You are on page 1of 18

Veterinary Quarterly

2023, VOL. 43, NO. 1, 1–18


https://doi.org/10.1080/01652176.2023.2234430

Mechanisms of circovirus immunosuppression and pathogenesis with a


focus on porcine circovirus 2: a review
Enikő Fehéra,b , Ferenc Jakabb and Krisztián Bányaia,c
Veterinary Medical Research Institute, Budapest, Hungary; bNational Laboratory of Virology, Szentágothai Research Centre,
a

University of Pécs, Pécs, Hungary; cDepartment of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest,
Hungary

ABSTRACT ARTICLE HISTORY


Certain pathogens, due to their adverse effects on the immune reaction, aggravate the course Received 21 September
of concomitant heterologous infections. Here we summarize mechanisms by which circoviruses, 2022
including the most studied porcine circovirus 2, and other mammalian and avian circoviruses, Accepted 3 July 2023
trigger their own replication and confound the hosts’ immune response. At different stages of KEYWORDS
infection, from latent state to disease induction, these viruses markedly influence the cellular Pig; porcine; circovirus;
signaling pathways. Circoviruses have been found to interfere with interferon and PCV2;
proinflammatory cytokine producing and responsive pathways. Apoptotic processes, altered immunosuppression;
cellular transport and constraint of the mitotic phase all support the viral replication. The pathogenesis; review
cytokine imbalance and lymphocyte depletion, thus the impaired immunity, favors invasion of
super- or co-infecting agents, which in concert with circoviruses induce illnesses with increased
severity. The information summarized in this review point out the diversity of host and viral
factors involved in the mechanisms of disease progression during circovirus infections.

1. Introduction IFN molecules as well as the proinflammatory


pathways act in concert and ensure feedback to each
The formation of an antiviral state in host cells is the to restrict viral propagation in and between suscep-
result of sequential steps of variable cascade mecha- tible cells. To by-pass this fine-tuned network, viruses
nisms and interactions between molecular networks, evolved countermeasures at variable points by tar-
which activate the innate immune response as a first geting and interacting with host-origin molecules
step. A key functional component of the innate (Garcia-Sastre 2017; Rojas et al. 2021). From an
immunity is the initiation of cytokine expression. immunological view, the general presentation of the
Interferons (IFNs) constitute a class of cytokines that virus-host interactions goes beyond the scope of this
are released in response to pathogens in humans review. Here we attempt to summarize how porcine
and animals (Schneider et al. 2014; Rojas et al. 2021; circovirus type 2 (PCV2), an immunosuppressive
Walker et al. 2021). IFNs mediate autocrine and para- self-replicating DNA virus with minimal genome and
crine signaling that ultimately contributes to expres- coding capacity, disturbs cellular processes in order
sion of IFN stimulated genes (ISGs). ISGs encode to diminish the innate antiviral responses of host
proteins with direct and indirect antiviral effects that cells and antagonize the cytokine signaling, and how
inhibit different steps of the viral cell cycle, facilitate these actions promote co-infective pathogens to
antigen recognition and presentation, as well as multiply.
secretion of distinct classes of cytokines, or stabilize
elements of the signaling pathways. IFNs activate
immune cells (NK cells, natural killer cells; macro- 2. Antiviral pathways of DNA viruses
phages) and components of the adaptive immunity
(Schneider et al. 2014; Rojas et al. 2021). Another piv- The viral nucleic acids and proteins are perceived by
otal point is the stimulation and fine regulation of variable host pattern recognition receptors (PRRs)
proinflammatory cytokine secretion to activate the depending on the composition of the viral nucleic
adaptive immunity in which transcription factor acid and on the phase of the viral life cycle (Ablasser
nuclear factor κB (NF-κB)-mediated signaling has a and Hur 2020; Rojas et al. 2021; Walker et al. 2021).
central role (Driessler et al. 2004; Cao et al. 2006; PRRs involve the cell membrane or endosome-associ-
Ablasser and Hur 2020; Rojas et al. 2021). ated Toll-like receptors (TLRs), the RIG-I-like receptors

CONTACT Enikő Fehér feher.eniko@vmri.hu Veterinary Medical Research Institute, Hungária krt. 21, H-1143 Budapest, Hungary.
© 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/),
which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited. The terms on which this article
has been published allow the posting of the Accepted Manuscript in a repository by the author(s) or with their consent.
2 E. FEHÉR ET AL.

(e.g. RIG-I, retinoic acid-inducible gene-I receptor; with IRF9 constitute the transcription factor ISGF3.
MDA5, melanoma differentiation-associated gene 5 ISGF3 translocates to the nucleus, where it interacts
receptor) and the cytosolic GMP-AMP synthase with IFN-stimulated response element (ISRE) pro-
(cGAS) (Li et al. 2013; Ablasser and Hur 2020; moter sequences of ISGs (Lee and Ashkar 2018).
Eaglesham and Kranzusch 2020; Rojas et al. 2021; After binding to its receptor, IFN-γ activates Jak1/
Walker et al. 2021). Jak2, and controls the expression of further regula-
Activation of RIG-I and MDA-5 molecules lead to tory elements (i.e. transcription factors) by phosphor-
interaction with the mitochondrial antiviral adaptor ylated STAT1 homodimers (Schroder et al. 2004).
protein (MAVS), a protein located in the intracellular Beside specific heterodimeric receptors, type III IFNs
membranes of mitochondria, endoplasmatic reticu- also uses the interleukin (IL)-10 receptor 2 to induce
lum (ER) and peroxisome (Ablasser and Hur 2020; signal transduction via the JAK/STAT cascade (Stanifer
Rojas et al. 2021). MAVS activates inhibitor of kap- et al. 2020). In addition to the classical ones, a num-
pa-B kinase epsilon (IKKε) and TANK binding kinase 1 ber of distinct pathways are initiated by IFNs with
(TBK1), two enzymes that phosphorylate either inter- contribution of variable types of STATs, IRFs and
feron regulatory factor (IRF) 3 or IRF7. The homodi- kinases (Schroder et al. 2004; Lee and Ashkar 2018;
mers of IRF3 and IRF7 proteins translocate to the Stanifer et al. 2020).
nucleus and initiate transcription of Type I (IFN-α and
IFN-β) and Type III (IFN-λ) IFNs (Ablasser and Hur
2020; Rojas et al. 2021). Type I and Type III IFNs stim-
3. Introduction to circoviruses
ulate pathways leading to an antiviral state of cells
(Lee and Ashkar 2018; Rojas et al. 2021; Walker et al. Circoviruses (Circovirus genus, Circoviridae family) are
2021). Type II IFN (IFN-γ) production is predominantly small, non-enveloped, icosahedral viruses comprising
triggered by Type I IFNs (Lee and Ashkar 2018). IFN-γ a circular ssDNA genome of ~1600–2200 nt in length
plays a key role in antigen presentation and macro- (Rosario et al. 2017; Feher et al. 2022). The ambisense
phage activation, and as an immunomodulatory and genome contains two main open reading frames
proinflammatory protein it contributes to the activa- (ORFs) encoding one or more replication-associated
tion of the adaptive immune system (Lee and Ashkar proteins (Reps, ORF1) on the viral strand and capsid
2018; Rojas et al. 2021; Walker et al. 2021). protein (Cap, ORF2) on the complementary, replica-
The expression of proinflammatory cytokines is tive intermediate DNA strand (Lv et al. 2014; 2015;
mostly induced by MAVS via the NF-κB pathway Rosario et al. 2017). In the absence of virus-encoded
(Driessler et al. 2004; Cao et al. 2006; Ablasser and DNA polymerase, the replication of circoviruses
Hur 2020; Rojas et al. 2021). The inactivated form of depends on the cell cycle and is linked to the
the most abundant mammalian NF-κB p65/p50 het- nucleus; in susceptible mitotic cells, a large amount
erodimer is associated with the IκB (NF-κB inhibitor) of nascent virus particles is produced (Tang et al.
inhibitory complex. Phosphorylation of IκB by IκB 2013; Saikumar and Das 2019). Circoviral proteins,
kinase (IKK), followed by polyubiquitination and deg- including Cap, Rep, ORF3, ORF4 and ORF5 may sup-
radation, activates NF-κB that acts as transcription port viral replication interacting with the viral DNA
factor and transactivate the promoter of the targeted and regulating cellular proteins (Lv et al. 2014; 2015;
genes through the p65 protein (Driessler et al. 2004; Rosario et al. 2017).
Cao et al. 2006). A wide variety of circoviruses has been identified
The connection of viral DNA with cGAS triggers in vertebrates and insects, often without association
the catalytic activity of the enzyme. As a result, cGAS with pathogenic changes or clinical signs (Rosario
synthesizes cyclic GMP-AMP (cGAMP) that activates et al. 2017). However, some avian and mammalian
STING (stimulator of interferon genes) dimers within circoviruses are considered of high veterinary impor-
the ER (Ablasser and Hur 2020; Eaglesham and tance, which comes from their capacity to induce
Kranzusch 2020; Rojas et al. 2021). Oligomerized marked immunosuppression. Beside the direct patho-
STINGs translocate from the ER to the Golgi appara- genic role, the immunosuppressive effect together
tus and recruit TBK1 for downstream signaling. At with co-infecting agents and secondary infections
this point, the processes overlap with pathways con- will determine the outcome of the infection caused
nected to RIG-I and MDA5 (Ablasser and Hur 2020; by these circoviruses (Abadie et al. 2001; Schmidt
Eaglesham and Kranzusch 2020; Rojas et al. 2021). et al. 2008; Saikumar and Das 2019; Kroeger
via TBK1, STING stimulates the NF-κB pathway as et al. 2022).
well (Ablasser and Hur 2020; Eaglesham and Avian circoviruses, including beak and feather dis-
Kranzusch 2020). ease virus (BFDV), goose circovirus (GoCV), duck cir-
Type I IFNs induce the dimerization of IFN recep- covirus (DuCV), pigeon circovirus (PiCV), finch
tor 1 and 2 that elicit the IFN response. The receptor circovirus and canary circoviruses, may generate leth-
complex, and the associated tyrosine kinase (Tyk) 2 argy, weight loss, as well as beak deformation, feath-
and Janus tyrosine kinase (Jak) 1 phosphorylate each ering issues and gastrointestinal disorders (Ritchie
other and signal transducer and activator of tran- et al. 1989; Soike et al. 1999; Todd et al. 2001;
scription (STAT) proteins (Rojas et al. 2021). In the Shivaprasad et al. 2004; Raue et al. 2005; Todd et al.
canonical IFN response pathway the phosphorylated 2007; Guo et al. 2011; Circella et al. 2014; Feher et al.
STAT1 and STAT2 form heterodimers and together 2022). Microscopic lesions associated with avian
Veterinary Quarterly 3

circoviruses are lymphocyte and macrophage infiltra- and PCV3, but it can be the consequence of the
tion, atrophy and necrotic areas in the lymphoid and often diagnosed co-infection of PCVs and other
non-lymphoid tissues; however, subclinical infection pathogens (Wang et al. 2022a). Some reports sug-
without clinical signs and lesions may also occur gest that PCV4 might be pathogenic alone; the virus
(Abadie et al. 2001; Hong et al. 2018; Wang et al. recovered from a cloned genome induced inflamma-
2022b). Avian circoviruses are widely distributed in tion and deformities in the spleen, lung, kidney, liver
wild and domestic fowl, and are regularly diagnosed and lymph nodes of inoculated piglets (Niu
together with other avian pathogens. This co-occur- et al. 2022).
rence can worsen the course of infection and makes PCV1, PCV2 and PCV3 are widely distributed in
the etiological role of circoviruses in disease develop- pigs and wild boars. Furthermore, PCV2 has also
ment difficult to determine (Kozdruń et al. 2012; been identified in cattle, dog, deer, chamois, mouse,
Stenzel and Koncicki 2017; Hong et al. 2018; Kaszab as well as arthropods (such as ticks and mosquitoes)
et al. 2020; Sahindokuyucu et al. 2022). (Iizuka et al. 1989; Cao et al. 2018; Saikumar and Das
At present, four species of porcine circoviruses 2019; Yang et al. 2019; Zhang et al. 2020a; Amoroso
(PCVs) are distinguished. PCV1 was discovered nearly et al. 2021; Cui et al. 2022; Fanelli et al. 2022; Hu
50 years ago in a porcine kidney cell culture, PK-15, et al. 2022; Igriczi et al. 2022; Kroeger et al. 2022;
and it is considered an apathogenic virus (Tischer Luka et al. 2022; Vargas-Bermudez et al. 2022).
et al. 1974; Iizuka et al. 1989). On the contrary, PCV2 Reports about the less prevalent PCV4 in swine, wild
induces diseases in their host species (Iizuka et al. boars and cattle originate from Southeast Asia, China
1989; Nayar et al. 1997; Saikumar and Das 2019). and South Korea (Wu et al. 2022; Xu et al. 2022;
PCV2 infection remains subclinical, or manifests in Wang et al. 2022a).
variable forms commonly referred to as PCV- Canine circovirus (CaCV) is also characterized as a
associated diseases (PCVAD). PCVAD involves widely distributed pathogenic mammalian circovirus
post-weaning multisystemic wasting syndrome that may cause hemorrhagic gastrointestinal and
(PMWS), porcine respiratory disease complex, porcine respiratory diseases of dogs (Anderson et al. 2017;
dermatitis and nephropathy syndrome and reproduc- Dankaona et al. 2022). As for avian and porcine cir-
tive and enteric disease forms (Iizuka et al. 1989; coviruses, co-infections contribute probably to the
Nayar et al. 1997; Allan et al. 1998; Ellis et al. 1998; development or exacerbation of CaCV related dis-
Saikumar and Das 2019). eases (Anderson et al. 2017; Dankaona et al. 2022).
PCV2 causes a wide-range of clinical signs depend- The lack of cell culture systems is detrimental to
ing on the condition of the host. The PCV2-asociated the exploration of the pathomechanisms for the vast
immunosuppression is characterized by lymphope- majority of circoviruses. However, PCV1, PCV2 and
nia, lymphoid cell (B- and T-cell) depletion and PCV3 can be propagated both in lymphoid and
altered cytokine production (Darwich et al. 2003b; non-lymphoid cell cultures, a finding that permits
Saikumar and Das 2019). PCV2 is most often diag- insight into molecular aspects of virus-host interac-
nosed with concurrent pathogens, including classical tions (Tu et al. 2015; Kroeger et al. 2022).
swine fever virus (CSFV), pseudorabies virus (PRV),
porcine reproductive and respiratory syndrome virus
(PRRSV) and porcine parvovirus (PPV) (Iizuka et al.
4. Target cells of circoviruses
1989; Saikumar and Das 2019). Due to the interfer-
ence with the host immune system, PCV2 and con- The primary sites of active replication remain to be
current porcine pathogens could provoke more clarified for many circoviruses (Abadie et al. 2001;
severe disease together than those separately (Rovira Schmidt et al. 2008; Steiner et al. 2008; Guo et al.
et al. 2002; Kim et al. 2006; Kekarainen et al. 2008b; 2011; Robino et al. 2014; Hong et al. 2018; Kroeger
Shi et al. 2010; Sinha et al. 2011; Gao et al. 2014a; et al. 2022). Viral components of PCV2 were identi-
Ouyang et al. 2019; Opriessnig et al. 2020). fied in monocytic cells and other blood cells, in lym-
The most recently identified PCV species, PCV3 phoid tissues, as well as in endothelial cells and
and PCV4, have been also linked to variable dis- epithelial cells of different organs (such as liver, lung,
eases. PCV3 is commonly detected in swine with kidney) (Rosell et al. 1999; Darwich et al. 2004). Pig
acute porcine dermatitis and nephropathy syn- fetuses were shown to carry PCV2 in a latent form in
drome-like signs, reproductive failures, cardiac dis- the medulla of thymus and other lymphoid organs
eases and multisystemic inflammation (Phan et al. (Dong et al. 2015; Sydler et al. 2016). The virus causes
2016; Palinski et al. 2017). Histological findings NK-, T- and B-cell depletion in PCVAD, in part, by
include dermatitis and epidermitis, necrotizing vas- direct destruction of the immune cells and by other,
culitis, pneumonia, glomerulonephritis, granuloma- currently not fully understood mechanisms (Rosell
tous lymphadenitis, myocarditis and cardiac et al. 1999; Darwich et al. 2004).
arteriolitis (Phan et al. 2016; Palinski et al. 2017; Evidence indicate that PCV2 primarily infects
Jiang et al. 2020; Sirisereewan et al. 2022; Yang et al. monocytic cells, including monocytes, macrophages,
2022). Lymphocyte and macrophage infiltration is dendritic cell (DC) precursors, myeloid DCs and plas-
usually observed in the affected tissues (Phan et al. macytoid DCs (pDC). These cells are not the main
2016; Kim et al. 2018; Jiang et al. 2020). PCV4 is virus factories but promote dissemination of the
known from similar diseases as described for PCV2 virus into variable tissues (Figure 1) (Gilpin et al.
4 E. FEHÉR ET AL.

Figure 1. Putative course of circovirus infection through the example of porcine circovirus 2 (PCV2). PCV2 infects primarily
monocytic cells that aid dissemination of the virus throughout the body, allowing infection of multiple cell types. The impaired
immune functions, provoked by the virus, together with IFNs elicited by co-infective agents, promote initiation of PCV2 repli-
cation in permissive cells in the early phase of infection. Cap (capsid), Rep (replication-associated protein) and open reading
frame (ORF) 4 proteins contribute to activation of virus replication, as well as maintenance of anti-apoptotic state. In the late
stage of infection excessive PCV2 replication is associated with apoptotic processes that is supported by upregulation of Cap,
Rep, ORF3 and ORF5 proteins of the virus. Enhancement of immunosuppression favors replication of other pathogens that may
lead to development of severe diseases.

2003; Vincent et al. 2003; 2005; Chang et al. 2006; maturation, immune cell (NK, T and B lymphocyte)
Darwich and Mateu 2012). PCV2 binds heparan sul- activation and direct antiviral effect (Vincent et al.
fate, chondroitin sulfate B and dermatan sulfate 2005; 2007; Ablasser and Hur 2020). PCV2 inhibits
receptors and enters the monocytic cells via clath- pathogen recognition and, with the dissemination
rin-mediated endocytosis, where the virus persists in into a wide range of organs, it facilitates the deteri-
latency (Misinzo et al. 2005; Vincent et al. 2005; oration of tissues against other pathogens (Figure 1)
Misinzo et al. 2006; Wei et al. 2018; Huang et al. (Vincent et al. 2005; 2007).
2021). The amino acid characteristics and distribution PCV2 affects the differentiation of immune cells
of the Cap has an impact on the PCV2 binding to indirectly through cytokine changes elicited from
the receptors, thus the uptake of distinct virus strains DCs. PCV2 infection and PCV2/PRRSV co-infection of
shows differences (Wei et al. 2019; Huang et al. DCs induce transforming growth factor beta 1 (TGF-
2021). In contrast to monocytic cells, actin- and small β)-dependent regulatory T-cell (Treg) development. A
GTPase-regulated clathrin- and caveolin-independent similar mechanism was observed in experiments with
pathways support PCV2 internalization and replica- IPEC-J2 intestinal porcine enterocytes (Cecere et al.
tion in swine kidney and testicle epithelial cells, 2012). In these cells, the activation of NF-κB increased
while the virus enters T-lymphoblasts by various the transforming growth factor (TGF)-β level that
ways (Misinzo et al. 2009; Wei et al. 2019). PCV3 stimulated CD4+ T-cell differentiation into Treg cells
invades PK-15 cells through clathrin- and via extracellular signal-regulated kinase (ERK) phos-
dynamin-2-mediated endocytosis (Stenzel et al. 2019). phorylation (Liu et al. 2022). The increased amount
In cultured DCs, PCV2 alone does not influence of Treg cell leads to reduced responsiveness of the
cell differentiation, antigen processing and presenta- host immune system and to chronic infections
tion, or proinflammatory cytokine production (Maizels and Smith 2011; Liu et al. 2022).
(Vincent et al. 2005). The virus does not affect the PCV3 antigen/nucleic acid is commonly identified
IFN-α/tumor necrosis factor (TNF)-α induced DC in lymph nodes, intestines, cardiac cells, various
maturation, and major histocompatibility complex internal organs and in the dermis, as well as in
class II (MHCII) antigen presenting protein and inflammatory cells (presumably in macrophages) of
CD80/86 T-cell co-stimulatory molecule expression. the infected pigs (Phan et al. 2016; Palinski et al.
However, the virus inhibits IFN-α and TNF-α expres- 2017; Deim et al. 2019; Kroeger et al. 2022). Similarly,
sion induced by distinct swine pathogens (such as lymphoid cells and macrophages tested positive for
PRV, CSFV and transmissible gastroenteritis virus or CaCV nucleic acid by in situ hybridization (Dankaona
TGEV) or by ligands in pDCs that impairs DC et al. 2022). However, the role of these cells in the
Veterinary Quarterly 5

viral life cycle and in the pathogenesis is still in In addition to the viral DNA, virus-encoded pro-
question (Phan et al. 2016; Palinski et al. 2017; Kim teins also modulate the cellular immune mechanisms.
et al. 2018). PCV2 Cap-gC1qR (receptor of the globular head of
PiCV, DuCV, GoCV, and BFDV infections are also complement component 1q, also called C1QBP or
characterized with lymphocyte depletion, and with p32) interaction up- or downregulates variable sig-
marked atrophy and high apoptotic cell rate in the naling pathways early after the virus enters the cells
lymphoid tissues of the bursa of Fabricius, thymus, (Choi et al. 2015; Du et al. 2016). In PK-15 cells, PCV2
spleen and bone marrow (Raue et al. 2005; Schmidt activates the phosphoinositide 3-kinase/protein
et al. 2008; Guo et al. 2011; Robino et al. 2014; kinase B (PI3K/Akt) pathway by replication indepen-
Huang et al. 2017b; Hong et al. 2018). Circoviruses dent manner, probably at the step of virus attach-
and virus-associated inclusion bodies are often ment/cell entry that support viral replication,
detected in multiple tissues of the avian hosts, which translation, and maintenance of an anti-apoptotic
is most pronounced in lymohoid tissues, chiefly in state via the cleavage of poly-ADP ribose polymerase
the bursa of Fabricius. Since inclusion bodies first (PARP) and caspase-3 (Figure 2) (Wei et al. 2012).
appear in macrophages of this organ, PiCV is sug- Furthermore, HSP70, a cellular chaperon also inhibits
gested to have a primary bursal tropism (Abadie caspase-3 at this stage of infection in monocytic cells
et al. 2001; Schmidt et al. 2008). During progression (Liu et al. 2013). PCV2 Cap interacts with nucleosome
of the infection, bursal atrophy and lymphocyte assembly protein 1-like 4 (NAP1L4) protein; the
depletion can be observed, and mononuclear cells silencing of NAP1L4 results in the upregulation of
loaded with inclusion bodies infiltrate other lym- Cap and Rep expression and enhances IFN-β expres-
phoid organs or tissues that begins to deteriorate sion via cGAS activation, a process that promotes
(Abadie et al. 2001; Stenzel et al. 2020). In the late virus replication (Wang et al. 2020).
stage of PiCV infection the host is affected by sec- The viral Cap modulates the cellular transport pro-
ondary and opportunistic infections as a likely conse- cesses that trigger virus replication. PCV2 Cap aug-
quence of suppressed immune functions (Abadie ments the phagocytic activity of macrophages via
et al. 2001). stabilization of cytoplasmic gC1qR and activation of
As the above-mentioned data imply, the main tar- PI3K (Choi et al. 2015). The gC1qR-Cap interaction
gets of circoviruses are monocytic cells. After dissem- promotes the phosphorylation of protein kinase C
ination and invasion of permissive cells, circoviruses (PKC)-δ and recruitment of p-PKC-δ that lead to the
obtain excessive replication and immune impairment rearrangement of nuclear lamina and facilitate the
that predisposes the host to be invaded by patho- viral egress in cultured cells. At later phase of infec-
gens, advocating disease development. tion, Jun N-terminal protein kinase (JNK) and ERK
signaling further enhance these processes (Wang
et al. 2019). PCV2 Cap binds and supports nuclear
transport of the virus binding dynein thus inducing
5. Immunomodulatory elements of circoviruses
α-tubulin acetylation (Cao et al. 2015). With down-
The genomic DNA of circoviruses has immunomodu- regulation of porcine makorin RING finger protein 1
latory effects in the infected cells. As demonstrated Cap avoids its own degradation and triggers PCV2
for UV-treated PCV2 in pDCs, the viral genomic DNA replication via induction of stress mechanisms and
influences the IFN production independent of viral apoptosis (Wang et al. 2018; Zhang et al. 2019). Both
replication, probably by unmethylated CpG motifs Cap and Rep antagonize the immune response at
(Hasslung et al. 2003; Vincent et al. 2007; Wikstrom several points that was investigated in cell culture
et al. 2007; Kekarainen et al. 2008a). CpG-ODNs based overexpression experiments (see next sections)
(cytosine-phosphorothioate-guanine synthetic oli- (Du et al. 2016; 2018; Wu et al. 2019).
godeoxynucleotides), that are often used to study As for PCV2, the Cap of PCV3 intersects variable
DNA motifs, act on the IFN expression via TLR depen- cellular signaling pathways. The Cap-G3BP1 (GTPase-
dent (e.g. TLR9) and independent pathways (Vincent activating protein-(SH3 domain)-binding protein 1)
et al. 2007; Wikstrom et al. 2007; Hansoongnern et al. interaction inhibits viral DNA recognition by cGAS
2022). The fact that incorporated tandem CpG-ODNs that hampers IFN-β production in HEK-293T cells
increase the PCV2 virus-like particle (VLP) vaccine (Zhang et al. 2020c). The Cap of both PCV2 and PCV3
efficacy underlines the importance of CpG motifs in interact with nucleolar phosphoprotein nucleophos-
modulation of cellular signals (Hansoongnern et al. min-1 that contributes to virus replication (Zhou
2022). In addition to the DNA sequence, the second- et al. 2020; Song et al. 2021). PCV3 Cap, but not Rep,
ary and tertiary structure of the circular genomic was described to induce IL-6 and TNF-α expression
DNA markedly influence the final action on the via the NF-κB pathway, possibly through the activa-
immune reaction (Wikstrom et al. 2007; Kekarainen tion of RIG-I/MDA5, as well as TLR-MyD88 pathways
et al. 2008a; 2008b; Balmelli et al. 2011). The purified in HEK-293T cells (Liu et al. 2021). In IFN-β treated
replicative dsDNA of PCV2 was found to induce cyto- PK-15 and HEK-293T cells PCV3 Cap inhibits tran-
skeletal rearrangements via the impairment of actin scription activation of ISRE, but phosphorylation and
polymerization, a mechanism that inhibits endocyto- heterodimerization of STAT1 and STAT2 are not
sis, secretion of cytokines as well as antigen presen- affected. Although Cap binds karyopheryn alpha
tation in DCs (Balmelli et al. 2011). (KPNA) 1, involved in nuclear protein import, this
6 E. FEHÉR ET AL.

Figure 2. Anti-apoptotic and pre-apoptotic processes in porcine circovirus 2 (PCV2) infection. In the early phase of infection,
PCV2 activates the PI3K and the NF-κB pathways, and upregulates HSP70 and PCV2 open reading frame (ORF) 4 expression
that inhibit pre-apoptotic processes and support virus replication. In the late phase the excessive PCV2 replication contribute
to expression of the ORF3 encoded protein, and to activation of NF-κB, p38-MAPK and JNK1/2 pathways. Moreover, the viral
protein of ORF5 induces endoplasmic reticulum stress (ERS). These together may further enhance virus replication, and trigger
autophagy and apoptotic processes in the infected cells.

interaction does not inhibit the translocation of the Ca2+ and reactive oxygen species (ROS) (Lv et al.
STAT complex to nucleus. It is suggested that PCV3 2015; Zhou et al. 2016; Lv et al. 2020). These pro-
Cap interacts with the transactivation domain of cesses activate the NF-κB pathway in PK-15 cells by
STAT2 that prevents ISGF3 binding to ISRE (Shen increasing IκBα phosphorylation and degradation,
et al. 2020). and p65 nuclear translocation (Figure 2) (Wei et al.
Although the protein encoded by ORF5 is a non- 2008; Zhang et al. 2013). This signaling seems to be
essential factor in respect to PCV2 replication, it sup- necessary for excessive replication and viral protein
ports the virus propagation by induction of expression, and for the enhancement of proinflam-
prolonged S-phase and arresting of cell proliferation matory processes, thus disease progression. Likewise,
in alveolar macrophage cells with the downregula- expression of proinflammatory proteins (IL-6, IL-8
tion of transmembrane glycoprotein NMB and cyclin and COX-2) can be induced by the NF-κB pathway in
A expression (Tang et al. 2013; Lv et al. 2015; Guo porcine alveolar macrophages (PAMs) (Lv et al. 2015).
et al. 2018). On the other hand, ORF5 protein, local- The ORF5-associated triggering of autophagy and
ized in the ER, enhances the replication of PCV2 by apoptosis proceed via the phosphorylation of PERK
inducing ER associated stress that leads to autoph- and eukaryotic initiation factor 2a (eIF2α) that acti-
agy, protein kinase RNA-like endoplasmic reticulum vate activating transcription factor 4 (ATF4) and C/
kinase (PERK)-mediated unfolded protein response EBP homologous protein (CHOP, an UPR component)
(UPR), and ultimately to apoptosis contributed by in PK-15 cells (Figure 2). Furthermore, it mediates
Veterinary Quarterly 7

the phosphorylation of adenosine monophos- (Xiang et al. 2012; Lv et al. 2014; 2015; Rosario et al.
phates-activated protein kinase and the activation of 2017). Apoptosis was linked to viral protein expres-
ERK1/2 that downregulates the cell survival support- sion in BFDV affected cells as revealed by TUNEL
ing and autophagy regulator mammalian target of assay and large caspase-1 positive cell rate in BFDV
rapamycin (mTOR) pathway (Figure 2) (Zhu et al. infected parrots (Robino et al. 2014). The load of
2012; Zhou et al. 2016; Lv et al. 2020). The mTOR is PiCV seems to correlate with the disease severity;
a target molecule of PCV3 as well; PCV3 Cap sup- the virus inhibits the humoral immunity and induces
ports autophagosome formation and induces auto- apoptosis of B lymphocytes (Stenzel et al. 2020).
phagy via inhibition of mTOR phosphorylation in Taken together, the circovirus nucleic acid, as well
HEK293T cells (Geng et al. 2020). Both Cap and Rep as the structural and non-structural proteins work in
of PCV2 utilize the PERK-eIF2α-ATF4-CHOP axis for concert with each other to take the control over the
upregulation of ER oxidoreductase and ROS genera- cells and to determine the course of infection.
tion that promote nucleocytoplasmic migration of
high-mobility group box 1 protein (HMGB1) in PK-15
cells. The decreased retention of nuclear HMGB1
6. Cytokine changes in circovirus infection
releases the HMGB1-bound viral DNA, allowing
higher virus replication rate (Sun et al. 2021). In order to evaluate the immune response to PCV2
The expression and stabilization of p53 may have infection, a number of studies investigated the
a central role in the above-mentioned processes (Liu changes of cytokine level in sera, tissues or isolated
et al. 2005; 2006; Wei et al. 2009; Pan et al. 2018; blood cells of healthy naïve and vaccinated pigs, and
Deng et al. 2022). The PERK-ROS axis promotes p53 in pigs diagnosed with subclinical infections or
upregulation in PCV2 inoculated PAMs and the accu- PCVAD. Furthermore, the modulatory effect of PCV2
mulated phosphorylated p53 leads to cell cycle arrest has been frequently studied in relation to co-infec-
at S-phase and enhances viral replication. It remains tion with other pathogens or vaccination. A huge
a question whether this phenomenon is associated variety of experimental design has been imple-
with ORF5 (Deng et al. 2022). In other experimental mented leading to diverse, sometimes controversial,
design, the prolongation of the S-phase has been results that are occasionally hard to interpret.
retrieved in PAMs by ORF5 (Lv et al. 2015; Zhou et al. Cytokine response to PCV2, PCV2 DNA, CpG-ODNs
2016; Lv et al. 2020). In addition, the excessive viral and viral proteins in blood cells were intensively
replication and translation downregulate HSP70 and examined, often giving dissimilar data in peripheral
upregulate the phospholipase C/inositol 1,4,5-tris- blood mononuclear cells (PBMC) and monocytic cell
phosphate receptor pathways leading to apoptotic cultures. In PBMC of PRV naïve pigs PCV2 induced
processes through p53 and caspase-3 activation, as negligible amount of IFN-α, while most of CpG-ODNs
well as Ca2+-release-mediated ER stress (Figure 2) used for PCV2 DNA analysis triggered its production.
(Pan et al. 2018; Wang et al. 2021a). In PBMC of PRV-immunized animals, PRV-induced
In the later phases of infection, PCV2 stimulates IFN-α production was inhibited by PCV2, but not by
p38-mitogen-activated protein kinase (MAPK) and CpG-ODNs (Wikstrom et al. 2007; Kekarainen et al.
JNK1/2 signal transductions linked to viral replication 2008a; 2008b). Regarding IFN-γ, neither PCV2 nor
processes. p38-MAPK and JNK1/2 pathways have a CpG-ODNs provoked alone the production of this
role in the transcription and translation of viral pro- cytokine, but a decrease of PRV induced IFN-γ mRNA
teins, release of progeny viruses, whilst triggering could be detected in PBMCs of PRV/PCV2 co-infected
apoptosis (Figure 2) (Wei et al. 2009). Similarly to pigs (Gao et al. 2014a). In the PBMCs of PRV immu-
ORF5, ORF3 is nonessential with regard to virus rep- nized pigs, PCV2 and most of the CpG-ODNs did not
lication, but it is considered an important element of stimulate the production of IFN-γ and IL-2, while sig-
PCV2 pathogenesis that triggers apoptosis of the nificant downregulation of the PRV induced IFN-γ
infected cells and thus virus proliferation (Liu et al. and IL-2 could be seen for both PCV2 and the CpG-
2005; 2006). p38-MAPK and JNK1/2 support the sta- ODNs (Kekarainen et al. 2008a; 2008b; Gao et al.
bilization of p53 that mediates PCV2 ORF3 protein 2014a). Decreased levels of IFN-γ mRNA have been
induced apoptosis (Liu et al. 2005; 2006; Wei et al. also detected in PBMC of PRRSV/PCV2 co-infected
2009). In contrast, ORF4 was found to be an piglets compared to the control groups (Shi et al.
anti-apoptotic viral protein. It inhibits the virus repli- 2010). IL-10 production is influenced also by PCV2.
cation in early phase of infection and decreases the The complete virus, but not CpG-ODNs, induced high
expression of the pre-apoptotic ORF3 protein (Figure level of this cytokine in PBMC of both naïve and PRV
2) (He et al. 2013; Gao et al. 2014b). immunized animals. In this mixed cell population,
The promotion of apoptotic processes was also the monocytic cell fraction may be the source of
described for avian circoviruses, but few details are IL-10 (Kekarainen et al. 2008a; 2008b; Gao et al.
known about the viral immunomodulatory elements 2014a). As IL-10 inhibitory experiments revealed,
(Raue et al. 2005; Schmidt et al. 2008; Guo et al. IL-10 can negatively affect IL-12 and IFN-γ levels in
2011; Robino et al. 2014; Huang et al. 2017b; Hong PBMCs of PRV immunized animals during PRV recall,
et al. 2018). Like the homologous protein of PCV2, while IL-2 inhibition seemed to be independent of
the product of DuCV ORF3 was found to contribute the IL-10 activation (Kekarainen et al. 2008a; 2008b;
to the viral pathogenesis via activation of apoptosis Ablasser and Hur 2020). VLPs did not provoke or
8 E. FEHÉR ET AL.

influence significantly the expression of these cyto- TNF-α, IL-8 and monocyte chemotactic protein-1 lev-
kines in PBMCs (Kekarainen et al. 2008a). els were observed in PCV2 infected alveolar macro-
In BMDCs (bone-marrow-derived dendritic cells), phages with simultaneous reduction of the
PCV2 and rep derived CpG-ODNs (but not PCV2 VLP) microbicidal capacity and phagocytic activity (Chang
inhibited IFN-α secretion during PRV challenge et al. 2006). Higher TNF-α levels were detected in
(Kekarainen et al. 2008a; 2008b). Furthermore, PCV2 alveolar macrophages in co-infection with PPV and
has been proved to downregulate IFN production PCV2, and in concavalin A stimulated PBMCs of
induced by CpG-ODNs, CSFV and TGEV in monocytic PRRSV-PCV2 co-infected pigs compared to the con-
cells (Vincent et al. 2007; Kekarainen et al. 2008a; Li trols (Kim et al. 2006; Shi et al. 2010). In in the same
et al. 2018; Ablasser and Hur 2020). The measure- time, decreased TNF-α expression was measured in
ment of IL-12 revealed that PCV2 and VLPs (but not PBMC of PRRSV-PCV2 co-infected pigs and in PCV2-
the used CpG-ODNs) induce its production in BMDCs. PRV inoculated PK-15 cell culture (Tu et al. 2015; Li
However, the upregulation of IL-10 expression, an et al. 2022). Elevated IL-1β expression has been mea-
important feature of PCV2, reduced IL-12 secretion sured in PMWS and in co-infections of PCV2 with
(Kekarainen et al. 2008a; 2008b). Overall, the varia- PRRSV or PRV, but the level of this cytokine was
tion seen in cytokine production of blood cells seems shown to depend on the phase of infection similarly
to depend on composition of the tested cell popula- to IL-8 (Darwich et al. 2003b; Sipos et al. 2004;
tion, on the stage of infection, on the vaccination Borghetti et al. 2013; Tu et al. 2015; Li et al. 2022).
status or the presence of co-infective agents. In gen- These alterations, that may be different in variable
eral, PCV2 alone does not increase significantly the tissues and disease status, disturb the overall cyto-
expression of IFNs, but decreases the IFN levels kine expression and hamper the microbe clearance,
induced by other agents, while IL-10 and IL-12 but attract macrophages and other target cells to
expression could be directly influenced by PCV2. the site of infection in order to support cell survival
VLPs and the variably constructed CpG-ODNs act in and the spread of PCV2 (Chang et al. 2006). The
a different way on cytokine expression, if compared impaired immunological reactivity exposes the host
to the complete virus. to secondary or opportunistic infections (Darwich
Numerous studies investigated the cytokine et al. 2003a; 2003b; Chang et al. 2006; Kim et al.
response to PCV2 infection without focusing on a 2006; Shi et al. 2010).
certain structural element of the virus. Although The regulation of cytokine levels relies on the
IFN-γ downregulation has been usually noted in cooperation of cytokines and cellular factors. As an
monocytic cells, in PBMCs or in internal organs both example, suppressor of cytokine signaling family 3
in subclinical and PCVAD cases, an increased IFN-γ (SOCS3) protein has been identified as a regulator of
expression was occasionally observed (Darwich et al. cytokine signaling that functions in the negative
2003a; 2003b; Li et al. 2022). In PCV2 inoculated gno- feedback to proinflammatory cytokine production.
tobiotic pigs the immune response showed marked TNF-α and IL-6 induce removal of IκBα bound to
variances with regard to the neutralizing antibody NF-κB, then NF-κB activation initiate gene expression
titer and IFN-γ expression of PBMCs even within an of proinflammatory cytokines (Yasukawa et al. 2003;
experiment. Neutralizing antibody and increased Hayden and Ghosh 2014). In PCV2 challenge, PBMCs
IFN-γ mRNA levels were shown in some cases with- of piglets with subclinical infections showed an
out detectable PCV2, while lower neutralizing anti- increased SOCS3 level, and inhibited IκBα degrada-
body and indifferent IFN-γ mRNA levels with variable tion and NF-κB signaling was detected. Likewise,
PCV2 titers were seen in others (Meerts et al. 2005b). SOCS3 level was elevated after PCV2 inoculation of
The IFN-γ mRNA level may only slightly change in PK-15 cells. SOC3 interacted with STAT3 (mediator of
animals vaccinated against PCV2 (Borghetti IL-6 signaling) and TNF-receptor-associated factor 2
et al. 2013). (mediator of TNF-α signaling), inhibiting IL-6 and
PCV2 and concomitant infectious agents may TNF-α-mediated IκBα degradation and NF-κB activa-
interfere with expression of multiple cytokines, tion (Zhu et al. 2016). It was concluded that anti-in-
including those responsible for T-cell differentiation flammatory reactions result in failure of immune
and homeostasis, such as IL-2 and IL-4, as well as the response to PCV2, supporting the change of PCV2
proinflammatory cytokines IL-6 and IL-12. These cyto- infection from the latent to an active state, while
kines are typically downregulated by PCV2, but upregulated proinflammatory cytokine production
increased levels have been detected as well depend- promotes progression of PCV2 infection (Zhu
ing on the type of examined cells and tissues, and et al. 2016).
the stage of infection (Sipos et al. 2004; Li et al. PCV1 and PCV2 have been investigated often
2022). Elevated levels of TNF-α and IL-8 were com- together in order to compare the cellular processes
monly measured during PCV2 infection (Darwich activated during the infections. Although, some cyto-
et al. 2003a; 2003b; Chang et al. 2006; Darwich et al. kine response could be measured in PCV1 inoculated
2008). However, in healthy pigs and severe PCVAD animals and cell cultures, these changes are not sig-
lower levels of IL-8 (a chemoattractant of blood cells) nificant when compared to controls (Kekarainen
have been found than in animals with moderate et al. 2008b; Du et al. 2016; 2018; Wang et al. 2022c).
signs and lower PCV2 titer (Darwich et al. 2003a; Similarly to PCV2, PCV3 infection can markedly alter
2003b; Borghetti et al. 2013; Lv et al. 2015). Increased the cytokine response, but few data are currently
Veterinary Quarterly 9

available. Elevated IL-8 and undetectable IL-1β, IL-4, cGAS expression, STING dimerization and perinuclear
IL-10 levels were reported in serum of pigs showing translocation (Huang et al. 2017a; 2018; Wang et al.
subclinical PCV3 infection, a finding that is consistent 2020). The cGAS-STING pathway reacts for the viral
with that of seen in early stages of PCV2 infection DNA, while the RIG-I/MDA5/MAVS/IRF3/IRF7 signaling
(Temeeyasen et al. 2020). The downregulation of is activated by RNAs connected to viral gene expres-
IFN-β and upregulation of IL-6 and TNF-α expression sion processes (Huang et al. 2017a; 2018). In PAMs,
were observed in PCV3 inoculated HEK-293T cells PCV2 enhances the expression level of IKKα, IRF3 and
(Zhang et al. 2020c). Cytokine response to avian cir- IRF7 via MyD88 signaling that leads to IFN-α and
covirus infection is also poorly studied and results IFN-β production (Chen et al. 2016).
come mainly from vaccine experiments. Recombinant The beneficial effect of IFNs on PCV2 replication
PiCV Cap VLPs are promising vaccine candidates initiation could explain why PCV2 remains in a
inducing T-cell proliferation and increased IFN-γ level, latent state until IFN triggering stimuli (including
but decreasing TGF-β expression in lymphocytes non-circovirus agents) have an impact on the course
from spleen of PiCV uninfected pigeons, when com- of infection (Allan et al. 2000; Rovira et al. 2002).
pared to mock control (Huang et al. 2021). The PCV2 may promote IFN production under certain
recombinant Cap elevated the CD4, CD8 and IFN-γ conditions, but the underlying factors are not
expression levels in splenic mononuclear cells both understood.
in PiCV infected and uninfected pigeons, but the
humoral immunity was delayed in subclinical PiCV
cases (Stenzel et al. 2019). In DuCV infected Cherry 8. Molecular background of IFN interference
Valley ducklings, increased IL-10, decreased IL-12 and
IFN-γ levels, and decreased soluble CD4 and CD8 Although IFNs contribute to the induction of PCV2
amounts could be measured that may indicate an replication, PCV2 counteracts IFN-I producing and
impaired T-cell function. In the same time, DuCV signaling pathways from cGAS receptor signaling to
infection was shown to enhance the pathogenicity of STAT molecules, which supports the excessive repli-
avian pathogenic Escherichia coli (APEC) (Wang cation, and invasion of PCV2 and other pathogens
et al. 2022b). (Wu et al. 2021). Beside silencing of the catalytic
activity in early phase of infection, phosphorylation
of porcine cGAS at S278 via PI3K/AKT signaling (but
not via p38-MAPK, JNK, ERK) facilitates the K48-linked
7. Exploiting of IFNs
poly-ubiquitination and p62-mediated autophagic
As it has been exemplified, via its immunomodula- degradation of cGAS protein in PK-15 cells (Wang
tory sequence motifs or proteins, PCV2 generates et al. 2021b). The PCV2 Cap-gC1qR connection helps
insignificant levels of IFNs and decreases IFN expres- recruit p-PKCδ and promotes histone deacetylase 6
sion induced by other microbes (Hasslung et al. activation that mediates transport of the ubiquiti-
2003; Vincent et al. 2007; Wikstrom et al. 2007; nated cGAS from the cytosol to autolysosomes
Kekarainen et al. 2008a). Whilst inhibition of the IFN (Wang et al. 2021b).
response supports the immune evasion and disease The PCV2 dependent inhibition of IFN-α and
progression, IFNs up to a certain amount has been IFN-β production enhances PPV infection in piglets
found to support PCV2 uptake and replication initia- compared to those infected solely by PPV (Ouyang
tion in early stages of infection. et al. 2019; Wu et al. 2021). PCV2 hampers the pro-
An increased internalization of PCV2 VLPs is seen duction of IFN-α and IFN-β that is accompanied by
in the presence of externally added IFN-γ that prob- decreased ISG (such as ISG15; ISG56; IFIT2, IFN-
ably enhances endocytic activity of 3D4/31 cells induced protein with tetratricopeptide repeats;
(Meerts et al. 2005a). IFN-α and IFN-γ increase the CXCL10, C-X-C motif chemokine ligand 10) mRNA
number of infected cells and enhance PCV2 replica- levels in PBMCs of PCV2/PPV co-infected piglets (Wu
tion in PK-15 and 3D4/31 (porcine monocytic) cells, et al. 2021). PCV2 infection blocks the cGAMP
except that pretreatment with IFN-α inhibits viral induced IFN-β production in PPV co-infected PK-15
replication in pK-15 cells (Meerts et al. 2005a; culture by inhibiting K63 ubiquitination of STING
Ramamoorthy et al. 2009; Mutthi et al. 2018). An through activation of p38-MAPK pathway. p38-MAPK
ISRE-like sequence has been identified in the rep pro- signaling leads to phosphorylation and activation of
moter of PCV2 that promotes the propagation of the USP21 deubiquitinating enzyme that hydrolyzes
PCV2 in PK-15 and 3D4/31 cells in cooperation with polyubiquitin chains of STING. Deubiquitination of
the IFNs (Ramamoorthy et al. 2009). Concerted STING causes lower phosphorylation levels for IRF3
actions of the ISRE, TATA box and other (e.g. cis-act- and TBK1, weaker interaction of these molecules
ing) elements, together with variable interacting and decreased translocation to the nucleus (Wu
transcription factors, determine the response of PCV2 et al. 2021). The pIRF3 translocation can be inhibited
to these cytokines (Ramamoorthy et al. 2009). at other signaling points as well. Nuclear transport
PCV2-induced IFN production supports PCV2 rep- of IRF3 is mediated by KPNA3 and KPNA4 (Zhang
lication, too. PCV2 upregulates IFN-β production in et al. 2020b). PCV2 has been reported to be able to
PK-15 cells via RIG-I/MDA-5/MAVS/IRF3 signaling, as disrupt the interaction of KPNA3 with p-IRF3 in
well as through cGAS signaling with enhancement of order to block pIRF3 translocation to the nucleus for
10 E. FEHÉR ET AL.

the inhibition of IFN-β induction in PK-15 cells (Li G3BP1-cGAS interaction promotes DNA binding of
et al. 2018; Zhang et al. 2020b). cGAS and downstream signaling that lead to IFN-β
The Cap of PCV2, in concert with the cellular expression via TBK1 and IRF3 phosphorylation in
gC1qR protein, inhibits Type I IFN response as well. PK-15 and HEK-293T cells (Zhang et al. 2020c). The
Cap decreases the phosphorylation level of STAT1 interaction between PCV3 Cap and G3BP1 prevents
and STAT2 and their interaction with IRF9 upon IFN-α cGAS from recognizing the viral DNA that inhibits
treatment of PK-15 cells (Wang et al. 2022c). IFN-β production (Zhang et al. 2020c). In the same
Heterotrimer formation, translocation of ISGF3 to the time, IFN stimulated gene expression (ISG15, OAS1,
nucleus and binding to ISRE promoter of ISGs are MX1, MX2, IFIT3) have been shown to be upregu-
inhibited by both the Rep and Cap. Thus, PCV2 lated in lung samples of PCV3 infected piglets, but
reduces activation of the ISRE promoter and expres- the mechanisms behind are unexplored (Jiang
sion of ISGs (CXCL10; IFIT1; MX1, interferon-induced et al. 2020).
GTP-binding protein) (Wang et al. 2022c).
Downregulation of IFN-β production and JAK/STAT
signaling has been also reported in PCV2-PRV co-in- 9. Molecular background of IL-10/IL-12
fected PK-15 cells (Li et al. 2022). Along with Cap, pathway modulations
one has to highlight the role of ORF5 in IFN antago-
nism and support of PCV2 replication. As revealed by IL-10 is an immunoregulatory cytokine with anti-in-
mRNA analysis, the ORF5 protein of PCV2 antago- flammatory properties, produced primarily by mono-
nizes the transcription of genes involved in Type I cytic cells and B-cells. IL-10 hampers excessive
IFN production and IFN response in PK-15 cells, inflammation, allergy or autoimmune responses of
including RIG-I (DDX58), LGP2 (DHX58), MDA5 (IFIH1), the host. Some pathogens exploit its anti-inflamma-
IFIT1 and IFIT3, IFITM2 and IFITM3 (IFN induced tory effect to evade the immune response (Driessler
transmembrane proteins), IRF7 and IRF9, ISG15 and et al. 2004; Cao et al. 2006). The often seen downreg-
TLR3 (Choi et al. 2018). ulation of proinflammatory cytokines has been found

Figure 3. Effect of porcine circovirus 2 (PCV2) infection on IL-10 and IL-12 production in monocytic cells. PCV2 capsid protein
(Cap) interacts with gC1qR or other cellular proteins, activating PI3K/akt pathway typically in the early phase of infection, and
p38-MAPK and ERK in late phases. The activated transcription factors enhance IL-10 expression, while inhibition of NF-κB p65/
p50 and the upregulation of virus-triggered miRNAs result in IL-12 and IFN-γ transcription downregulation, and posttransla-
tional decrease of the IL-12 level.
Veterinary Quarterly 11

to associate with elevated IL-10 expression and with infected animals. Moreover, PCV2 promoted replica-
disease development in PCV2 infections (Figure 3) tion of the PRRSV and H. parasuis (Du et al. 2018).
(Darwich et al. 2003a; 2003b; Sipos et al. 2004; PCV2 Cap-gC1qR interaction has a key role in
Stevenson et al. 2006; Darwich et al. 2008; Shi et al. IL-12p40 decrease in PAMs with miR-23a, miR-23b
2010; Borghetti et al. 2013; Gao et al. 2014a; and miR-29b upregulation via PI3K/Akt1, and with
Richmond et al. 2015; Du et al. 2019). IL-10 supports miR-29a and miR-29b upregulation via the p38 MAPK
PCV2 replication in the late phase of infection and pathway (Du et al. 2018). miR-23a and miR-23b act
the viral load correlates with the IL-10 level (Darwich at posttranscriptional level, while miR-29a and miR-
et al. 2008; Du et al. 2019). The IL-10 mRNA level has 29b influence both transcription and the posttransla-
been shown to increase in the lymphoid tissues and tional level of IL-12p40. miR-23a and miR-29b were
has been related with the degree of lymphocyte shown to be the most important suppressors of
depletion in the thymus of pigs with PCVAD (Darwich IL-12p40 expression (Figure 3). The activation of Akt1
et al. 2003b). IL-10 causes CD4+ and CD8+ T-cell and p38 signaling leads to NF-κB p65 downregula-
depletion in the spleen, and inhibits the infiltration tion in macrophages and this prevents the binding
of these cells into the lung in mice (Du et al. 2019). of the transcription factor to the il12B (IL-12p40) pro-
The primary targets of PCV2 are monocytic cells in moter (Figure 3) (Du et al. 2018).
which the virus activates cellular pathways leading to In contrast to results related to downregulation of
IL-10 expression. IL-10 production in PAMs is induced IL-12 in monocytic cells, the decrease of IL-4 and
when viral Cap interacts with the ubiquitous cellular enhancement of IL-12 production has been mea-
protein gC1qR. Both in early and late phase of infec- sured in lymphocytes. It could be obtained via the
tion this complex triggers PI3K/Akt signaling that NF-κB pathway by TLR2, TLR3, TLR4, TLR9 and MyD88
cooperates with the NF-κB pathway, while p38-MAPK signal adaptor activation (Duan et al. 2014). It is in
signaling is highly active in later phases of infection accordance with studies revealing that the mono-
(Figure 2) (Du et al. 2016). Furthermore, the ERK cytic cell fraction has a primary role in reduction of
pathway can be upregulated by the Cap in parallel IL-12 in the PBMC populations (Kekarainen et al.
with p38-MAPK independent of gC1qR. At last, a 2008a; 2008b; Gao et al. 2014a).
number of transcription factors, including NF-κB p50,
AP1 and CREB (activated via phosphorylated PI3K/
Akt), as well as SP1 (activated via phosphorylated
10. Summary of the circovirus-associated
p38-MAPK and ERK) bind the IL-10 promoter and
immunosuppressive mechanisms
induce transcription of IL-10 (Figure 3) (Du et al.
2016). While Cap induces IL-10 expression early after The evaluation of published data on cellular and
PCV2 enters the cells, Rep promotes IL-10 activation immunological changes caused by animal circovirus
in later phase of infection in PAMs. Interaction of infection is challenging due to the complexity of the
PCV2 Rep with the cellular protein thymine DNA gly- molecular signaling and regulation of this network,
cosylase induces p38 phosphorylation, and the acti- and to the differences of the experimental condi-
vated p38-MAPK pathway, but not ERK and PI3K/Akt, tions. At present, the largest data set is available for
induces binding of NF-κB p50 and Sp1 to the IL-10 PCV2, a readily culturable circovirus with well-estab-
promoter (Figure 3) (Wu et al. 2019). The overex- lished animal model, thus presentation of the circo-
pressed IL-10, as one of the main targets, blocks IκBα virus-associated immunosuppression could be
degradation, thus inhibiting the release of NF-κB formulated for it.
p65/p50 and the nuclear translocation of p65. Monocytic cells, including macrophages/mono-
Furthermore, IL-10 aids the translocation and DNA cytes and DCs, have been identified as primary tar-
binding of the NF-κB1 p50/p50 complex in mono- gets of PCV2 and PiCV. These cells are not the main
cytic cells; p50 transactivates the IL-10 gene and location of viral replication but serve as a site for
mediates anti-inflammatory effect of IL-10 protein latent infection (Gilpin et al. 2003; Vincent et al. 2003;
(Figure 3) (Driessler et al. 2004; Cao et al. 2006). 2005; Chang et al. 2006; Darwich and Mateu 2012).
IL-12, a heterodimeric molecule composed of p35 In early phase of PCV2 infection most functions of
and p40 protein chains, is produced primarily by monocytic cells are intact, but in case of a ligand
antigen presenting cells, and plays a pivotal role in stimulus (e.g. concomitant infections) PCV2 impairs
cytotoxic T-lymphocyte and NK cell activation, and the cytokine response and raises an imbalance in the
CD4+ T lymphocyte maturation to IFN-γ producing activation of immune cells, hence interferes with the
Th1 cells (Ma et al. 2015). As mentioned above, innate and adaptive immunity (Vincent et al. 2005;
PCV2 induced IL-10 appears to be crucial in the 2007; Ablasser and Hur 2020). The alterations are
impairment of proinflammatory processes antago- achieved in a replication independent manner by
nizing the IL-12 and IFN-γ expression (Kekarainen structural components of PCV2, such as the genomic
et al. 2008a; 2008b; Gao et al. 2014a; Du et al. 2019). DNA and Cap protein (Vincent et al. 2007; Wikstrom
IL-12p40 and IFN-γ levels, as well as Th1 cell propor- et al. 2007; Kekarainen et al. 2008a; Balmelli et al.
tion were shown to decrease in PBMCs, and lung 2011). A key element of the Cap induced modifica-
and pulmonary lymph nodes in pigs challenged tions is the interaction of this protein with the cellu-
with PRRSV and Haemophilus parasuis following lar gC1qR molecule (Choi et al. 2015; Du et al. 2016;
PCV2 infection, compared to PCV1 and mock 2018; Wang et al. 2019; 2021b; 2022c). PCV2 not only
12 E. FEHÉR ET AL.

silences, but at some stages of infection triggers and number of newly identified species highlights the
exploits cytokines to initiate its own replication and diversity of circoviruses. As circoviruses have direct
protein expression, and to promote invasion of the pathological role and affect the outcome of concur-
target cells (Huang et al. 2017a; 2018; Wang et al. rent infections, it is crucial to gather more informa-
2020). At early stages of infections and subclinical tion about the genetics, evolution, pathomechanisms
cases upregulation of some cytokines (TNF-α, IL-1β, and immunomodulatory strategies of these viruses
IL-8) may help the survival and dissemination of and gain further insight into the interactions in the
monocytic cells that serve as portals of PCV2 to affected hosts. These data will be essential to identify
enter various tissues, while elevated IL-10 level results new targets relevant to disease prevention or treat-
in T-cell imbalances (Gilpin et al. 2003; Vincent et al. ment strategies.
2003; 2005; Chang et al. 2006; Darwich and Mateu
2012; Chen et al. 2016). Meanwhile, the ORF4 protein
and Cap support the maintenance of anti-apoptotic Disclosure statement
state of the cells (He et al. 2013; Liu et al. 2013; Gao The authors report there are no competing interests to
et al. 2014b). declare.
PCV2 itself does not typically cause severe disease
and in PCVAD the virus is often detected together
with co-infective microbes. As recorded, the swine Funding
pathogens may generate alone less serious disease
This work was supported by the (RRF-2.3.1-21-2022-
than in concert with PCV2 (Kim et al. 2006; Kekarainen
00010) of the University of Pécs, Hungary. Project no.
et al. 2008b; Shi et al. 2010; Sinha et al. 2011; Gao TKP2021-NVA-07 has been implemented with the support
et al. 2014a; Ouyang et al. 2019; Opriessnig et al. provided from the financed under the TKP2021-NVA fund-
2020). IFNs, induced by PCV2 or concomitant infec- ing scheme.
tive agents, may trigger expression of the viral Rep
through the ISRE motifs in the rep promoter, thus
advocate the virus replication in permissive cells ORCID
(Ramamoorthy et al. 2009; Sinha et al. 2011; Huang
Enikő Fehér http://orcid.org/0000-0001-7778-9116
et al. 2017a). At later phase of infection circoviruses Krisztián Bányai http://orcid.org/0000-0002-6270-1772
(as seen for PCV2 and PiCV) are detected in different
cell types and organs (Rosell et al. 1999; Darwich
et al. 2004). On the other hand, with progression of References
the infection, PCV2 reduces IFN production and pro-
Abadie J, Nguyen F, Groizeleau C, Amenna N, Fernandez B,
voke severe immunosuppression via more intense
Guereaud C, Guigand L, Robart P, Lefebvre B, Wyers M.
production of IL-10 that has detrimental effect on
2001. Pigeon circovirus infection: pathological observa-
the proinflammatory cytokine expression and on the tions and suggested pathogenesis. Avian Pathol.
activation of the adaptive immunity (Kekarainen 30(2):149–158. doi: 10.1080/03079450124811.
et al. 2008a; 2008b). Ablasser A, Hur S. 2020. Regulation of cGAS- and RLR-
The excessive replication of PCV2 requires the cel- mediated immunity to nucleic acids. Nat Immunol.
lular machinery, thus the viral life cycle is connected 21(1):17–29. doi: 10.1038/s41590-019-0556-1.
to the nucleus of mitotic cells. Modification of the Allan GM, McNeilly F, Ellis J, Krakowka S, Meehan B, McNair
cytokine expression, arrest of the cell cycle with pro- I, Walker I, Kennedy S. 2000. Experimental infection of
longation of the S-phase and progression of the colostrum deprived piglets with porcine circovirus 2
infection are supported by both structural and (PCV2) and porcine reproductive and respiratory syn-
drome virus (PRRSV) potentiates PCV2 replication. Arch
non-structural viral proteins. Furthermore, the repli-
Virol. 145(11):2421–2429. doi: 10.1007/s007050070031.
cative, intermediate dsDNA inhibits transport pro-
Allan GM, McNeilly F, Kennedy S, Daft B, Clarke EG, Ellis JA,
cesses, endocytosis, antigen presentation and Haines DM, Meehan BM, Adair BM. 1998. Isolation of
cytokine secretion (Liu et al. 2005; 2006; Balmelli porcine circovirus-like viruses from pigs with a wasting
et al. 2011; Tang et al. 2013; Lv et al. 2015; Guo et al. disease in the USA and Europe. J Vet Diagn Invest.
2018; Ablasser and Hur 2020; Lv et al. 2020). 10(1):3–10. doi: 10.1177/104063879801000102.
Lymphocyte depletion, typical for PCVAD, may be Amoroso MG, Serra F, Esposito C, D’Alessio N, Ferrara G,
the consequence of altered cytokine expression and Cioffi B, Anzalone A, Pagnini U, De Carlo E, Fusco G,
intense virus propagation that is enhanced by initia- et al. 2021. Prevalence of infection with porcine circovi-
tion of ER stress, autophagy and apoptotic processes rus types 2 and 3 in the wild boar population in the
supported by the Cap, Rep, ORF3 and ORF5 proteins campania region (Southern Italy). Animals (Basel).
11(11):3215. doi: 10.3390/ani11113215.
(Tang et al. 2013; Lv et al. 2015; Zhou et al. 2016;
Anderson A, Hartmann K, Leutenegger CM, Proksch AL,
Guo et al. 2018; Lv et al. 2020). At last, these changes
Mueller RS, Unterer S. 2017. Role of canine circovirus in
generate multiorgan failures and fatal events in dogs with acute haemorrhagic diarrhoea. Vet Rec.
the host. 180(22):542. doi: 10.1136/vr.103926.
PCV2 is fairly well controlled by vaccines. However, Balmelli C, Steiner E, Moulin H, Peduto N, Herrmann B,
emerging of novel PCV genotypes may lead to Summerfield A, McCullough K. 2011. Porcine circovirus
escape from immunity raised by vaccines currently in type 2 DNA influences cytoskeleton rearrangements in
use (Saikumar and Das 2019). Also, the increasing plasmacytoid and monocyte-derived dendritic cells.
Veterinary Quarterly 13

Immunology. 132(1):57–65. doi: temic wasting syndrome in response to mitogen, super-


10.1111/j.1365-2567.2010.03339.x. antigen or recall viral antigens. J Gen Virol. 84(Pt
Borghetti P, Morganti M, Saleri R, Ferrari L, De Angelis E, 12):3453–3457. doi: 10.1099/vir.0.19364-0.
Cavalli V, Cacchioli A, Corradi A, Martelli P. 2013. Innate Darwich L, Mateu E. 2012. Immunology of porcine circovi-
pro-inflammatory and adaptive immune cytokines in rus type 2 (PCV2). Virus Res. 164(1-2):61–67. doi:
PBMC of vaccinated and unvaccinated pigs naturally ex- 10.1016/j.virusres.2011.12.003.
posed to porcine circovirus type 2 (PCV2) infection vary Darwich L, Pie S, Rovira A, Segales J, Domingo M, Oswald
with the occurrence of the disease and the viral burden. IP, Mateu E. 2003b. Cytokine mRNA expression profiles in
Vet Microbiol. 163(1-2):42–53. doi: 10.1016/j.vet- lymphoid tissues of pigs naturally affected by postwean-
mic.2012.12.007. ing multisystemic wasting syndrome. J Gen Virol. 84(Pt
Cao J, Lin C, Wang H, Wang L, Zhou N, Jin Y, Liao M, Zhou 8):2117–2125. doi: 10.1099/vir.0.19124-0.
J. 2015. Circovirus transport proceeds via direct interac- Darwich L, Segales J, Mateu E. 2004. Pathogenesis of post-
tion of the cytoplasmic dynein IC1 subunit with the viral weaning multisystemic wasting syndrome caused by
capsid protein. J Virol. 89(5):2777–2791. doi: 10.1128/ Porcine circovirus 2: an immune riddle. Arch Virol.
JVI.03117-14. 149(5):857–874. doi: 10.1007/s00705-003-0280-9.
Cao L, Sun W, Lu H, Tian M, Xie C, Zhao G, Han J, Wang W, Darwich L, Segales J, Resendes A, Balasch M, Plana-Duran
Zheng M, Du R, et al. 2018. Genetic variation analysis of J, Mateu E. 2008. Transient correlation between viremia
PCV1 strains isolated from Guangxi Province of China in levels and IL-10 expression in pigs subclinically infected
2015. BMC Vet Res. 14(1):43. doi: 10.1186/s12917-018- with porcine circovirus type 2 (PCV2). Res Vet Sci.
1345-z. 84(2):194–198. doi: 10.1016/j.rvsc.2007.04.005.
Cao S, Zhang X, Edwards JP, Mosser DM. 2006. NF-kappaB1 Deim Z, Dencső L, Erdélyi I, Valappil SK, Varga C, Pósa A,
(p50) homodimers differentially regulate pro- and an- Makrai L, Rákhely G. 2019. Porcine circovirus type 3 de-
ti-inflammatory cytokines in macrophages. J Biol Chem. tection in a Hungarian pig farm experiencing reproduc-
281(36):26041–26050. doi: 10.1074/jbc.M602222200. tive failures. Vet Rec. 185(3):84. doi: 10.1136/vr.104784.
Cecere TE, Todd SM, Leroith T. 2012. Regulatory T cells in Deng Z, Sun R, Han X, Zhang Y, Zhou Y, Shan Y, Xu J, Li X,
arterivirus and coronavirus infections: do they protect He F, Fang W. 2022. Porcine circovirus 2 activates the
against disease or enhance it? Viruses. 4(5):833–846. doi: PERK-reactive oxygen species axis to induce p53 phos-
10.3390/v4050833. phorylation with subsequent cell cycle arrest at s phase
Chang HW, Jeng CR, Lin TL, Liu JJ, Chiou MT, Tsai YC, Chia in favor of its replication. J Virol. 96(22):e0127422. doi:
MY, Jan TR, Pang VF. 2006. Immunopathological effects 10.1128/jvi.01274-22.
of porcine circovirus type 2 (PCV2) on swine alveolar Dong VH, Tu PY, Tsai PC, Lin YH, Chang HL, Kuo TY, Chiou
macrophages by in vitro inoculation. Vet Immunol MT, Lin CN, Chung WB. 2015. Expression of Toll-like re-
Immunopathol. 110(3–4):207–219. doi: 10.1016/j.ve- ceptor signaling-related genes in pigs co-infected with
timm.2005.09.016. porcine reproductive and respiratory syndrome virus
Chen M, Han J, Zhang Y, Duan D, Zhang S. 2016. Porcine and porcine circovirus type 2. Res Vet Sci. 101:180–186.
circovirus type 2 induces type I interferon production doi: 10.1016/j.rvsc.2015.05.006.
via MyD88-IKKalpha-IRFs signaling rather than NF- Driessler F, Venstrom K, Sabat R, Asadullah K, Schottelius AJ.
kappaB in porcine alveolar macrophages in vitro. Res Vet 2004. Molecular mechanisms of interleukin-10-mediated in-
Sci. 104:188–194. doi: 10.1016/j.rvsc.2015.12.016. hibition of NF-kappaB activity: a role for p50. Clin Exp
Choi CY, Choi YC, Park IB, Lee CH, Kang SJ, Chun T. 2018. Immunol. 135(1):64–73. doi: 10.1111/j.1365-2249.2004.02342.x.
The ORF5 protein of porcine circovirus type 2 enhances Duan D, Zhang S, Li X, Guo H, Chen M, Zhang Y, Han J, Lv
viral replication by dampening type I interferon expres- Y. 2014. Activation of the TLR/MyD88/NF-kappaB signal
sion in porcine epithelial cells. Vet Microbiol. 226:50–58. pathway contributes to changes in IL-4 and IL-12 pro-
doi: 10.1016/j.vetmic.2018.10.005. duction in piglet lymphocytes infected with porcine cir-
Choi CY, Oh HN, Jun Lee S, Chun T. 2015. ORF2 protein of covirus type 2 in vitro. PLoS One. 9(5):e97653. doi:
porcine circovirus type 2 promotes phagocytic activity 10.1371/journal.pone.0097653.
of porcine macrophages by inhibiting proteasomal deg- Du Q, Huang Y, Wang T, Zhang X, Chen Y, Cui B, Li D, Zhao
radation of complement component 1, q subcomponent X, Zhang W, Chang L, et al. 2016. Porcine circovirus type
binding protein (C1QBP) through physical interaction. J 2 activates PI3K/Akt and p38 MAPK pathways to pro-
Gen Virol. 96(11):3294–3301. doi: 10.1099/jgv.0.000282. mote interleukin-10 production in macrophages via Cap
Circella E, Legretto M, Pugliese N, Caroli A, Bozzo G, Accogli interaction of gC1qR. Oncotarget. 7(14):17492–17507.
G, Lavazza A, Camarda A. 2014. Psittacine beak and feather doi: 10.18632/oncotarget.7362.
disease-like illness in Gouldian finches (Chloebia gouldiae). Du Q, Wu X, Wang T, Yang X, Wang Z, Niu Y, Zhao X, Liu
Avian Dis. 58(3):482–487. doi: 10.1637/10745-121113Case.1. SL, Tong D, Huang Y. 2018. Porcine circovirus Type 2 sup-
Cui Y, Hou L, Pan Y, Feng X, Zhou J, Wang D, Guo J, Liu C, presses IL-12p40 induction via Capsid/gC1qR-mediated
Shi Y, Sun T, et al. 2022. Reconstruction of the evolution- MicroRNAs and signalings. J Immunol. 201(2):533–547.
ary origin, phylodynamics, and phylogeography of the doi: 10.4049/jimmunol.1800250.
porcine circovirus type 3. Front Microbiol. 13:898212. Du Q, Zhang H, He M, Zhao X, He J, Cui B, Yang X, Tong D,
doi: 10.3389/fmicb.2022.898212. Huang Y. 2019. Interleukin-10 promotes porcine circovi-
Dankaona W, Mongkholdej E, Satthathum C, Piewbang C, rus type 2 persistent infection in mice and aggravates
Techangamsuwan S. 2022. Epidemiology, genetic diversi- the tissue lesions by suppression of T cell infiltration.
ty, and association of canine circovirus infection in dogs Front Microbiol. 10:2050. doi: 10.3389/fmicb.2019.02050.
with respiratory disease. Sci Rep. 12(1):15445. doi: Eaglesham JB, Kranzusch PJ. 2020. Conserved strategies for
10.1038/s41598-022-19815-z. pathogen evasion of cGAS-STING immunity. Curr Opin
Darwich L, Balasch M, Plana-Duran J, Segales J, Domingo Immunol. 66:27–34. doi: 10.1016/j.coi.2020.04.002.
M, Mateu E. 2003a. Cytokine profiles of peripheral blood Ellis J, Hassard L, Clark E, Harding J, Allan G, Willson P,
mononuclear cells from pigs with postweaning multisys- Strokappe J, Martin K, McNeilly F, Meehan B, et al. 1998.
14 E. FEHÉR ET AL.

Isolation of circovirus from lesions of pigs with post- sis of porcine circovirus 2 in wild boar from Jiangxi
weaning multisystemic wasting syndrome. Can Vet J. Province of China. Animals (Basel. 12(16):2021. doi:
39(1):44–51. 10.3390/ani12162021.
Fanelli A, Pellegrini F, Camero M, Catella C, Buonavoglia D, Huang YL, Castaneda OA, Thongchan D, Khatri-Chhetri R,
Fusco G, Martella V, Lanave G. 2022. Genetic diversity of Tsai SS, Wu HY. 2017b. Pigeon circovirus infection in dis-
porcine circovirus types 2 and 3 in wild boar in Italy. qualified racing pigeons from Taiwan. Avian Pathol.
Animals (Basel. 12(8):953. doi: 10.3390/ani12080953. 46(4):359–366. doi: 10.1080/03079457.2017.1284305.
Feher E, Kaszab E, Bali K, Hoitsy M, Sos E, Banyai K. 2022. Huang B, Li J, Zhang X, Zhao Q, Lu M, Lv Y. 2017a. RIG-1
Novel circoviruses from birds share common evolution- and MDA-5 signaling pathways contribute to IFN-beta
ary roots with fish origin circoviruses. Life (Basel. production and viral replication in porcine circovirus vi-
12(3):368. doi: 10.3390/life12030368. rus type 2-infected PK-15 cells in vitro. Vet Microbiol.
Gao Z, Dong Q, Jiang Y, Opriessnig T, Wang J, Quan Y, Yang 211:36–42. doi: 10.1016/j.vetmic.2017.09.022.
Z. 2014b. ORF4-protein deficient PCV2 mutants enhance Huang HY, Silva BBI, Tsai SP, Tsai CY, Tyan YC, Lin TC, Flores
virus-induced apoptosis and show differential expression RJD, Chuang KP. 2021. Immunogenicity and protective
of mRNAs in vitro. Virus Res. 183:56–62. doi: 10.1016/j. activity of pigeon circovirus recombinant capsid protein
virusres.2014.01.024. virus-like particles (PiCV rCap-VLPs) in pigeons (Columba
Gao F, Xie JL, Jia CW, Ren HY, Zhou SH. 2014a. Effects of livia) experimentally infected with PiCV. Vaccines (Basel).
porcine circovirus type 2 and pseudorabies vaccine 9(2):98. doi: 10.3390/vaccines9020098.
co-inoculation on regulatory cytokine mRNA expression Huang B, Zhang L, Lu M, Li J, Lv Y. 2018. PCV2 infection
in pig peripheral blood mononuclear cells. Genet Mol activates the cGAS/STING signaling pathway to promote
Res. 13(1):1540–1547. doi: 10.4238/2014.March.12.6. IFN-beta production and viral replication in PK-15 cells.
Garcia-Sastre A. 2017. Ten strategies of interferon evasion Vet Microbiol. 227:34–40. doi: 10.1016/j.vet-
by viruses. Cell Host Microbe. 22(2):176–184. doi: mic.2018.10.027.
10.1016/j.chom.2017.07.012. Igriczi B, Denes L, Biksi I, Albert E, Revesz T, Balka G. 2022.
Geng SC, Li XL, Fang WH. 2020. Porcine circovirus 3 capsid High prevalence of porcine circovirus 3 in hungarian pig
protein induces autophagy in HEK293T cells by inhibit- herds: results of a systematic sampling protocol. Viruses.
ing phosphorylation of the mammalian target of rapa- 14(6):1219. doi: 10.3390/v14061219.
mycin. J Zhejiang Univ Sci B. 21(7):560–570. doi: 10.1631/ Iizuka N, Kohara M, Hagino-Yamagishi K, Abe S, Komatsu T,
jzus.B1900657. Tago K, Arita M, Nomoto A. 1989. Construction of less
Gilpin DF, McCullough K, Meehan BM, McNeilly F, McNair I, neurovirulent polioviruses by introducing deletions into
Stevenson LS, Foster JC, Ellis JA, Krakowka S, Adair BM, the 5’ noncoding sequence of the genome. J Virol.
et al. 2003. In vitro studies on the infection and replica- 63(12):5354–5363. doi: 10.1128/JVI.63.12.5354-5363.1989.
tion of porcine circovirus type 2 in cells of the porcine Jiang H, Wei L, Wang D, Wang J, Zhu S, She R, Liu T, Tian
immune system. Vet Immunol Immunopathol. 94(3– J, Quan R, Hou L, et al. 2020. ITRAQ-based quantitative
4):149–161. doi: 10.1016/s0165-2427(03)00087-4. proteomics reveals the first proteome profiles of piglets
Guo J, Tian J, Tan X, Yu H, Ding S, Sun H, Yu X. 2011. infected with porcine circovirus type 3. J Proteomics.
Pathological observations of an experimental infection 212:103598. doi: 10.1016/j.jprot.2019.103598.
of geese with goose circovirus. Avian Pathol. 40(1):55– Kaszab E, Lengyel G, Marton S, Dan A, Banyai K, Feher E.
61. doi: 10.1080/03079457.2010.538371. 2020. Occurrence and genetic diversity of CRESS DNA
Guo K, Xu L, Wu M, Hou Y, Jiang Y, Lv J, Xu P, Fan Z, Zhang viruses in wild birds: a Hungarian study. Sci Rep.
R, Xing F, et al. 2018. A host factor GPNMB restricts por- 10(1):7036. doi: 10.1038/s41598-020-63795-x.
cine circovirus type 2 (PCV2) replication and interacts Kekarainen T, Montoya M, Dominguez J, Mateu E, Segales
with PCV2 ORF5 protein. Front Microbiol. 9:3295. doi: J. 2008a. Porcine circovirus type 2 (PCV2) viral compo-
10.3389/fmicb.2018.03295. nents immunomodulate recall antigen responses. Vet
Hansoongnern P, Phecharat N, Wasanasuk K, Tommeurd W, Immunol Immunopathol. 124(1-2):41–49. doi: 10.1016/j.
Chankeeree P, Lekcharoensuk C, Semkum P, Pinitkiatisakul vetimm.2008.01.031.
S, Lekcharoensuk P. 2022. Encapsidated-CpG ODN en- Kekarainen T, Montoya M, Mateu E, Segales J. 2008b.
hances immunogenicity of porcine circovirus type 2 vi- Porcine circovirus type 2-induced interleukin-10 modu-
rus-like particles. Vet Microbiol. 275:109583. doi: lates recall antigen responses. J Gen Virol. 89(Pt 3):760–
10.1016/j.vetmic.2022.109583. 765. doi: 10.1099/vir.0.83354-0.
Hasslung FC, Berg M, Allan GM, Meehan BM, McNeilly F, Kim J, Ha Y, Chae C. 2006. Potentiation of porcine circovirus
Fossum C. 2003. Identification of a sequence from the 2-induced postweaning multisystemic wasting syndrome
genome of porcine circovirus type 2 with an inhibitory by porcine parvovirus is associated with excessive pro-
effect on IFN-alpha production by porcine PBMCs. J Gen duction of tumor necrosis factor-alpha. Vet Pathol.
Virol. 84(Pt 11):2937–2945. doi: 10.1099/vir.0.19362-0. 43(5):718–725. doi: 10.1354/vp.43-5-718.
Hayden MS, Ghosh S. 2014. Regulation of NF-kappaB by Kim SH, Park JY, Jung JY, Kim HY, Park YR, Lee KK, Lyoo YS,
TNF family cytokines. Semin Immunol. 26(3):253–266. Yeo SG, Park CK. 2018. Detection and genetic character-
doi: 10.1016/j.smim.2014.05.004. ization of porcine circovirus 3 from aborted fetuses and
He J, Cao J, Zhou N, Jin Y, Wu J, Zhou J. 2013. Identification pigs with respiratory disease in Korea. J Vet Sci.
and functional analysis of the novel ORF4 protein en- 19(5):721–724. doi: 10.4142/jvs.2018.19.5.721.
coded by porcine circovirus type 2. J Virol. 87(3):1420– Kozdruń W, Woźniakowski G, Samorek-Salamonowicz E, Czekaj
1429. doi: 10.1128/JVI.01443-12. H. 2012. Viral infections in goose flocks in Poland. Pol J Vet
Hong YT, Kang M, Jang HK. 2018. Pathogenesis of duck cir- Sci. 15(3):525–530. doi: 10.2478/v10181-012-0080-9.
covirus genotype 1 in experimentally infected Pekin Kroeger M, Temeeyasen G, Pineyro PE. 2022. Five years of
ducks. Poult Sci. 97(9):3050–3057. doi: 10.3382/ps/pey177. porcine circovirus 3: what have we learned about the
Hu X, Chen Z, Li Y, Ding Z, Zeng Q, Wan T, Wu H. 2022. clinical disease, immune pathogenesis, and diagnosis.
Detection of porcine circovirus 1/2/3 and genetic analy- Virus Res. 314:198764. doi: 10.1016/j.virusres.2022.198764.
Veterinary Quarterly 15

Lee AJ, Ashkar AA. 2018. The dual nature of type I and Meerts P, Misinzo G, Nauwynck HJ. 2005a. Enhancement of
type II interferons. Front Immunol. 9:2061. doi: 10.3389/ porcine circovirus 2 replication in porcine cell lines by
fimmu.2018.02061. IFN-gamma before and after treatment and by IFN-alpha
Li X, Chen S, Zhang L, Niu G, Zhang X, Yang L, Ji W, Ren L. after treatment. J Interferon Cytokine Res. 25(11):684–
2022. Coinfection of porcine circovirus 2 and pseudora- 693. doi: 10.1089/jir.2005.25.684.
bies virus enhances immunosuppression and inflamma- Meerts P, Van Gucht S, Cox E, Vandebosch A, Nauwynck HJ.
tion through NF-kappaB, JAK/STAT, MAPK, and NLRP3 2005b. Correlation between type of adaptive immune
pathways. IJMS. 23(8):4469. doi: 10.3390/ijms23084469. response against porcine circovirus type 2 and level of
Li J, Lu M, Huang B, Lv Y. 2018. Porcine circovirus type 2 virus replication. Viral Immunol. 18(2):333–341. doi:
inhibits inter-beta expression by targeting Karyopherin 10.1089/vim.2005.18.333.
alpha-3 in PK-15 cells. Virology. 520:75–82. doi: 10.1016/j. Misinzo G, Delputte PL, Lefebvre DJ, Nauwynck HJ. 2009.
virol.2018.05.008. Porcine circovirus 2 infection of epithelial cells is clath-
Liu J, Bai J, Zhang L, Jiang Z, Wang X, Li Y, Jiang P. 2013. rin-, caveolae- and dynamin-independent, actin and
Hsp70 positively regulates porcine circovirus type 2 rep- Rho-GTPase-mediated, and enhanced by cholesterol de-
lication in vitro. Virology. 447(1–2):52–62. doi: 10.1016/j. pletion. Virus Res. 139(1):1–9. doi: 10.1016/j.virus-
virol.2013.08.025. res.2008.09.005.
Liu J, Chen I, Du Q, Chua H, Kwang J. 2006. The ORF3 pro- Misinzo G, Delputte PL, Meerts P, Lefebvre DJ, Nauwynck
tein of porcine circovirus type 2 is involved in viral HJ. 2006. Porcine circovirus 2 uses heparan sulfate and
pathogenesis in vivo. J Virol. 80(10):5065–5073. doi: chondroitin sulfate B glycosaminoglycans as receptors
10.1128/JVI.80.10.5065-5073.2006. for its attachment to host cells. J Virol. 80(7):3487–3494.
Liu J, Chen I, Kwang J. 2005. Characterization of a previous- doi: 10.1128/JVI.80.7.3487-3494.2006.
ly unidentified viral protein in porcine circovirus type Misinzo G, Meerts P, Bublot M, Mast J, Weingartl HM,
2-infected cells and its role in virus-induced apoptosis. J Nauwynck HJ. 2005. Binding and entry characteristics of
Virol. 79(13):8262–8274. doi: 10.1128/JVI.79.13.8262- porcine circovirus 2 in cells of the porcine monocytic
8274.2005. line 3D4/31. J Gen Virol. 86(Pt 7):2057–2068. doi:
Liu X, Shen H, Zhang X, Liang T, Ban Y, Yu L, Zhang L, Liu 10.1099/vir.0.80652-0.
Y, Dong J, Zhang P, et al. 2021. Porcine circovirus type 3 Mutthi P, Theerawatanasirikul S, Roytrakul S, Paemanee A,
capsid protein induces NF-kappaB activation and upreg- Lekcharoensuk C, Hansoongnern P, Petcharat N,
ulates pro-inflammatory cytokine expression in HEK- Thangthamniyom N, Lekcharoensuk P. 2018. Interferon
293T cells. Arch Virol. 166(8):2141–2149. doi: 10.1007/ gamma induces cellular protein alteration and increases
s00705-021-05104-z. replication of porcine circovirus type 2 in PK-15 cells.
Liu X, Wang Y, Han C, Li Q, Hou X, Song Q, Zhou S, Li H. Arch Virol. 163(11):2947–2957. doi: 10.1007/s00705-018-
2022. TGF-beta from the porcine intestinal cell line 3944-1.
IPEC-J2 induced by porcine circovirus 2 increases the fre- Nayar GP, Hamel A, Lin L. 1997. Detection and characteri-
quency of treg cells via the activation of ERK (in CD4(+) zation of porcine circovirus associated with postweaning
T Cells) and NF-kappaB (in IPEC-J2). Viruses. 14(11):2466. multisystemic wasting syndrome in pigs. Can Vet J.
doi: 10.3390/v14112466. 38(6):385–386.
Li XD, Wu J, Gao D, Wang H, Sun L, Chen ZJ. 2013. Pivotal Niu G, Zhang X, Ji W, Chen S, Li X, Yang L, Zhang L, Ouyang H,
roles of cGAS-cGAMP signaling in antiviral defense and Li C, Ren L. 2022. Porcine circovirus 4 rescued from an infec-
immune adjuvant effects. Science. 341(6152):1390–1394. tious clone is replicable and pathogenic in vivo. Transbound
doi: 10.1126/science.1244040. Emerg Dis. 69(5):e1632–e1641. doi: 10.1111/tbed.14498.
Luka PD, Adedeji AJ, Jambol AR, Ifende IV, Luka HG, Choji Opriessnig T, Karuppannan AK, Castro A, Xiao CT. 2020.
ND, Weka R, Settypalli TBK, Achenbach JE, Cattoli G, Porcine circoviruses: current status, knowledge gaps and
et al. 2022. Coinfections of African swine fever virus, challenges. Virus Res. 286:198044. doi: 10.1016/j.virus-
porcine circovirus 2 and 3, and porcine parvovirus 1 in res.2020.198044.
swine in Nigeria. Arch Virol. 167(12):2715–2722. doi: Ouyang T, Zhang X, Liu X, Ren L. 2019. Co-infection of
10.1007/s00705-022-05593-6. swine with porcine circovirus type 2 and other swine
Lv Q, Guo K, Xu H, Wang T, Zhang Y. 2015. Identification of viruses. Viruses. 11(2):185. doi: 10.3390/v11020185.
putative ORF5 protein of porcine circovirus type 2 and Palinski R, Pineyro P, Shang P, Yuan F, Guo R, Fang Y, Byers
functional analysis of GFP-Fused ORF5 protein. PLoS E, Hause BM. 2017. A novel porcine circovirus distantly
One. 10(6):e0127859. doi: 10.1371/journal.pone.0127859. related to known circoviruses is associated with porcine
Lv QZ, Guo KK, Zhang YM. 2014. Current understanding of dermatitis and nephropathy syndrome and reproductive
genomic DNA of porcine circovirus type 2. Virus Genes. failure. J Virol. 91(1):e01879–16. doi: 10.1128/JVI.01879-16.
49(1):1–10. doi: 10.1007/s11262-014-1099-z. Pan Y, Li P, Jia R, Wang M, Yin Z, Cheng A. 2018. Regulation
Lv J, Jiang Y, Feng Q, Fan Z, Sun Y, Xu P, Hou Y, Zhang X, of apoptosis during porcine circovirus type 2 infection.
Fan Y, Xu X, et al. 2020. Porcine circovirus type 2 ORF5 Front Microbiol. 9:2086. doi: 10.3389/fmicb.2018.02086.
protein induces autophagy to promote viral replication Phan TG, Giannitti F, Rossow S, Marthaler D, Knutson TP, Li
via the PERK-eIF2alpha-ATF4 and mTOR-ERK1/2-AMPK L, Deng X, Resende T, Vannucci F, Delwart E. 2016.
signaling pathways in PK-15 cells. Front Microbiol. Detection of a novel circovirus PCV3 in pigs with cardiac
11:320. doi: 10.3389/fmicb.2020.00320. and multi-systemic inflammation. Virol J. 13(1):184. doi:
Ma X, Yan W, Zheng H, Du Q, Zhang L, Ban Y, Li N, Wei F. 10.1186/s12985-016-0642-z.
2015. Regulation of IL-10 and IL-12 production and func- Ramamoorthy S, Huang FF, Huang YW, Meng XJ. 2009.
tion in macrophages and dendritic cells. F1000Res. Interferon-mediated enhancement of in vitro replication
4:1465. doi: 10.12688/f1000research.7010.1. of porcine circovirus type 2 is influenced by an interfer-
Maizels RM, Smith KA. 2011. Regulatory T cells in infection. on-stimulated response element in the PCV2 genome.
Adv Immunol. 112:73–136. doi: 10.1016/B978-0-12- Virus Res. 145(2):236–243. doi: 10.1016/j.virusres.
387827-4.00003-6. 2009.07.009.
16 E. FEHÉR ET AL.

Raue R, Schmidt V, Freick M, Reinhardt B, Johne R, tions. J Leukoc Biol. 75(2):163–189. doi: 10.1189/
Kamphausen L, Kaleta EF, Muller H, Krautwald-Junghanns jlb.0603252.
ME. 2005. A disease complex associated with pigeon cir- Shen H, Liu X, Zhang P, Wang S, Liu Y, Zhang L, Song C.
covirus infection, young pigeon disease syndrome. Avian 2020. Porcine circovirus 3 Cap inhibits type I interferon
Pathol. 34(5):418–425. doi: 10.1080/03079450500267825. signaling through interaction with STAT2. Virus Res.
Richmond O, Cecere TE, Erdogan E, Meng XJ, Pineyro P, 275:197804. doi: 10.1016/j.virusres.2019.197804.
Subramaniam S, Todd SM, LeRoith T. 2015. The PD-L1/ Shi KC, Guo X, Ge XN, Liu Q, Yang HC. 2010. Cytokine
CD86 ratio is increased in dendritic cells co-infected mRNA expression profiles in peripheral blood mononu-
with porcine circovirus type 2 and porcine reproductive clear cells from piglets experimentally co-infected with
and respiratory syndrome virus, and the PD-L1/PD-1 axis porcine reproductive and respiratory syndrome virus
is associated with anergy, apoptosis, and the induction and porcine circovirus type 2. Vet Microbiol. 140(1–
of regulatory T-cells in porcine lymphocytes. Vet 2):155–160. doi: 10.1016/j.vetmic.2009.07.021.
Microbiol. 180(3–4):223–229. doi: 10.1016/j.vet- Shivaprasad HL, Hill D, Todd D, Smyth JA. 2004. Circovirus
mic.2015.09.014. infection in a Gouldian finch (Chloebia gouldiae). Avian
Ritchie BW, Niagro FD, Lukert PD, Steffens WL, 3rd, Latimer Pathol. 33(5):525–529. doi: 10.1080/03079450400003585.
KS. 1989. Characterization of a new virus from cockatoos Sinha A, Shen HG, Schalk S, Beach NM, Huang YW, Meng
with psittacine beak and feather disease. Virology. XJ, Halbur PG, Opriessnig T. 2011. Porcine reproductive
171(1):83–88. doi: 10.1016/0042-6822(89)90513-8. and respiratory syndrome virus (PRRSV) influences infec-
Robino P, Grego E, Rossi G, Bert E, Tramuta C, Stella MC, tion dynamics of porcine circovirus type 2 (PCV2) sub-
Bertoni P, Nebbia P. 2014. Molecular analysis and associ- types PCV2a and PCV2b by prolonging PCV2 viremia
ated pathology of beak and feather disease virus isolat- and shedding. Vet Microbiol. 152(3–4):235–246. doi:
ed in Italy from young Congo African grey parrots 10.1016/j.vetmic.2011.05.005.
(Psittacus erithacus) with an "atypical peracute form" of Sipos W, Duvigneau JC, Willheim M, Schilcher F, Hartl RT,
the disease. Avian Pathol. 43(4):333–344. doi: Hofbauer G, Exel B, Pietschmann P, Schmoll F. 2004.
10.1080/03079457.2014.934660. Systemic cytokine profile in feeder pigs suffering from
Rojas JM, Alejo A, Martin V, Sevilla N. 2021. Viral patho- natural postweaning multisystemic wasting syndrome
gen-induced mechanisms to antagonize mammalian in- (PMWS) as determined by semiquantitative RT-PCR and
terferon (IFN) signaling pathway. Cell Mol Life Sci. flow cytometric intracellular cytokine detection. Vet
78(4):1423–1444. doi: 10.1007/s00018-020-03671-z. Immunol Immunopathol. 99(1–2):63–71. doi: 10.1016/j.
Rosario K, Breitbart M, Harrach B, Segales J, Delwart E, vetimm.2004.01.001.
Biagini P, Varsani A. 2017. Revisiting the taxonomy of the Sirisereewan C, Thanawongnuwech R, Kedkovid R. 2022.
family Circoviridae: establishment of the genus Cyclovirus Current understanding of the pathogenesis of porcine
and removal of the genus Gyrovirus. Arch Virol. circovirus 3. Pathogens. 11(1):64. doi: 10.3390/patho-
162(5):1447–1463. doi: 10.1007/s00705-017-3247-y. gens11010064.
Rosell C, Segales J, Plana-Duran J, Balasch M, Rodriguez- Soike D, Kohler B, Albrecht K. 1999. A circovirus-like infec-
Arrioja GM, Kennedy S, Allan GM, McNeilly F, Latimer KS, tion in geese related to a runting syndrome. Avian
Domingo M. 1999. Pathological, immunohistochemical, Pathol. 28(2):199–202. doi: 10.1080/03079459994939.
and in-situ hybridization studies of natural cases of post- Song J, Hou L, Wang D, Wei L, Zhu S, Wang J, Quan R,
weaning multisystemic wasting syndrome (PMWS) in Jiang H, Shi R, Liu J. 2021. Nucleolar phosphoprotein
pigs. J Comp Pathol. 120(1):59–78. doi: 10.1053/ NPM1 interacts with porcine circovirus type 3 cap pro-
jcpa.1998.0258. tein and facilitates viral replication. Front Microbiol.
Rovira A, Balasch M, Segales J, Garcia L, Plana-Duran J, Rosell 12:679341. doi: 10.3389/fmicb.2021.679341.
C, Ellerbrok H, Mankertz A, Domingo M. 2002. Experimental Stanifer ML, Guo C, Doldan P, Boulant S. 2020. Importance
inoculation of conventional pigs with porcine reproductive of type I and III interferons at respiratory and intestinal
and respiratory syndrome virus and porcine circovirus 2. J barrier surfaces. Front Immunol. 11:608645. doi: 10.3389/
Virol. 76(7):3232–3239. doi: 10.1128/jvi.76.7.3232-3239.2002. fimmu.2020.608645.
Sahindokuyucu I, Turkmen MB, Sumer T, Elhag AE, Alcigir Steiner E, Balmelli C, Herrmann B, Summerfield A,
ME, Yazici Z, Barry G, Gulbahar MY, Kul O. 2022. First de- McCullough K. 2008. Porcine circovirus type 2 displays
tection and molecular characterisation of a pigeon pluripotency in cell targeting. Virology. 378(2):311–322.
aviadenovirus A and pigeon circovirus co-infection asso- doi: 10.1016/j.virol.2008.06.009.
ciated with Young Pigeon Disease Syndrome (YPDS) in Stenzel T, Dziewulska D, Śmiałek M, Tykałowski B, Kowalczyk
Turkish pigeons (Columba livia domestica). Vet Med Sci. J, Koncicki A. 2019. Comparison of the immune response
8(1):139–149. doi: 10.1002/vms3.662. to vaccination with pigeon circovirus recombinant cap-
Saikumar G, Das T. 2019. Porcine circovirus. In: Malik Y, sid protein (PiCV rCP) in pigeons uninfected and subclin-
Singh R, Yadav M, editors. Recent advances in animal vi- ically infected with PiCV. PLoS One. 14(6):e0219175. doi:
rology. Singapore: Springer; p. 171–195. doi: 10.1007/978- 10.1371/journal.pone.0219175.
981-13-9073-9_10. Stenzel T, Dziewulska D, Tykałowski B, Koncicki A. 2020. The
Schmidt V, Schlomer J, Luken C, Johne R, Biere B, Muller H, clinical infection with pigeon circovirus (PiCV) leads to
Krautwald-Junghanns ME. 2008. Experimental infection lymphocyte B apoptosis but has no effect on lympho-
of domestic pigeons with pigeon circovirus. Avian Dis. cyte T subpopulation. Pathogens. 9(8):632. doi: 10.3390/
52(3):380–386. doi: 10.1637/8188-120407-Reg. pathogens9080632.
Schneider WM, Chevillotte MD, Rice CM. 2014. Interferon- Stenzel T, Koncicki A. 2017. The epidemiology, molecular
stimulated genes: a complex web of host defenses. characterization and clinical pathology of circovirus in-
Annu Rev Immunol. 32:513–545. doi: 10.1146/an- fections in pigeons - current knowledge. Vet Q.
nurev-immunol-032713-120231. 37(1):166–174. doi: 10.1080/01652176.2017.1325972.
Schroder K, Hertzog PJ, Ravasi T, Hume DA. 2004. Interferon- Stevenson LS, McCullough K, Vincent I, Gilpin DF,
gamma: an overview of signals, mechanisms and func- Summerfield A, Nielsen J, McNeilly F, Adair BM, Allan
Veterinary Quarterly 17

GM. 2006. Cytokine and C-reactive protein profiles in- Wang Z, Chen J, Wu X, Ma D, Zhang X, Li R, Han C, Liu H,
duced by porcine circovirus type 2 experimental infec- Yin X, Du Q, et al. 2021b. PCV2 targets cGAS to inhibit
tion in 3-week-old piglets. Viral Immunol. 19(2):189–195. type I interferon induction to promote other DNA virus
doi: 10.1089/vim.2006.19.189. infection. PLoS Pathog. 17(9):e1009940. doi: 10.1371/
Sun R, Deng Z, Han X, Zhang Y, Zhou Y, Shan Y, Fang W, Li journal.ppat.1009940.
X. 2021. Porcine circovirus 2 manipulates the PERK- Wang Z, Chen J, Zhang QG, Huang K, Ma D, Du Q, Tong D,
ERO1alpha axis of the endoplasmic reticulum to favor its Huang Y. 2022c. Porcine circovirus type 2 infection inhib-
replication by derepressing viral DNA from HMGB1 se- its the activation of type I interferon signaling via capsid
questration within nuclei. J Virol. 95(19):e0100921. doi: protein and host gC1qR. Vet Microbiol. 266:109354. doi:
10.1128/JVI.01009-21. 10.1016/j.vetmic.2022.109354.
Sydler T, Bragger S, Handke M, Hartnack S, Lewis FI, Sidler Wang T, Du Q, Niu Y, Zhang X, Wang Z, Wu X, Yang X, Zhao
X, Brugnera E. 2016. Latent porcine circovirus type 2-in- X, Liu SL, Tong D, et al. 2019. Cellular p32 is a critical reg-
fected domestic pigs: a potential infection model for the ulator of porcine circovirus type 2 nuclear egress. J Virol.
effective development of vaccines against latent or 93(23): doi: 10.1128/JVI.00979-19.
chronic virus induced diseases. Vaccine. 34(8):1047–1053. Wang T, Du Q, Wu X, Niu Y, Guan L, Wang Z, Zhao X, Liu
doi: 10.1016/j.vaccine.2016.01.005. SL, Tong D, Huang Y. 2018. Porcine MKRN1 modulates
Tang Q, Li S, Zhang H, Wei Y, Wu H, Liu J, Wang Y, Liu D, the replication and pathogenesis of porcine circovirus
Zhang Z, Liu C. 2013. Correlation of the cyclin A expres- type 2 by inducing capsid protein ubiquitination and
sion level with porcine circovirus type 2 propagation degradation. J Virol. 92(21):e00100–00118. doi: 10.1128/
efficiency. Arch Virol. 158(12):2553–2560. doi: 10.1007/ JVI.00100-18.
s00705-013-1785-5. Wang X, Li L, Shang H, Zhou F, Wang C, Zhang S, Gao P,
Temeeyasen G, Lierman S, Arruda BL, Main R, Vannucci F, Guo P, Zhu R, Sun Z, et al. 2022b. Effects of duck circo-
Gimenez-Lirola LG, Pineyro PE. 2020. Pathogenicity and virus on immune function and secondary infection of
immune response against porcine circovirus type 3 in- Avian Pathogenic Escherichia coli. Poult Sci.
fection in caesarean-derived, colostrum-deprived pigs. J 101(5):101799. doi: 10.1016/j.psj.2022.101799.
Gen Virol. 102(11):001502. doi: 10.1099/jgv.0.001502. Wang S, Li C, Sun P, Shi J, Wu X, Liu C, Peng Z, Han H, Xu
Tischer I, Rasch R, Tochtermann G. 1974. Characterization S, Yang Y, et al. 2021a. PCV2 triggers PK-15 cell apopto-
of papovavirus-and picornavirus-like particles in perma- sis through the PLC-IP3R-Ca(2+) signaling pathway. Front
nent pig kidney cell lines. Zentralbl Bakteriol Orig A. Microbiol. 12:674907. doi: 10.3389/fmicb.2021.674907.
226(2):153–167. Wang D, Mai J, Yang Y, Xiao CT, Wang N. 2022a. Current
Todd D, Scott AN, Fringuelli E, Shivraprasad HL, Gavier- knowledge on epidemiology and evolution of novel por-
Widen D, Smyth JA. 2007. Molecular characterization of cine circovirus 4. Vet Res. 53(1):38. doi: 10.1186/s13567-
novel circoviruses from finch and gull. Avian Pathol. 022-01053-w.
36(1):75–81. doi: 10.1080/03079450601113654. Wang S, Ren X, Li J, Lin C, Zhou J, Zhou J, Gu J., 2020.
Todd D, Weston J, Ball NW, Borghmans BJ, Smyth JA, NAP1L4 inhibits porcine circovirus type 2 replication via
Gelmini L, Lavazza A. 2001. Nucleotide sequence-based IFN-beta signaling pathway. Vet Microbiol. 246:108692.
identification of a novel circovirus of canaries. Avian doi: 10.1016/j.vetmic.2020.108692.
Pathol. 30(4):321–325. doi: 10.1080/03079450120066322. Wei L, Kwang J, Wang J, Shi L, Yang B, Li Y, Liu J. 2008.
Tu PY, Tsai PC, Lin YH, Liu PC, Chang HL, Kuo TY, Chung Porcine circovirus type 2 induces the activation of nucle-
WB. 2015. Expression profile of Toll-like receptor mRNA ar factor kappa B by IkappaBalpha degradation. Virology.
in pigs co-infected with porcine reproductive and respi- 378(1):177–184. doi: 10.1016/j.virol.2008.05.013.
ratory syndrome virus and porcine circovirus type 2. Res Wei R, Trus I, Yang B, Huang L, Nauwynck HJ. 2018. Breed
Vet Sci. 98:134–141. doi: 10.1016/j.rvsc.2014.12.003. differences in PCV2 uptake and disintegration in porcine
Vargas-Bermudez DS, Mogollon JD, Jaime J. 2022. The preva- monocytes. Viruses. 10(10):562. doi: 10.3390/v10100562.
lence and genetic diversity of PCV3 and PCV2 in Colombia Wei, R., Van Renne, N., Nauwynck, H.J., 2019. Strain-
and PCV4 survey during 2015-2016 and 2018-2019. dependent porcine circovirus type 2 (PCV2) entry and
Pathogens. 11(6):633. doi: 10.3390/pathogens11060633. replication in T-lymphoblasts. Viruses, 911, 813. doi:
Vincent IE, Balmelli C, Meehan B, Allan G, Summerfield A, 10.3390/v11090813.
McCullough KC. 2007. Silencing of natural interferon Wei L, Zhu S, Wang J, Liu J. 2012. Activation of the phos-
producing cell activation by porcine circovirus type 2 phatidylinositol 3-kinase/Akt signaling pathway during
DNA. Immunology. 120(1):47–56. doi: porcine circovirus type 2 infection facilitates cell survival
10.1111/j.1365-2567.2006.02476.x. and viral replication. J Virol. 86(24):13589–13597. doi:
Vincent IE, Carrasco CP, Guzylack-Piriou L, Herrmann B, 10.1128/JVI.01697-12.
McNeilly F, Allan GM, Summerfield A, McCullough KC. Wei L, Zhu Z, Wang J, Liu J. 2009. JNK and p38 mitogen-ac-
2005. Subset-dependent modulation of dendritic cell ac- tivated protein kinase pathways contribute to porcine
tivity by circovirus type 2. Immunology. 115(3):388–398. circovirus type 2 infection. J Virol. 83(12):6039–6047. doi:
doi: 10.1111/j.1365-2567.2005.02165.x. 10.1128/JVI.00135-09.
Vincent IE, Carrasco CP, Herrmann B, Meehan BM, Allan Wikstrom FH, Meehan BM, Berg M, Timmusk S, Elving J, Fuxler
GM, Summerfield A, McCullough KC. 2003. Dendritic L, Magnusson M, Allan GM, McNeilly F, Fossum C. 2007.
cells harbor infectious porcine circovirus type 2 in the Structure-dependent modulation of alpha interferon pro-
absence of apparent cell modulation or replication of duction by porcine circovirus 2 oligodeoxyribonucleotide
the virus. J Virol. 77(24):13288–13300. doi: 10.1128/ and CpG DNAs in porcine peripheral blood mononuclear
jvi.77.24.13288-13300.2003. cells. J Virol. 81(10):4919–4927. doi: 10.1128/JVI.02797-06.
Walker FC, Sridhar PR, Baldridge MT. 2021. Differential roles Wu H, Hou C, Wang Z, Meng P, Chen H, Cao H. 2022. First
of interferons in innate responses to mucosal viral infec- complete genomic sequence analysis of porcine circovi-
tions. Trends Immunol. 42(11):1009–1023. doi: 10.1016/j. rus type 4 (PCV4) in wild boars. Vet Microbiol. 273:109547.
it.2021.09.003. doi: 10.1016/j.vetmic.2022.109547.
18 E. FEHÉR ET AL.

Wu X, Wang Z, Qiao D, Yuan Y, Han C, Yang N, Li R, Du Q, Zhang H, Liu C, Cheng S, Wang X, Li W, Charreyre C,
Tong D, Huang Y. 2021. Porcine circovirus type 2 infec- Audonnet JC, He Q. 2013. Porcine CD74 is involved in
tion attenuates the K63-linked ubiquitination of STING the inflammatory response activated by nuclear factor
to inhibit IFN-beta induction via p38-MAPK pathway. Vet kappa B during porcine circovirus type 2 (PCV-2) infec-
Microbiol. 258:109098. doi: 10.1016/j.vetmic.2021.109098. tion. Arch Virol. 158(11):2285–2295. doi: 10.1007/s00705-
Wu X, Wang X, Shi T, Luo L, Qiao D, Wang Z, Han C, Du Q, Tong 013-1750-3.
D, Huang Y. 2019. Porcine Circovirus Type 2 Rep Enhances Zhang L, Qiu S, Lu M, Huang C, Lv Y. 2020b. Nuclear trans-
IL-10 Production in Macrophages via Activation of p38-MAPK porter karyopherin subunit alpha 3 levels modulate
Pathway. Viruses. 11(12):1141. doi: 10.3390/v11121141. Porcine circovirus type 2 replication in PK-15 cells.
Xiang QW, Wang X, Xie ZJ, Sun YN, Zhu YL, Wang SJ, Liu Virology. 548:31–38. doi: 10.1016/j.virol.2020.06.003.
HJ, Jiang SJ. 2012. ORF3 of duck circovirus: a novel pro- Zhang P, Shen H, Liu X, Wang S, Liu Y, Xu Z, Song C. 2020c.
tein with apoptotic activity. Vet Microbiol. 159(1-2):251– Porcine circovirus type 3 cap inhibits type i interferon
256. doi: 10.1016/j.vetmic.2012.03.045. induction through interaction with G3BP1. Front Vet Sci.
Xu T, Chen XM, Fu Y, Ai Y, Wang DM, Wei ZY, Li XS, Zheng 7:594438. doi: 10.3389/fvets.2020.594438.
LL, Chen HY. 2022. Cross-species transmission of an Zhang Y, Sun R, Geng S, Shan Y, Li X, Fang W. 2019. Porcine
emerging porcine circovirus (PCV4): First molecular de- circovirus type 2 induces ORF3-independent mitochon-
tection and retrospective investigation in dairy cows. Vet drial apoptosis via PERK activation and elevation of cy-
Microbiol. 273:109528. doi: 10.1016/j.vetmic.2022.109528. tosolic calcium. J Virol. 93(7):e01784–01718. doi: 10.1128/
Yang K, Jiao Z, Zhou D, Guo R, Duan Z, Tian Y. 2019. JVI.01784-18.
Development of a multiplex PCR to detect and discrim- Zhou J, Dai Y, Lin C, Zhang Y, Feng Z, Dong W, Jin Y, Yan Y,
inate porcine circoviruses in clinical specimens. BMC Zhou J, Gu J. 2020. Nucleolar protein NPM1 is essential for
Infect Dis. 19(1):778. doi: 10.1186/s12879-019-4398-0. circovirus replication by binding to viral capsid. Virulence.
Yang Z, Marthaler DG, Rovira A. 2022. Frequency of porcine 11(1):1379–1393. doi: 10.1080/21505594.2020.1832366.
circovirus 3 detection and histologic lesions in clinical Zhou Y, Qi B, Gu Y, Xu F, Du H, Li X, Fang W. 2016. Porcine
samples from swine in the United States. J Vet Diagn circovirus 2 deploys PERK pathway and GRP78 for its en-
Invest. 34(4):602–611. doi: 10.1177/10406387221099538. hanced replication in PK-15 cells. Viruses. 8(2):56. doi:
Yasukawa H, Ohishi M, Mori H, Murakami M, Chinen T, Aki 10.3390/v8020056.
D, Hanada T, Takeda K, Akira S, Hoshijima M, et al. 2003. Zhu X, Bai J, Liu P, Wang X, Jiang P. 2016. Suppressor of
IL-6 induces an anti-inflammatory response in the ab- cytokine signaling 3 plays an important role in porcine
sence of SOCS3 in macrophages. Nat Immunol. 4(6):551– circovirus type 2 subclinical infection by downregulating
556. doi: 10.1038/ni938. proinflammatory responses. Sci Rep. 6:32538. doi:
Zhang HH, Hu WQ, Li JY, Liu TN, Zhou JY, Opriessnig T, Xiao 10.1038/srep32538.
CT. 2020a. Novel circovirus species identified in farmed Zhu B, Zhou Y, Xu F, Shuai J, Li X, Fang W. 2012. Porcine
pigs designated as Porcine circovirus 4, Hunan province, circovirus type 2 induces autophagy via the AMPK/ERK/
China. Transbound Emerg Dis. 67(3):1057–1061. doi: TSC2/mTOR signaling pathway in PK-15 cells. J Virol.
10.1111/tbed.13446. 86(22):12003–12012. doi: 10.1128/JVI.01434-12.

You might also like