tris-buffered saline and incubated
overnight at 4-8ºC. Dako EnVision System-HRP Labelled Polymer Anti-Mouse (Dako, ref. K4001) was
Page 9/22
used as secondary antibody and incubated during 60 min at RT. Sections were finally incubated in
diaminobenzidine (DAB)–hydro- gen peroxide solution for 10 min, counterstained with Harris’
haematoxylin, dehydrated and coverslipped. 

Sections from a previous case of PCV-2-SD diagnosed in Spain were used as positive control and tissues
from pigs from an experimental farm with no history of PCV-2 infection as negative controls.

To evaluate the number of immunolabeled cells, the sections were randomly chosen and blinded to which
group was being analyzed and counted by an experienced pathologist (J.S.). PCV-2 antigen detection by
immunohistochemistry was graded by score as low (1), moderate (2) and intense (3) amount of antigen.

Histopathology

Sections of tissue samples embedded in paraffin wax were cut at 3 µm and stained with hematoxylin and
eosin (HE) for further histological examinations. The presence of lymphocyte depletion, infiltrating
histiocytic and multinucleated giant cells in LNs and tonsils, the lymph-epithelium necrosis in tonsils, as
well as the interstitial pneumonia, type-II pneumocyte hyperplasia and alveolar edema in lungs were
graded as absent or 0% (0), mild or 1% to 30% affected (1), moderate or 30% to 70% affected (2), and
severe or 70% to 100% affected (3). The referred pathological findings were independently examined by
two blinded and experienced observers (E.F-C. and M.A.R.).

α-Gal content in PCV-2

The PCV-2 infective inoculum and an uninfected PK15 cell culture were tested with in-house direct ELISA
to detect α-Gal, following the protocol described by Villar et al. (2021) [58] with slight modifications. First,
protein concentration in the inoculum and in the cells was determined with the Bicinchoninic Acid (BCA)
Protein Assay Kit (Thermo Fisher Scientific, Waltham, MA, USA), following manufacturer’s instructions.
Thereafter, 96-well plates were coated with four serial dilutions of protein (from 0.1 to 0.00001 µg/µL per
well) from the viral inoculum and the PK15 cells in duplicate, as well as five serial dilutions of α-Gal (from
2 x 10-7 to 1.25 x 10-8  µg/µL per well), in carbonate/bicarbonate buffer and incubated overnight at 4°C.
Wells were subsequently washed with PBS solution containing 0.05% Tween-20 (PBST) (Tween 20;
Sigma-Aldrich Quimica S.A., Madrid, Spain) three times and blocked with 5% skimmed milk powder
solution in PBS (SM) for 1 h at RT. Then, wells were emptied and the anti-α-Gal epitope monoclonal
antibody (M86; Enzo Life Sciences) was added at 1:100 dilution in PBS and incubated for 1 h at 37°C.
After three washes with PBST, goat anti-mouse IgM (μ-chain specific) peroxidase-conjugated antibody
(Sigma-Aldrich Quimica S.A.) was added at 1:2000 dilution in PBS. After three washes with PBST, 100 μL
of 3,3´,5,5´- Tetramethylbenzidine (Promega, Madison, WI, USA) were incubated for 20 min in the dark at
RT. The colorimetric reaction was stopped with H2SO4 3N and the optical density (OD) was measured at
450 nm with a Multiskan FC ELISA reader (Thermo Fisher Scientific). The average value of the blanks
(uncoated wells; n = 2) was subtracted from all reads and the mean value of the replicates (wells coated
with the same sample at the same protein dilution; n = 2) was calculated. α-Gal content (ng/μg) in each
sample (PCV2 inoculum and uninfected PK15 cells) was calculated from an α-Gal standard curve. 
Page 10/22
Statistical analysis

Statistical analysis was performed using R version 4.0.3 (R Core Team, 2020) using packages
lme4 [59] and ggplot2 [60]. All data were assessed to calculate mean ± standard deviation (SD) or error
(SE) values. The variables viremia, viral burden, antibody titers, increments in weight gain and increments
in body temperature were fitted with linear mixed-effects models including the categorical variables
“group” and “day” as fixed factors with interaction, as well as “individual” as random factor [response
variable ~ group * day + (1 | individual)]. Differences in macroscopic and microscopic lesion scores
between groups were tested through Mann-Whitney U test. P values ≤ 0.05 were considered statistically
significant with a confidence level (CL) of 95%.

Declarations
Ethics approval

Handling of the animals and sampling was performed according to European (Council Directive 86/609)
and Spanish Legislation (RD53/2013) and approved by the Ethics Committee (Complutense University of
Madrid) and the Regional Agriculture Authority (Comunidad de Madrid; permit number: PROEX: 66/16).

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author
on reasonable request.

Competing interests

The authors declare that they have no competing interests.

Funding

The present study has been funded by project MYCOTRAINING SBPLY/19/180501/000174 (Junta de
Castilla-La Mancha, Spain, and EU-FEDER). E. Ferreras-Colino (2020/3836) and R. Vaz-Rodrigues
(2022/20675) were supported by the predoctoral contract from Universidad de Castilla-La Mancha
(UCLM), co-financed by the European Social Fund (ESF). Marinela Contreras was supported by the
Ministerio de Ciencia, Innovación y Universidades, Spain, grant IJC2020-042710-I.

Author’s contributions

CG, MAR, JF, JMG, LD: conceived, designed and supervised the study. JMG: prepared the
immunostimulant. MS, JS: prepared the viral inoculum and performed PCR and IHQ to detect PCV2. JAB,
MM, FC, MAR: performed the necropsies, evaluated macroscopic lesions and collected the samples. MAR:
performed ELISA to detect anti-PCV2 antibodies. EFC and MAR: evaluated microscopic lesions. EFC and
MC: performed ELISA to detect anti-gal antibodies. EFC and RVR: performed the statistical analyses and

Page 11/22
graphic visualization. EFC: prepared the first manuscript draft. All authors read and approved the final
manuscript. 

Acknowledgements

The authors would like to thank the staff of VISAVET and IREC for their technical assistance, as well as to
Mónica Pérez, Anna Llorens and Eva Huerta at IRTA-CReSA for their technical assistance on
immunohistochemistry, virology and qPCR, respectively.

References
1. Netea MG, Quintin J, Van Der Meer JWM. Trained immunity: a memory for innate host defense. Cell
Host Microbe [Internet]. Cell Host Microbe; 2011 [cited 2022 Feb 14];9:355–61. Available from:
https://pubmed.ncbi.nlm.nih.gov/21575907/
2. Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, et al. Defining trained
immunity and its role in health and disease [Internet]. Nat. Rev. Immunol. Nature Research; 2020
[cited 2021 Mar 31]. p. 375–88. Available from: https://pubmed.ncbi.nlm.nih.gov/32132681/
3. Fanucchi S, Domínguez-Andrés J, Joosten LAB, Netea MG, Mhlanga MM. The Intersection of
Epigenetics and Metabolism in Trained Immunity. Immunity. Cell Press; 2021. p. 32–43.
4. Domínguez-Andrés J, Fanucchi S, Joosten LAB, Mhlanga MM, Netea MG. Advances in understanding
molecular regulation of innate immune memory. Curr. Opin. Cell Biol. Elsevier Ltd; 2020. p. 68–75.
5. Taks EJM, Moorlag SJCFM, Netea MG, van der Meer JWM. Shifting the Immune Memory Paradigm:
Trained Immunity in Viral Infections. Annu Rev Virol. Annual Reviews; 2022;9:469–89.
6. Singh AK, Netea MG, Bishai WR. BCG turns 100: its nontraditional uses against viruses, cancer, and
immunologic diseases. J Clin Invest [Internet]. J Clin Invest; 2021 [cited 2022 Jul 3];131. Available
from: https://pubmed.ncbi.nlm.nih.gov/34060492/
7. de Bree LCJ, Marijnissen RJ, Kel JM, Huber SKR, Aaby P, Benn CS, et al. Bacillus Calmette-Guérin-
Induced trained immunity is not protective for experimental influenza A/Anhui/1/2013 (H7N9)
infection in mice. Front Immunol [Internet]. Frontiers Media S.A.; 2018 [cited 2021 Jan 30];9.
Available from: https://pubmed.ncbi.nlm.nih.gov/29760700/
8. Álvarez-Rodríguez M, Pereiro P, Reyes-López FE, Tort L, Figueras A, Novoa B. Analysis of the long-
lived responses induced by immunostimulants and their effects on a viral infection in Zebrafish
(Danio rerio). Front Immunol [Internet]. Frontiers Media S.A.; 2018 [cited 2021 Jan 30];9. Available
from: https://pubmed.ncbi.nlm.nih.gov/30038625/
9. Librán-Pérez M, Costa MM, Figueras A, Novoa B. β-glucan administration induces metabolic changes
and differential survival rates after bacterial or viral infection in turbot (Scophthalmus maximus).
Fish Shellfish Immunol [Internet]. Fish Shellfish Immunol; 2018 [cited 2021 Dec 13];82:173–82.
Available from: https://pubmed.ncbi.nlm.nih.gov/30081180/

Page 12/22
10. Kaufmann E, Khan N, Tran KA, Ulndreaj A, Pernet E, Fontes G, et al. BCG vaccination provides
protection against IAV but not SARS-CoV-2. Cell Rep. Elsevier B.V.; 2022;38.
11. White AD, Sibley L, Sarfas C, Morrison AL, Bewley K, Churchward C, et al. Influence of Aerosol
Delivered BCG Vaccination on Immunological and Disease Parameters Following SARS-CoV-2
Challenge in Rhesus Macaques. Front Immunol. 2022;12:1–15.
12. Lodmell DL, Ewalt LC. Induction of enhanced resistance against encephalomyocarditis virus
infection of mice by nonviable Mycobacterium tuberculosis: Mechanisms of protection. Infect
Immun [Internet]. American Society for Microbiology (ASM); 1978 [cited 2022 Apr 7];22:740–5.
Available from: /pmc/articles/PMC422222/?report = abstract
13. Kim Y sin, Ke F, Zhang QY. Effect of β-glucan on activity of antioxidant enzymes and Mx gene
expression in virus infected grass carp. Fish Shellfish Immunol [Internet]. Elsevier Ltd; 2009;27:336–
40. Available from: http://dx.doi.org/10.1016/j.fsi.2009.06.006
14. Salem A, Nofal A, Hosny D. Treatment of common and plane warts in children with topical viable
bacillus calmette-guerin. Pediatr Dermatol. 2013;30:60–3.
15. Podder I, Bhattacharya S, Mishra V, Sarkar TK, Chandra S, Sil A, et al. Immunotherapy in viral warts
with intradermal Bacillus Calmette-Guerin vaccine versus intradermal tuberculin purified protein
derivative: A double-blind, randomized controlled trial comparing effectiveness and safety in a
tertiary care center in Eastern I. Indian J Dermatol Venereol Leprol. Medknow Publications;
2017;83:411.
16. Xue H, Gan F, Qian G, Hu J, Hao S, Xu J, et al. Astragalus polysaccharides attenuate PCV2 infection
by inhibiting endoplasmic reticulum stress in vivo and in vitro. Sci Rep [Internet]. Nature Publishing
Group; 2017;7:1–12. Available from: http://dx.doi.org/10.1038/srep40440
17. Tsai CY, Hu SY, Santos HM, Catulin GEM, Tayo LL, Chuang KP. Probiotic supplementation containing
Bacillus velezensis enhances expression of immune regulatory genes against pigeon circovirus in
pigeons (Columba livia). J Appl Microbiol. 2021;130:1695–704.
18. Krishnan R, Jang YS, Oh MJ. Beta glucan induced immune priming protects against nervous
necrosis virus infection in sevenband grouper. Fish Shellfish Immunol [Internet]. Fish Shellfish
Immunol; 2022 [cited 2022 Feb 7];121:163–71. Available from:
https://pubmed.ncbi.nlm.nih.gov/35017048/
19. Galili U. Evolution in primates by “Catastrophic-selection” interplay between enveloped virus
epidemics, mutated genes of enzymes synthesizing carbohydrate antigens, and natural anti-
carbohydrate antibodies [Internet]. Am. J. Phys. Anthropol. John Wiley & Sons, Ltd; 2019 [cited 2023
Feb 14]. p. 352–63. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/ajpa.23745
20. Vaz-Rodrigues R, Mazuecos L, Fuente J de la. Current and Future Strategies for the Diagnosis and
Treatment of the Alpha-Gal Syndrome (AGS). J Asthma Allergy [Internet]. J Asthma Allergy; 2022
[cited 2023 Apr 17];15:957–70. Available from: https://pubmed.ncbi.nlm.nih.gov/35879928/
21. Galili U. Host Synthesized Carbohydrate Antigens on Viral Glycoproteins as “Achilles’’ Heel" of Viruses
Contributing to Anti-Viral Immune Protection.” Int J Mol Sci [Internet]. Int J Mol Sci; 2020 [cited 2022

Page 13/22
Apr 4];21:1–26. Available from: https://pubmed.ncbi.nlm.nih.gov/32933166/
22. De La Fuente J, Gortázar C, Cabezas-Cruz A. Immunity to glycan α-Gal and possibilities for the
control of COVID-19. Immunotherapy [Internet]. Immunotherapy; 2021 [cited 2023 Feb 14];13:185–8.
Available from: https://pubmed.ncbi.nlm.nih.gov/33307805/
23. Juste RA, Ferreras-Colino E, de la Fuente J, Domínguez M, Risalde MA, Domínguez L, et al. Heat
inactivated mycobacteria, alpha-Gal and zebrafish: Insights gained from experiences with two
promising trained immunity inductors and a validated animal model. Immunology. 2022;
24. Hayashi S, Ogawa S, Takashima Y, Otsuka H. The neutralization of pseudorabies virus by anti-α-
galactocyl natural antibody in normal serum. Virus Res. Elsevier; 2004;99:1–7.
25. Rother RP, Fodor WL, Springhorn JP, Birks CW, Setter E, Sandrin MS, et al. A Novel Mechanism of
Retrovirus Inactivation in Human Serum Mediated by Anti-ot-Galactosyl Natural Antibody. J Exp Med.
1995;182:1345–55.
26. Abdel-Motal UM, Guay HM, Wigglesworth K, Welsh RM, Galili U. Immunogenicity of Influenza Virus
Vaccine Is Increased by Anti-Gal-Mediated Targeting to Antigen-Presenting Cells. J Virol.
2007;81:9131–41.
27. Abdel-Motal UM, Wigglesworth K, Galili U. Mechanism for increased immunogenicity of vaccines that
form in vivo immune complexes with the natural anti-Gal antibody. Vaccine [Internet]. Vaccine; 2009
[cited 2022 Apr 4];27:3072–82. Available from: https://pubmed.ncbi.nlm.nih.gov/19428921/
28. Henion TR, Gerhard W, Anaraki F, Galili U. Synthesis of α-gal epitopes on influenza virus vaccines, by
recombinant α1,3galactosyltransferase, enables the formation of immune complexes with the
natural anti-Gal antibody. Vaccine. Elsevier; 1997;15:1174–82.
29. Galili U. Increasing Efficacy of Enveloped Whole-Virus Vaccines by In situ Immune-Complexing with
the Natural Anti-Gal Antibody. Med Res Arch [Internet]. Med Res Arch; 2021 [cited 2022 Apr 4];9.
Available from: https://pubmed.ncbi.nlm.nih.gov/34853815/
30. Murphy EA, Davis JM, Brown AS, Carmichael MD, Ghaffar A, Mayer EP. Effects of oat β-glucan on the
macrophage cytokine response to herpes simplex virus 1 infection in vitro. J Interf Cytokine Res.
2012;32:362–7.
31. Kulkarni S, Mukherjee S, Pandey A, Dahake R, Padmanabhan U, Chowdhary AS. Bacillus Calmette-
Guérin Confers Neuroprotection in a Murine Model of Japanese Encephalitis.
Neuroimmunomodulation [Internet]. Karger Publishers; 2016 [cited 2022 Nov 8];23:278–86. Available
from: https://www.karger.com/Article/FullText/452171
32. Rahali N, Bahloul C. Induction of Cross-Reacting Antibodies Against the COVID-19 by BCG
Vaccination in the Mouse Model. Curr Microbiol [Internet]. Springer; 2022 [cited 2022 Nov 8];79:1–6.
Available from: https://link.springer.com/article/10.1007/s00284-022-02971-w
33. Arts RJW, Moorlag SJCFM, Novakovic B, Li Y, Wang SY, Oosting M, et al. BCG Vaccination Protects
against Experimental Viral Infection in Humans through the Induction of Cytokines Associated with
Trained Immunity. Cell Host Microbe [Internet]. Cell Press; 2018 [cited 2021 Jan 30];23:89–100.e5.
Available from: https://pubmed.ncbi.nlm.nih.gov/29324233/
Page 14/22
34. Angulo M, Angulo C. Trained immunity against diseases in domestic animals. Acta Trop [Internet].
Acta Trop; 2022 [cited 2022 Oct 12];229. Available from:
https://pubmed.ncbi.nlm.nih.gov/35149041/
35. Jung K, Ha Y, Ha SK, Han DU, Kim DW, Moon WK, et al. Antiviral effect of Saccharomyces cerevisiae
β-glucan to swine influenza virus by increased production of interferon-γ and nitric oxide. J Vet Med
Ser B Infect Dis Vet Public Heal. 2004;51:72–6.
36. Tran HTT, Truong AD, Chu NT, Vu HN, Nguyen HT, Nguyen T, et al. Inhibition of African swine fever
virus replication by β-glucan. Open Vet J [Internet]. Open Vet J; 2022 [cited 2023 Jan 24];12:1027–34.
Available from: https://pubmed.ncbi.nlm.nih.gov/36650869/
37. Byrne KA, Tuggle CK, Loving CL. Differential induction of innate memory in porcine monocytes by β-
glucan or bacillus Calmette-Guerin. Innate Immun. 2020;
38. Vaz-Rodrigues R, Ferreras-Colino E, Ugarte-Ruíz M, Pesciaroli M, Thomas J, García-Seco T, et al.
Nonspecific protection of heat-inactivated Mycobacterium bovis against Salmonella Choleraesuis
infection in pigs. Vet Res [Internet]. BioMed Central; 2022;53:31. Available from:
https://doi.org/10.1186/s13567-022-01047-8
39. Ferreras-Colino E, Moreno I, Risalde MA, Sevilla I, Agulló-Ros I, Martinez-Camacho R, et al. Efecto
protector de Mycobacterium bovis inactivado por calor frente a Leishmnia. XXXIII Reun la Soc
Española Anat Patol Vet. 2022.
40. Contreras M, Kasaija PD, Merino O, De La Cruz-Hernandez NI, Gortazar C, De La Fuente J. Oral
Vaccination With a Formulation Combining Rhipicephalus microplus Subolesin With Heat
Inactivated Mycobacterium bovis Reduces Tick Infestations in Cattle. Front Cell Infect Microbiol
[Internet]. Front Cell Infect Microbiol; 2019 [cited 2022 Jan 26];9. Available from:
https://pubmed.ncbi.nlm.nih.gov/30881925/
41. Guo J, Hou L, Zhou J, Wang D, Cui Y, Feng X, et al. Porcine Circovirus Type 2 Vaccines: Commercial
Application and Research Advances. Viruses [Internet]. MDPI; 2022 [cited 2023 Jan 25];14:2005.
Available from: https://www.mdpi.com/1999-4915/14/9/2005/htm
42. Risco D, Fernández-Llario P, García-Jiménez WL, Gonçalves P, Cuesta JM, Martínez R, et al. Influence
of porcine circovirus type 2 infections on bovine tuberculosis in wild boar populations. Transbound
Emerg Dis. 2013;60:121–7.
43. Segalés J, Domingo M, Chianini F, Majó N, Domínguez J, Darwich L, et al. Immunosuppression in
postweaning multisystemic wasting syndrome affected pigs. Vet Microbiol [Internet]. Elsevier; 2004
[cited 2023 Mar 8];98:151–8. Available from: https://pubmed.ncbi.nlm.nih.gov/14741127/
44. Darwich L, Segalés J, Mateu E. Pathogenesis of postweaning multisystemic wasting syndrome
caused by Porcine circovirus 2: An immune riddle. Arch Virol [Internet]. Arch Virol; 2004 [cited 2023
Mar 8];149:857–74. Available from: https://pubmed.ncbi.nlm.nih.gov/15098103/
45. Beltrán-Beck B, Ballesteros C, Vicente J, De La Fuente J, Gortázar C. Progress in oral vaccination
against tuberculosis in its main wildlife reservoir in Iberia, the Eurasian wild boar. Vet Med Int.
2012;2012.

Page 15/22
46. Beach NM, Meng XJ. Efficacy and future prospects of commercially available and experimental
vaccines against porcine circovirus type 2 (PCV2). Virus Res [Internet]. Elsevier B.V.; 2012;164:33–42.
Available from: http://dx.doi.org/10.1016/j.virusres.2011.09.041
47. Xu XG, Zhao HN, Zhang Q, Ding L, Li ZC, Li W, et al. Oral vaccination with attenuated Salmonella
enterica serovar Typhimurium expressing Cap protein of PCV2 and its immunogenicity in mouse and
swine models. Vet Microbiol [Internet]. Elsevier B.V.; 2012;157:294–303. Available from:
http://dx.doi.org/10.1016/j.vetmic.2012.01.008
48. Wang L, Zhao D, Sun B, Yu M, Wang Y, Ru Y, et al. Oral vaccination with the porcine circovirus type 2
(PCV-2) capsid protein expressed by Lactococcus lactis induces a specific immune response against
PCV-2 in mice. J Appl Microbiol. 2020;128:74–87.
49. Bucarey SA, Noriega J, Reyes P, Tapia C, Sáenz L, Zuñiga A, et al. The optimized capsid gene of
porcine circovirus type 2 expressed in yeast forms virus-like particles and elicits antibody responses
in mice fed with recombinant yeast extracts. Vaccine. 2009;27:5781–90.
50. Nainys J, Lasickiene R, Petraityte-burneikiene R, Dabrisius J, Lelesius R, Sereika V, et al. Generation in
yeast of recombinant virus-like particles of porcine circovirus type 2 capsid protein and their use for a
serologic assay and development of monoclonal antibodies. BMC Biotechnol [Internet]. BMC
Biotechnol; 2014 [cited 2023 Jan 31];14. Available from:
https://pubmed.ncbi.nlm.nih.gov/25487652/
51. Chen P, Zhang L, Chang N, Shi P, Gao T, Zhang L, et al. Preparation of virus-like particles for porcine
circovirus type 2 by YeastFab Assembly. Virus Genes [Internet]. Virus Genes; 2018 [cited 2023 Jan
31];54:246–55. Available from: https://pubmed.ncbi.nlm.nih.gov/29417333/
52. Patterson R, Eley T, Browne C, Martineau HM, Werling D. Oral application of freeze-dried yeast
particles expressing the PCV2b Cap protein on their surface induce protection to subsequent PCV2b
challenge in vivo. Vaccine [Internet]. Elsevier Ltd; 2015;33:6199–205. Available from:
http://dx.doi.org/10.1016/j.vaccine.2015.10.003
53. Garrido JM, Sevilla IA, Beltrán-Beck B, Minguijón E, Ballesteros C, Galindo RC, et al. Protection against
tuberculosis in eurasian wild boar vaccinated with heat-inactivated mycobacterium bovis. PLoS One
[Internet]. PLoS One; 2011 [cited 2021 Jan 15];6. Available from:
https://pubmed.ncbi.nlm.nih.gov/21935486/
54. Fort M, Sibila M, Nofrarías M, Pérez-Martín E, Olvera A, Mateu E, et al. Porcine circovirus type 2
(PCV2) Cap and Rep proteins are involved in the development of cell-mediated immunity upon PCV2
infection. Vet Immunol Immunopathol [Internet]. Vet Immunol Immunopathol; 2010 [cited 2021 Feb
5];137:226–34. Available from: https://pubmed.ncbi.nlm.nih.gov/20566220/
55. Fort M, Olvera A, Sibila M, Segalés J, Mateu E. Detection of neutralizing antibodies in postweaning
multisystemic wasting syndrome (PMWS)-affected and non-PMWS-affected pigs. Vet Microbiol.
Elsevier; 2007;125:244–55.
56. Thomas J, Infantes-Lorenzo JA, Moreno I, Cano-Terriza D, de Juan L, García-Bocanegra I, et al.
Validation of a new serological assay for the identification of Mycobacterium tuberculosis complex-

Page 16/22
specific antibodies in pigs and wild boar. Prev Vet Med [Internet]. Elsevier; 2019;162:11–7. Available
from: https://doi.org/10.1016/j.prevetmed.2018.11.004
57. Chianini F, Majó N, Segalés J, Domínguez J, Domingo M. Immunohistochemical characterisation of
PCV2 associate lesions in lymphoid and non-lymphoid tissues of pigs with natural postweaning
multisystemic wasting syndrome (PMWS). Vet Immunol Immunopathol. 2003;94:63–75.
58. Villar M, Pacheco I, Mateos-Hernández L, Cabezas-Cruz A, Tabor AE, Rodríguez-Valle M, et al.
Characterization of tick salivary gland and saliva alphagalactome reveals candidate alpha-gal
syndrome disease biomarkers. Expert Rev Proteomics [Internet]. Taylor & Francis; 2021;18:1099–116.
Available from: https://doi.org/10.1080/14789450.2021.2018305
59. Bates D, Mächler M, Bolker BM, Walker SC. Fitting Linear Mixed-Effects Models Using lme4. J Stat
Softw [Internet]. American Statistical Association; 2015 [cited 2023 Apr 17];67:1–48. Available from:
https://www.jstatsoft.org/index.php/jss/article/view/v067i01
60. Wickham H. ggplot2. ggplot2. Springer New York; 2009;
61. Kasaija PD, Contreras M, Kabi F, Mugerwa S, Garrido JM, Gortazar C, et al. Oral vaccine formulation
combining tick Subolesin with heat inactivated mycobacteria provides control of cross-species cattle
tick infestations. Vaccine [Internet]. Elsevier Ltd; 2022;40:4564–73. Available from:
https://doi.org/10.1016/j.vaccine.2022.06.036
62. Segalés J. Best practice and future challenges for vaccination against porcine circovirus type 2.
Expert Rev Vaccines. 2015;14:473–87.
63. Martelli P, Saleri R, Ferrarini G, De Angelis E, Cavalli V, Benetti M, et al. Impact of maternally derived
immunity on piglets’ immune response and protection against porcine circovirus type 2 (PCV2) after
vaccination against PCV2 at different age. BMC Vet Res [Internet]. BMC Vet Res; 2016 [cited 2022
Feb 7];12. Available from: https://pubmed.ncbi.nlm.nih.gov/27170186/
64. Kixmöller M, Ritzmann M, Eddicks M, Saalmüller A, Elbers K, Fachinger V. Reduction of PMWS-
associated clinical signs and co-infections by vaccination against PCV2. Vaccine [Internet]. Vaccine;
2008 [cited 2022 Apr 7];26:3443–51. Available from: https://pubmed.ncbi.nlm.nih.gov/18513842/
65. Martelli P, Ferrari L, Morganti M, De Angelis E, Bonilauri P, Guazzetti S, et al. One dose of a porcine
circovirus 2 subunit vaccine induces humoral and cell-mediated immunity and protects against
porcine circovirus-associated disease under field conditions. Vet Microbiol [Internet]. Vet Microbiol;
2011 [cited 2022 Feb 7];149:339–51. Available from: https://pubmed.ncbi.nlm.nih.gov/21216540/
66. Shen HG, Beach NM, Huang YW, Halbur PG, Meng XJ, Opriessnig T. Comparison of commercial and
experimental porcine circovirus type 2 (PCV2) vaccines using a triple challenge with PCV2, porcine
reproductive and respiratory syndrome virus (PRRSV), and porcine parvovirus (PPV). Vaccine
[Internet]. Vaccine; 2010 [cited 2023 Jan 31];28:5960–6. Available from:
https://pubmed.ncbi.nlm.nih.gov/20637768/
67. Segalés J, Rosell C, Domingo M. Pathological findings associated with naturally acquired porcine
circovirus type 2 associated disease. Vet Microbiol [Internet]. Vet Microbiol; 2004 [cited 2021 Oct 26].
p. 137–49. Available from: https://pubmed.ncbi.nlm.nih.gov/14741126/

Page 17/22
68. Segalés J. Porcine circovirus type 2 (PCV2) infections: Clinical signs, pathology and laboratory
diagnosis. Virus Res [Internet]. Elsevier B.V.; 2012;164:10–9. Available from:
http://dx.doi.org/10.1016/j.virusres.2011.10.007
69. Kristensen CS, Baadsgaard NP, Toft N. A meta-analysis comparing the effect of PCV2 vaccines on
average daily weight gain and mortality rate in pigs from weaning to slaughter. Prev Vet Med
[Internet]. Prev Vet Med; 2011 [cited 2023 Mar 8];98:250–8. Available from:
https://pubmed.ncbi.nlm.nih.gov/21239076/
70. da Silva N, Carriquiry A, O’Neill K, Opriessnig T, O’Connor AM. Mixed treatment comparison meta-
analysis of porcine circovirus type 2 (PCV2) vaccines used in piglets. Prev Vet Med [Internet]. Prev
Vet Med; 2014 [cited 2023 Mar 8];117:413–24. Available from:
https://pubmed.ncbi.nlm.nih.gov/25457512/
71. Seo HW, Han K, Park C, Chae C. Clinical, virological, immunological and pathological evaluation of
four porcine circovirus type 2 vaccines. Vet J [Internet]. Vet J; 2014 [cited 2023 Jan 31];200:65–70.
Available from: https://pubmed.ncbi.nlm.nih.gov/24618398/
72. Fort M, Animals S, Aut U, Mateu E, Segal J. Characterization of immune responses to porcine
circovirus type 2 (PCV2) infection and vaccination in pigs. PhD Thesis. 2009;2.
73. Wainberg MA, Israel E. Enhancement of avian sarcoma virus-induced tumor growth after
pretreatment with BCG. Infect Immun [Internet]. Infect Immun; 1978 [cited 2022 Feb 7];22:328–33.
Available from: https://pubmed.ncbi.nlm.nih.gov/215547/
74. Douglas JM, Vontver LA, Stamm WE, Reeves WC, Critchlow C, Remington ML, et al. Ineffectiveness
and toxicity of BCG vaccine for the prevention of recurrent genital herpes. Antimicrob Agents
Chemother [Internet]. American Society for Microbiology (ASM); 1985 [cited 2023 Jan 27];27:203.
Available from: /pmc/articles/PMC176238/?report = abstract
75. de Bree LCJ, Koeken VACM, Joosten LAB, Aaby P, Benn CS, van Crevel R, et al. Non-specific effects of
vaccines: Current evidence and potential implications. Semin Immunol. 2018;39:35–43.

Figures

Page 18/22
Figure 1

Experimental design. Sixteen 21 days-old Landrace x Large White hybrid female piglets were randomly
assigned to the immunized group (n=8), which received two oral doses of HIMB with an interval of 3
weeks, or the control group (n=8), which received PBS instead of the immunostimulant. Twenty-one days
after the second immunization dose, all the animals of both groups were infected by intranasal
inoculation with 105 TCID 50 of PCV-2. Twenty-one days after infection, all animals were euthanized and
necropsied.

Page 19/22
Figure 2

Temperature and body weight increments. Mean ± SE of body temperature (A) and body weight (B) in
immunized and control groups of animals infected with PCV-2.

Figure 3

Viral burden and antibody S/P ratio of PCV-2. A) PCV-2 DNA copies/mL of serum by individuals from
immunized and control groups. A cutoff value of 104 PCV-2 DNA copies/mL (black dashed line) was
considered the quantification limit of the PCV-2 qPCR. B) Mean ± SE of anti-PCV2 antibody S/P ratio by
group at each time point after PCV2 infection in immunized and control groups. An S/P ratio of 0.29
(black dashed line) or greater was considered seropositive for anti-PCV-2 antibodies.

Page 20/22
Figure 4

PCV-2 lesions at 21 days post-infection. PCV-2-infected pigs from both groups showed similar lesions,
presenting a moderate congestion and lymphadenomegaly, e.g. mediastinal lymph node (LN) (A). Mild
infiltrating of histiocytes and multinucleated giant cells was observed in the mandibular and pulmonary
LNs of some animals from both groups (B), associated to the presence of PCV-2 antigen (B, inset). Both,
immunized and control groups, also presented mild to moderate interstitial pneumonia (C), mainly
characterized by mononuclear leukocyte infiltrates in the pulmonary parenchyma (D). Hematoxylin and
eosin stain.

Page 21/22
Figure 5

Score of histopathological findings and antigen detection associated to PCV-2 infection. Mean of the
score for each microscopic lesion and PCV-2 antigen detection in mandibular (A) and pulmonary
(tracheobronchial and mediastinal) lymph nodes (B), tonsil (C) and lung (D). Lesions were graded by
score as absent or 0% (0), mild or 1% to 30% of the parenchyma affected (1), moderate or 30% to 70%
affected (2), and severe or 70% to 100% affected (3). PCV-2 antigen detection by immunohistochemistry
was graded by score as low (1), moderate (2) and intense (3) amount of antigen.

Page 22/22

You might also like