ArticlePDF Available

Bovine Papillomavirus Type 1 Infection in an Equine Congenital Papilloma

Authors:
  • Equine Practitioner

Abstract and Figures

Papillomas are benign epithelial lesions protruding on the epithelial surfaces as finger-like or warty projections. These lesions are often caused by papillomavirus (PV) infection. Congenital papillomas have been reported in foals. However, to date, no evidence of PV infection has been provided. In the present paper, we describe the main clinical-pathological features of a congenital papilloma observed in a foal. In addition, biomolecular tests demonstrated BPV1 infection in the case under study. Such data stimulate further investigations, even on archived samples, aiming to clarifying the etiology of equine congenital papilloma and the clinical relevance, if any, of BPV1 vertical transmission in horses.
Content may be subject to copyright.
Citation: Maggi, R.; De Paolis, L.; De
Santis, D.; Vellone, V.G.; De Ciucis,
C.G.; Fruscione, F.; Mazzocco, K.;
Ghelardi, A.; Marruchella, G.;
Razzuoli, E. Bovine Papillomavirus
Type 1 Infection in an Equine
Congenital Papilloma. Pathogens
2023,12, 1059. https://doi.org/
10.3390/pathogens12081059
Academic Editors: Laura Gallina
and Magda Dunowska
Received: 17 April 2023
Revised: 14 August 2023
Accepted: 15 August 2023
Published: 18 August 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
pathogens
Case Report
Bovine Papillomavirus Type 1 Infection in an Equine
Congenital Papilloma
Raffaella Maggi 1, Livia De Paolis 2, Daria De Santis 3, Valerio Gaetano Vellone 4, Chiara Grazia De Ciucis 2,
Floriana Fruscione 2, Katia Mazzocco 4, Alessandro Ghelardi 5, Giuseppe Marruchella 6, *
and Elisabetta Razzuoli 2, *
1Veterinary Practitioner, Via Cassia 829, 00189 Rome, Italy; raffy.maggi@hotmail.it
2Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, National Reference Center of
Veterinary and Comparative Oncology (CEROVEC), Piazza Borgo Pila 29/34, 16129 Genova, Italy;
livia.depaolis@izsto.it (L.D.P.); chiaragrazia.deciucis@izsto.it (C.G.D.C.); floriana.fruscione@izsto.it (F.F.)
3Veterinary Practitioner, Via San Manno 19, 03024 Cepranno, Italy; dariadesantisvet@gmail.com
4U.O.C. Anatomia Patologica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
valeriovellone@gaslini.org (V.G.V.); katiamazzocco@gaslini.org (K.M.)
5Azienda Usl Toscana-Ovest, UOC Ostetricia e Ginecologia, Ospedale Apuane, 54100 Massa, Italy;
ghelardi.alessandro@gmail.com
6Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy
*Correspondence: gmarruchella@unite.it (G.M.); elisabetta.razzuoli@izsto.it (E.R.);
Tel.: +39-086-126-6932 (G.M.); +39-010-542-274 (E.R.)
Abstract:
Papillomas are benign epithelial lesions protruding on the epithelial surfaces as finger-like
or warty projections. These lesions are often caused by papillomavirus (PV) infection. Congenital
papillomas have been reported in foals. However, to date, no evidence of PV infection has been
provided. In the present paper, we describe the main clinical–pathological features of a congenital
papilloma observed in a foal. In addition, biomolecular tests demonstrated BPV1 infection in the
case under study. Such data stimulate further investigations, even on archived samples, aiming to
clarifying the etiology of equine congenital papilloma and the clinical relevance, if any, of BPV1
vertical transmission in horses.
Keywords: horse; congenital papilloma; bovine papillomaviruses
1. Introduction
Papillomas are benign epithelial lesions protruding from the epithelial surfaces as
finger-like or warty projections [
1
]. Papillomas are usually caused by papillomaviruses
(PVs, fam. Papillomaviridae), non-enveloped and host-adapted viruses provided with a
circular dsDNA genome [
2
]. Microscopically, cutaneous papillomas consist of arborizing
fibrovascular fronds lined with a thickened epidermis, more prominent within the stratum
spinosum. Large, variably shaped keratohyalin granules, and degenerating cells (so called
koilocytes) are commonly observed. Moreover, a few intranuclear viral inclusions can be
detected throughout the stratum spinosum and granulosum [3,4].
As far as the horse is concerned, three syndromes caused by Equus caballus papillo-
maviruses (EcPVs) have been described:
classic equine viral papillomatosis via EcPV-1, which usually affects the muzzle and
lips of young horses (<3 years of age) and regresses spontaneously within 2–3 months;
equine genital papillomas caused by EcPV-2, which affect older horses and do not
resolve spontaneously;
equine ear papillomas caused by EcPV3-6, which present with bilateral and symmet-
ric white hyperkeratotic plaques confined to the pinnae, and rarely resolve sponta-
neously [3].
Pathogens 2023,12, 1059. https://doi.org/10.3390/pathogens12081059 https://www.mdpi.com/journal/pathogens
Pathogens 2023,12, 1059 2 of 9
In addition, equids develop sarcoids after bovine PV (BPV) type 1 and 2 infection, this
being among the very few cases of cross-species PV infection reported so far. Sarcoids are
locally invasive fibroblastic tumors, and they represent the most common and economically
relevant neoplasms in horses [3,5].
Congenital papillomas have also been reported in foals. To date, no evidence of
EcPV infection has been provided, suggesting that equine congenital papillomas might be
regarded as epidermal hamartomas [
6
,
7
]. The present paper describes the main clinical–
pathological features of a congenital papilloma recently observed in a foal. The presence of
PV infection was also investigated.
2. Case Description
In March 2022, a male, newborn quarter horse foal underwent a routine clinical
examination. The foal was born on his due date, and appeared healthy, bright, and alert.
However, a spheric, pedunculated, and non-painful mass was observed, which hung from
the upper lip and did not prevent suckling (Figure 1).
Pathogens 2023, 12, x FOR PEER REVIEW 2 of 10
In addition, equids develop sarcoids after bovine PV (BPV) type 1 and 2 infection,
this being among the very few cases of cross-species PV infection reported so far. Sarcoids
are locally invasive broblastic tumors, and they represent the most common and eco-
nomically relevant neoplasms in horses [3,5].
Congenital papillomas have also been reported in foals. To date, no evidence of EcPV
infection has been provided, suggesting that equine congenital papillomas might be re-
garded as epidermal hamartomas [6,7]. The present paper describes the main clinical
pathological features of a congenital papilloma recently observed in a foal. The presence
of PV infection was also investigated.
2. Case Description
In March 2022, a male, newborn quarter horse foal underwent a routine clinical ex-
amination. The foal was born on his due date, and appeared healthy, bright, and alert.
However, a spheric, pedunculated, and non-painful mass was observed, which hung from
the upper lip and did not prevent suckling (Figure 1).
Figure 1. Foal three days after birth. A globoid lesion is clearly seen, aached to the upper lip.
The mass was surgically removed three days later. To achieve this, the foal was drug-
free restrained [8], 1 mL of 2% lidocaine chlorydrate was locally injected around the base
of the peduncle, and the mass was fully removed and promptly xed in 10% neutral-bu-
ered formalin (Figure 2). The skin incision healed without any complication, and the
stiches were removed 10 days after surgery.
Figure 1. Foal three days after birth. A globoid lesion is clearly seen, attached to the upper lip.
The mass was surgically removed three days later. To achieve this, the foal was drug-
free restrained [
8
], 1 mL of 2% lidocaine chlorydrate was locally injected around the base of
the peduncle, and the mass was fully removed and promptly fixed in 10% neutral-buffered
formalin (Figure 2). The skin incision healed without any complication, and the stiches
were removed 10 days after surgery.
Formalin-fixed samples were embedded in paraffin and routinely processed in prepa-
ration for histopathological examination (hematoxylin and eosin stain, H and E). Micro-
scopically, the mass consisted of several finger-like fibro-vascular projections, which were
covered by a thickened epidermis with prominent acanthosis and orthokeratotic hyperker-
atosis (Figure 3). We observed evidence of koilocytosis, keratohyalin granule abnormalities,
and a pilosebaceous unit (Figure 4).
Pathogens 2023,12, 1059 3 of 9
Pathogens 2023, 12, x FOR PEER REVIEW 3 of 10
(a) (b)
Figure 2. Excised lesion after formalin xation. The mass is about 3 cm in diameter and shows a
verrucous, “true-like” surface (a). In the cut section, the arborized stromal scaold can be clearly
observed (b).
Formalin-xed samples were embedded in paran and routinely processed in prep-
aration for histopathological examination (hematoxylin and eosin stain, H and E). Micro-
scopically, the mass consisted of several nger-like bro-vascular projections, which were
covered by a thickened epidermis with prominent acanthosis and orthokeratotic hyper-
keratosis (Figure 3). We observed evidence of koilocytosis, keratohyalin granule abnor-
malities, and a pilosebaceous unit (Figure 4).
Based on the clinical history, gross ndings, and histopathological features, the diag-
nosis of equine congenital papilloma was made.
Figure 3. Histopathological examination of the excised lesion. In this nger-like projection, a central
brovascular backbone is clearly seen, which contains several blood and lymphatic vessels. The
stroma is covered by a thickened epidermis, the stratum spinosum being markedly expanded. H
and E stain. Final magnication ×200.
Figure 2.
Excised lesion after formalin fixation. The mass is about 3 cm in diameter and shows a
verrucous, “truffle-like” surface (
a
). In the cut section, the arborized stromal scaffold can be clearly
observed (b).
Pathogens 2023, 12, x FOR PEER REVIEW 3 of 10
(a) (b)
Figure 2. Excised lesion after formalin xation. The mass is about 3 cm in diameter and shows a
verrucous, “true-like” surface (a). In the cut section, the arborized stromal scaold can be clearly
observed (b).
Formalin-xed samples were embedded in paran and routinely processed in prep-
aration for histopathological examination (hematoxylin and eosin stain, H and E). Micro-
scopically, the mass consisted of several nger-like bro-vascular projections, which were
covered by a thickened epidermis with prominent acanthosis and orthokeratotic hyper-
keratosis (Figure 3). We observed evidence of koilocytosis, keratohyalin granule abnor-
malities, and a pilosebaceous unit (Figure 4).
Based on the clinical history, gross ndings, and histopathological features, the diag-
nosis of equine congenital papilloma was made.
Figure 3. Histopathological examination of the excised lesion. In this nger-like projection, a central
brovascular backbone is clearly seen, which contains several blood and lymphatic vessels. The
stroma is covered by a thickened epidermis, the stratum spinosum being markedly expanded. H
and E stain. Final magnication ×200.
Figure 3.
Histopathological examination of the excised lesion. In this finger-like projection, a central
fibrovascular backbone is clearly seen, which contains several blood and lymphatic vessels. The
stroma is covered by a thickened epidermis, the stratum spinosum being markedly expanded. H and
E stain. Final magnification ×200.
Based on the clinical history, gross findings, and histopathological features, the diag-
nosis of equine congenital papilloma was made.
Total DNA was extracted from three 5
µ
m thick sections of formalin-fixed and paraffin-
embedded (FFPE) samples using the AllPrep DNA FFPE kit (Qiagen, Milan, Italy), accord-
ing to the manufacturer’s instruction. The DNA concentration was measured using the
Qubit 3 fluorimeter (Thermo Fisher Scientific, Waltham, MA, USA). Aiming to investigate
the presence of the EcPV2/9/10 and BPV1/2/13 genome, 100 ng of DNA, 200 nM of
the probe, and 100 nM of each primer were added to 20
µ
L of the iTaq Universal Probes
Supermix (BioRad, Milan, Italy), in a total volume of 25
µ
L. Equine beta-2-microglobulin
(
β
2M) was used as the gene reference. Specific primer and probe sequences are reported in
Pathogens 2023,12, 1059 4 of 9
Table 1. Internal controls (block blanks, extraction blanks, and positive controls) were used
for each analytical session. Real-time PCR was performed using a CFX96 Real-Time System
(BioRad, Milan, Italy). A threshold cycle of 38 was set as the cut off for sample positivity.
All samples were tested in duplicate.
Pathogens 2023, 12, x FOR PEER REVIEW 4 of 10
Figure 4. In these sections, evidence of koilocytosis was observed. The right panels reveal a close-
up view of the inset. The arrows indicate some selected koilocytes. H and E stain. Final magnica-
tion ×200.
Total DNA was extracted from three 5 µm thick sections of formalin-xed and par-
an-embedded (FFPE) samples using the AllPrep DNA FFPE kit (Qiagen, Milan, Italy),
according to the manufacturer’s instruction. The DNA concentration was measured using
the Qubit 3 uorimeter (Thermo Fisher Scientic, Waltham, MA, USA). Aiming to inves-
tigate the presence of the EcPV2/9/10 and BPV1/2/13 genome, 100 ng of DNA, 200 nM of
the probe, and 100 nM of each primer were added to 20 µL of the iTaq Universal Probes
Supermix (BioRad, Milan, Italy), in a total volume of 25 µL. Equine beta-2-microglobulin
(β2M) was used as the gene reference. Specic primer and probe sequences are reported
in Table 1. Internal controls (block blanks, extraction blanks, and positive controls) were
used for each analytical session. Real-time PCR was performed using a CFX96 Real-Time
System (BioRad, Milan, Italy). A threshold cycle of 38 was set as the cut o for sample
positivity. All samples were tested in duplicate.
Table 1. Primer set and probes used in the virological investigations.
Target Gene Primer Sequences Amplicon
Length
Accession Num-
ber
EcPV2-L1 F-5-TTGTCCAGGAGAGGGGTTAG-3 80 NC_012123.1
R-5-TGCCTTCCTTTTCTTGGTGG-3
p-EcPV2-L1 FAM-CGTCCAGCACCTTCGACCACCA-TAMRA
EcPV9-L1 F-5-TTC ATC CCA GCT TGA GAC CA-3 116 MN117918.1 R-5-GCA GAT CAA TGG TCC AGA AGG-3
p-EcPV9-L1 p-FAM-ATT GCC TCC TCA GCC ACC CG-TAMRA
EcPV10-L1 F-5-GTG TCA CAG GTA ACC CCC TG-3 174 OP870083 R-5-AAG CGT GTC TTC CTC CAG TG-3
p-EcPV10-L1 p-FAM-TGC TGG TGG GTT GCA AGC CC-TAMRA
BPV1-L1 F-5-CAG GAC TGT TCA CAA CCC AA-3 96 NC_001522.1
R-5-CCC AGT TAC AGT ACC TCC AA-3
p-BPV1-L1 p-FAM-TGC AGG TGT CCA GAG GGC AG-TAMRA
BPV2-L1 F-5-ACA GCC CGT CCA TGT GTT A-3 115 MF045490
R-5-TCA GCA GCA CCA AAC CCT AT-3
Figure 4.
In these sections, evidence of koilocytosis was observed. The right panels reveal a close-up
view of the inset. The arrows indicate some selected koilocytes. H and E stain. Final magnification
×200.
Table 1. Primer set and probes used in the virological investigations.
Target Gene Primer Sequences Amplicon Length Accession Number
EcPV2-L1 F-50-TTGTCCAGGAGAGGGGTTAG-30
80 NC_012123.1
R-50-TGCCTTCCTTTTCTTGGTGG-30
p-EcPV2-L1 FAM-CGTCCAGCACCTTCGACCACCA-TAMRA
EcPV9-L1 F-50-TTC ATC CCA GCT TGA GAC CA-30
116 MN117918.1
R-50-GCA GAT CAA TGG TCC AGA AGG-30
p-EcPV9-L1 p-FAM-ATT GCC TCC TCA GCC ACC CG-TAMRA
EcPV10-L1 F-50-GTG TCA CAG GTA ACC CCC TG-30
174 OP870083
R-50-AAG CGT GTC TTC CTC CAG TG-30
p-EcPV10-L1 p-FAM-TGC TGG TGG GTT GCA AGC CC-TAMRA
BPV1-L1 F-50-CAG GAC TGT TCA CAA CCC AA-30
96 NC_001522.1
R-50-CCC AGT TAC AGT ACC TCC AA-30
p-BPV1-L1 p-FAM-TGC AGG TGT CCA GAG GGC AG-TAMRA
BPV2-L1 F-50-ACA GCC CGT CCA TGT GTT A-30
115 MF045490
R-50-TCA GCA GCA CCA AAC CCT AT-30
p-BPV2-L1 p-FAM-AGA AAA TGG TGC GTG TCC TCC T-TAMRA
BPV1-E5 F-50-TGCTTCAATGCAACTGCTGCT-30
77 MZ310894
R-50-AGGAGCACTCAAAATGATCCCAG-30
p-BPV1-E5 p-FAM-ACTCTTGTTTTTTCTTGTA-TAMRA
BPV1-E6 F-50-TGCTACTGTGGGGGCAAACT-30
110 MZ310894
R-50-CAGTCGTAGCAGCGTCCTCT-30
p-BPV1-E6 p-FAM-AGCCTTTCTGCAAAACCAGAGCT-TAMRA
BPV1-E7 F-50-GCTGTGGAAACTGCGGAAAA-30
122 MZ310894
R-50-GCGAGATTCACAACGTGGAC-30
p-BPV1-E7 p-FAM-GCTGACTTTTGCAGTGAAGACCAGC-TAMRA
BPV13-L1 F-50-GCA CCC CAC TTT TAA TGC CT-30
87 NC_030795
R-50-TCC TGT TTG CTT CCT GTC ATC-30
p-BPV13-L1 p-FAM-AGG AAA GTG ACC AGC CAA ACA ACA-TAMRA
B2M (Beta2-microglobulin) F-50-GGCTACTCTCCCTGACTGG-30
135 NM_001082502.3
R-50-TCAATCTCAGGCGGATGGAA-30
p-B2M p-FAM-ACTCACGTCACCCAGCAGAGA-TAMRA
Pathogens 2023,12, 1059 5 of 9
Real-time PCR demonstrated the presence of the BPV1 genome, while it gave a nega-
tive result for all the other PVs herein investigated (see Table 2and Figure S1 for details).
Table 2.
Real-time PCR targeting the PV genome. Data are expressed as Cq mean value
±
stan-
dard deviation.
Sample Real-Time PCR
B2M EcPV2-L1 EcPV9-L1 EcPV10-L1 BPV2-L1 BPV13-L1
29.4 ±0.1 >38 >38 >38 >38 >38
B2M BPV1-E5 BPV1-E6 BPV1-E7 BPV1-L1
29.1 ±0.1 21.5 ±0.2 21.7 ±0.4 21.5 ±0.2 22.5 ±0.5
Thereafter, to evaluate the expression of BPV1-L1 and oncogenes (namely, E5, E6, E7,
and L1), total RNA was extracted from three 5
µ
m thick sections of the FFPE samples
using the Maxwell
®
RSC RNA FFPE kit (Promega, Madison, WI, USA), according to the
manufacturer’s instructions. RNA concentration was evaluated using the Qubit 3 fluo-
rimeter (Thermo Fisher, Milan, Italy). The reverse transcription (RT) step was performed
using the Reliance Select cDNA Synthesis Kit (BioRad, Milan, Italy), adding 100 ng of
RNA. Then,
5µL
of 1:3 diluted complementary DNA (cDNA) was added to 20
µ
L PCR
mixture at the final concentration of 1
×
master mix (iTaq Universal Probes Supermix,
Bio-Rad, Irvine, CA, USA), with the following thermal profile: 95
C for 10
0
, then 39 cycles
of
95 C
for 15
00
and 60
C for 60
00
in a CFX96
Real-Time System. To exclude genomic
DNA contamination, a digestion using the DNase I RNase free kit (Qiagen, Milan, Italy)
was performed. The specific primer set and probes used for testing the expression of
BPV1 oncogenes are reported in Table 1. RT-Real-Time PCR demonstrated that BPV1 L1
(Cq = 37.1
±
0.3), E5 (
Cq = 30.1 ±0.2
), E6 (Cq = 34.2
±
0.2), and E7 (Cq = 33.8
±
0.3) were
expressed at the mRNA level (Table 3).
Table 3. BPV1 oncogene expression. Data are expressed as Cq mean value ±standard deviation.
Sample Real-Time PCR
B2M BPV1-E5 BPV1-E6 BPV1-E7 BPV1-L1
31.4 ±0.2 30.1 ±0.2 34.2 ±0.2 21.5 ±0.2 37.1 ±0.3
After this test, to evaluate the mRNA localization, RNA in situ hybridization was
performed using the RNAscope kit (Advanced Cell Diagnostics Inc., Hayward, CA, USA),
according to the manufacturer’s guidelines, as described in [
9
]. The RNAscope probe used
was the V-BPV-E-bovine-papillomavirus-1 complete genome E5 E6 E7 (Catalog Number
416831) and was designed to detect the mRNA expression of the E5, E6, and E7 genes
(National Center for Biotechnology Information Reference Sequence NC_001522.1). FFPE
tissue sections (of 5
µ
m thickness) were deparaffinized in xylene, and were subsequently
dehydrated in 100% ethanol. Tissue sections were exposed to hydrogen peroxide for 10 min,
followed by incubation in a target retrieval reagent maintained at a boiling temperature
(98 to 102
C) using a hot plate for 15 min, rinsed in deionized water, and immediately
treated with Protease Plus at 40
C for 30 min in a hybridization oven (TopBrite- Resnova,
RM, Italy). The tissue sections were then incubated at 40
C with a target probe of BPV-1
for 2 h. The target–probe hybridization was followed by a series of target-specific signal-
amplification steps. After each hybridization step, slides were washed with wash buffer
at room temperature twice, followed by staining with Fast Red dye. The sections were
then counterstained with 50% hematoxylin staining solution, and dehydrated using xylene,
before being mounted with a mounting medium (Eukitt). The positive signals were present
in the form of punctate cytoplasmic and nuclear red staining that was higher than the signal
on the negative control slide. Assays using FFPE specimens were performed in parallel
Pathogens 2023,12, 1059 6 of 9
with positive and negative control probes (positive probe: Ec-PPIB; negative control probe:
DapB), to ensure interpretable results (Figure 5).
Pathogens 2023, 12, x FOR PEER REVIEW 6 of 10
was the V-BPV-E-bovine-papillomavirus-1 complete genome E5 E6 E7 (Catalog Number
416831) and was designed to detect the mRNA expression of the E5, E6, and E7 genes
(National Center for Biotechnology Information Reference Sequence NC_001522.1). FFPE
tissue sections (of 5 µm thickness) were deparanized in xylene, and were subsequently
dehydrated in 100% ethanol. Tissue sections were exposed to hydrogen peroxide for 10
min, followed by incubation in a target retrieval reagent maintained at a boiling tempera-
ture (98 to 102 °C) using a hot plate for 15 min, rinsed in deionized water, and immediately
treated with Protease Plus at 40 °C for 30 min in a hybridization oven (TopBrite- Resnova,
RM, Italy). The tissue sections were then incubated at 40 °C with a target probe of BPV-1
for 2 h. The target–probe hybridization was followed by a series of target-specic signal-
amplication steps. After each hybridization step, slides were washed with wash buer
at room temperature twice, followed by staining with Fast Red dye. The sections were
then counterstained with 50% hematoxylin staining solution, and dehydrated using xy-
lene, before being mounted with a mounting medium (Euki). The positive signals were
present in the form of punctate cytoplasmic and nuclear red staining that was higher than
the signal on the negative control slide. Assays using FFPE specimens were performed in
parallel with positive and negative control probes (positive probe: Ec-PPIB; negative con-
trol probe: DapB), to ensure interpretable results (Figure 5).
Figure 5. RNAscope assay. (A) Tissue sections hybridized with the probe targeting BPV-1
E5-E6-E7 mRNA (V-BPV-E). A close-up view of the inset is visible in the right panel. Spe-
cic staining was observed in the congenital papilloma under investigation. In detail, scat-
tered red dots were detected within the cytoplasms (empty circles) and nuclei (black ar-
row) of epithelial cells. (B) On the contrary, no specic staining was observed in the neg-
ative control. Final magnication: ×400.
3. Discussion
Papillomaviruses are regarded as very important oncogenic viruses, able to induce
benign and often self-limiting epithelial neoplasms, which can occasionally progress to
malignancy [10,11]. In humans, cervical cancer represents the most common PV-associ-
ated cancer [12]. Moreover, the role of human PVs as causative agents of anogenital, head,
Figure 5.
RNAscope assay. (
A
) Tissue sections hybridized with the probe targeting BPV-1 E5-E6-E7
mRNA (V-BPV-E). A close-up view of the inset is visible in the right panel. Specific staining was
observed in the congenital papilloma under investigation. In detail, scattered red dots were detected
within the cytoplasms (empty circles) and nuclei (black arrow) of epithelial cells. (
B
) On the contrary,
no specific staining was observed in the negative control. Final magnification: ×400.
3. Discussion
Papillomaviruses are regarded as very important oncogenic viruses, able to induce
benign and often self-limiting epithelial neoplasms, which can occasionally progress to
malignancy [
10
,
11
]. In humans, cervical cancer represents the most common PV-associated
cancer [
12
]. Moreover, the role of human PVs as causative agents of anogenital, head, and
neck cancers is widely accepted [
13
]. Likewise, PVs have been associated with malignant
neoplasms in several animal species [
11
]. In horses, a growing body of evidence suggests
that EcPV2 infection contributes to the etiopathogenesis of squamous cell carcinomas,
which can affect the external genitalia, and laryngeal and gastric mucosa [
14
,
15
]. Other
types of equine papillomaviruses have been associated with different lesions. In particular,
papillomas seem to be caused by EcPV1, EcPV3-6, and EcPV8, while EcPV9 and EcPV10, to
date, are not associated with specific pathology [1618].
Papillomaviruses are strictly species-specific and show a distinct tropism towards
epithelial cells. Delta-PVs (e.g., BPV1 and BPV2) are exceptions to this general rule, as they
have a wider host range and can infect different cell types [
11
,
19
]. A recent case report by
Savini and co-workers (2020) demonstrated the ability of BPV1 to cause proliferative lesions
in sheep. These results suggest a different pathogenetic role of BPV-1, and confirm the
ability of BPV1 to infect different species, also causing a non-sarcoid outcome on cutaneous
surfaces [
20
]. Notably, phylogenetic investigations revealed that BPV1 originated in cattle,
while multiple cross-species transmissions occurred into horses [
21
]. The causal association
of BPV1 and BPV2 infection with equid sarcoid has been unequivocally demonstrated [
22
].
It was assumed that BPV1/2 infections were strictly confined to dermal fibroblasts in the
Pathogens 2023,12, 1059 7 of 9
horses, wherein they reside in a non-productive episomal form. Consequently, horses have
been long considered a dead-end host for BPV1/2 [
14
]. However, BPV1 infection has been
demonstrated to involve the epidermis of the horse, where it may be productive, albeit at
low level [
23
]. In addition, BPV1 can early-infect PBMCs in young horses, likely inducing a
viremic phase [
24
], and the BPV1 genome has been detected in the placenta and blood of
newborn foals [25].
Overall, biomolecular investigations clearly demonstrated BPV1 infection in the equine
congenital papilloma under study. As a matter of fact, the detection of BPV1 oncogene
mRNA and its cytoplasmatic localization ruled out any incidental contamination at the time
of sampling and/or during laboratory tests. Moreover, according to the literature, such data
would be unlikely to result from the selective infection of PBMCs, considering the high viral
load detected (the BPV1 DNA was 100-fold more concentrated than the equine genome,
when compared with the
β
2M), as well as the transcription of the L1 gene [
24
,
26
,
27
].
The present case report is apparently in contrast with those currently available in the
literature [
6
,
7
]. However, we remark that White et al. [
6
] and Postey et al. [
7
] only ruled
out the presence of EcPVs in equine congenital papillomas, while no data have ever been
provided about BPV1 infection in such lesions.
Although PV infections mainly spread through horizontal transmission, a growing in-
terest exists around intrauterine and perinatal transmission. In humans, the PV (human PV,
HPV) genome has been detected in the amniotic fluid, placenta, and umbilical cord
[28,29]
,
and transplacental infection might occur through the hematogenous or ascending route,
from the maternal genital tract [
30
]. The hypothesis of vertical transmission has been further
corroborated in cattle, as BPV2 was shown to productively infect the uterine epithelium and
chorionic placenta [
31
,
32
]. Accordingly, some evidence supports BPV vertical transmission
in sheep [
33
]. The mechanism of BPV transmission in horses is still debated; the BPV
genome has been detected in the equine placenta, and intrauterine transmission has been
speculated in this animal species [
25
]. Moreover, Silva and co-workers demonstrated the
virus’ presence in the PBMC and semen of healthy horses [
27
,
34
]. Therefore, recently, it
was speculated that, as in humans, infection could be ascribable to oocyte fecundation [
35
].
Considering this, the detection of BPV1 in congenital papillomas argues in favor of its
transplacental transmission in horses, stimulating further investigation to identify the risk
factors, prevalence, and clinical relevance of such event.
Congenital papillomas have been repeatedly reported in animals (including horses) and
regarded as hamartomatous lesions, with no viral origin having been demonstrated
[1,6,7]
. It
is worth noting that Roperto et al. [
33
] recently reported BPV-associated congenital lesions
in lambs after transplacental infection. Of course, the conclusive demonstration of the viral
etiology (if any) of equine congenital papilloma goes well beyond the scope of the present
case report. Moreover, the molecular detection of BPV1 in newborn healthy foals [
25
],
along with the conflicting data about BPV1 infection in the skin of healthy horses [
36
,
37
],
add further concerns about the significance of BPV1 in equine congenital papilloma. In
our opinion, the case described herein suggests that BPV vertical transmission might have
clinical relevance in different animal species, further highlighting the biological plasticity
of delta-PVs. As a matter of fact, BPV1/2 have been shown to infect several animal species,
targeting a wide range of cell types and causing different neoplastic lesions depending on
the host features (animal species, age, route of transmission).
4. Conclusions
The present case report found BPV1 infection in an equine congenital papilloma, with
very high viral load. This stimulates further investigations, even on archived samples,
aiming to clarify the etiology of equine congenital papilloma and the clinical relevance, if
any, of BPV1 vertical transmission in horses.
Pathogens 2023,12, 1059 8 of 9
Supplementary Materials:
The following supporting information can be downloaded at: https:
//www.mdpi.com/article/10.3390/pathogens12081059/s1, Figure S1: Curve of Real Time PCR to
detection of BPV1.
Author Contributions:
Conceptualization, G.M., R.M., A.G. and E.R.; methodology, G.M., R.M., F.F.,
D.D.S. and E.R.; formal analysis, G.M., L.D.P., C.G.D.C., F.F., V.G.V. and K.M.; investigation, G.M.,
R.M., L.D.P., C.G.D.C. and E.R.; resources, G.M. and E.R.; data curation, V.G.V., K.M., D.D.S., G.M.,
R.M. and E.R.; writing—original draft preparation, D.D.S., G.M., R.M. and E.R.; writing—review and
editing, V.G.V. and K.M.; visualization, G.M., A.G. and E.R.; supervision, G.M., A.G. and E.R.; project
administration, E.R.; funding acquisition, E.R. All authors have read and agreed to the published
version of the manuscript.
Funding:
This research was funded by the Italian Ministry of Health and the Liguria Region, grant
number IZS PLV 12/19 and 22L03. The APC was funded by IZS PLV 12/19 and 22L03.
Institutional Review Board Statement:
The study was approved by the Ethics Committee of Is-
tituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta (protocol code 14047 of
28 November 2019).
Informed Consent Statement:
Informed consent was obtained from the owner of the horse involved
in this study.
Data Availability Statement: All data deriving from the study are given in the article text.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Kumar, V.; Abbas, A.K.; Fausto, N. Neoplasia. In Pathologic Basis of Disease, 7th ed.; Kumar, V., Abbas, A.K., Fausto, N., Eds.;
Elsevier Saunders: Philadelphia, PA, USA, 2005; pp. 269–342.
2.
de Villiers, E.M.; Fauquet, C.; Broker, T.R.; Bernard, H.U.; zur Hausen, H. Classification of papillomaviruses. Virology
2004
,324,
17–27. [CrossRef] [PubMed]
3.
Mauldin, E.A.; Peters-Kennedy, J. Integumentary System. In Pathology of Domestic Animals, 6th ed.; Maxie, M.G., Ed.; Elsevier:
Alpharetta, GA, USA, 2015; pp. 509–736.
4.
Scott, D.W. Neoplastic diseases. In Large Animal Dermatology; Scott, D.W., Ed.; W.B. Saunders Company: Philadelphia, PA, USA,
1998; pp. 419–468.
5.
Goodrich, L.; Gerber, H.; Marti, E.; Antczak, D.F. Equine sarcoids. Vet. Clin. N. Am. Equine Pract.
1998
,14, 607–623. [CrossRef]
[PubMed]
6.
White, K.S.; Fuji, R.N.; Valentine, B.A.; Bildfell, R.J. Equine congenital papilloma: Pathological findings and results of papillo-
mavirus immunohistochemistry in five cases. Vet. Dermatol. 2004,15, 240–244. [CrossRef] [PubMed]
7.
Postey, R.C.; Appleyard, G.D.; Kidney, B.A. Evaluation of equine papillomas, aural plaques, and sarcoids for the presence of
Equine papillomavirus DNA and Papillomavirus antigen. Can. J. Vet. Res. 2007,71, 28–33. [PubMed]
8.
Christmann, U. Handling and restraint. In Equine Reproductive Procedures, 2nd ed.; Dascanio, J., McCue, P., Eds.; Wiley-Blackwell:
Hoboken, NJ, USA, 2021; pp. 681–686.
9.
Wang, F.; Flanagan, J.; Su, N.; Wang, L.C.; Bui, S.; Nielson, A.; Wu, X.; Vo, H.T.; Ma, X.J.; Luo, Y. RNAscope: A novel in situ RNA
analysis platform for formalin-fixed, paraffin-embedded tissues. J. Mol. Diagn. 2012,14, 22–29. [CrossRef] [PubMed]
10. Campo, M.S. Papillomavirus and disease in humans and animals. Vet. Comp. Oncol. 2003,1, 3–14. [CrossRef]
11.
Araldi, R.P.; Assaf, S.M.R.; Carvalho, R.F.; Carvalho, M.A.C.R.; Souza, J.M.; Magnelli, R.F.; Módolo, D.G.; Roperto, F.P.; de Cassia
Stocco, R.; Beçak, W. Papillomaviruses: A systematic review. Genet. Mol. Biol. 2017,40, 1–21. [CrossRef]
12. Okunade, K.S. Human papillomavirus and cervical cancer. J. Obstet. Gynaecol. 2020,40, 602–608. [CrossRef]
13. Tumban, E. A Current Update on Human Papillomavirus-Associated Head and Neck Cancers. Viruses 2019,11, 922. [CrossRef]
14.
Hainisch, E.K.; Jindra, C.; Kirnbauer, R.; Brandt, S. Papillomavirus-like Particles in Equine Medicine. Viruses
2023
,15, 345.
[CrossRef]
15.
Straticò, P.; Razzuoli, E.; Hattab, J.; Guerri, G.; Celani, G.; Palozzo, A.; Bonanni, D.; Fruscione, F.; Varasano, V.; Petrizzi, L.; et al.
Equine Gastric Squamous Cell Carcinoma in a Friesian Stallion. J. Equine Vet. Sci. 2022,117, 104087. [CrossRef] [PubMed]
16.
Linder, K.E.; Bizikova, P.; Luff, J.; Zhou, D.; Yuan, H.; Breuhaus, B.; Nelson, E.; Mackay, R. Generalized papillomatosis in three
horses associated with a novel equine papillomavirus (EcPV8). Vet. Dermatol. 2018,29, 72-e30. [CrossRef] [PubMed]
17.
Li, C.X.; Chang, W.S.; Mitsakos, K.; Rodger, J.; Holmes, E.C.; Hudson, B.J. Identification of a Novel Equine Papillomavirus in
Semen from a Thoroughbred Stallion with a Penile Lesion. Viruses 2019,11, 713. [CrossRef] [PubMed]
18.
Turco, S.; Gabbianelli, F.; Mavian, C.N.; Pietrucci, D.; De Paolis, L.; Gialletti, R.; Mechelli, L.; De Ciucis, C.G.; Cappelli, K.;
Dell’Anno, F.; et al. Genetic Characterization of a Novel Equus caballus Papillomavirus Isolated from a Thoroughbred Mare.
Viruses 2023,15, 650. [CrossRef]
19. Campo, M.S. Animal models of papillomavirus pathogenesis. Virus Res. 2002,89, 249–261. [CrossRef]
Pathogens 2023,12, 1059 9 of 9
20.
Savini, F.; Gallina, L.; Prosperi, A.; Puleio, R.; Lavazza, A.; Di Marco, P.; Tumino, S.; Moreno, A.; Lelli, D.; Guercio, A.; et al. Bovine
Papillomavirus 1 Gets Out of the Flock: Detection in an Ovine Wart in Sicily. Pathogens 2020,9, 429. [CrossRef]
21.
Trewby, H.; Ayele, G.; Borzacchiello, G.; Brandt, S.; Campo, M.S.; Del Fava, C.; Marais, J.; Leonardi, L.; Vanselow, B.; Biek, R.; et al.
Analysis of the long control region of bovine papillomavirus type 1 associated with sarcoids in equine hosts indicates multiple
cross-species transmission events and phylogeographical structure. J. Gen. Virol. 2014,95, 2748–2756. [CrossRef]
22.
Nasir, L.; Reid, S.W.J. Bovine papillomaviruses and equine sarcoids. In Papillomavirus Research: From Natural History to Vaccines
and Beyond; Campo, M.S., Ed.; Caister Academic Press: Norfolk, UK, 2006; pp. 389–397.
23.
Brandt, S.; Tober, R.; Corteggio, A.; Burger, S.; Sabitzer, S.; Walter, I.; Kainzbauer, C.; Steinborn, R.; Nasir, L.; Borzacchiello, G.
BPV-1 infection is not confined to the dermis but also involves the epidermis of equine sarcoids. Vet. Microbiol. 2011,150, 35–40.
[CrossRef]
24.
Hartl, B.; Hainisch, E.K.; Shafti-Keramat, S.; Kirnbauer, R.; Corteggio, A.; Borzacchiello, G.; Tober, R.; Kainzbauer, C.; Pratscher,
B.; Brandt, S. Inoculation of young horses with bovine papillomavirus type 1 virions leads to early infection of PBMCs prior to
pseudo-sarcoid formation. J. Gen. Virol. 2011,92, 2437–2445. [CrossRef]
25.
Savini, F.; Gallina, L.; Mazza, F.; Mariella, J.; Castagnetti, C.; Scagliarini, A. Molecular Detection of Bovine Papillomavirus DNA in
the Placenta and Blood of Healthy Mares and Respective Foals. Vet. Sci. 2019,6, 14. [CrossRef]
26.
Brandt, S.; Schoster, A.; Tober, R.; Kainzbauer, C.; Burgstaller, J.P.; Haralambus, R.; Steinborn, R.; Hinterhofer, C.; Stanek, C.
Consistent detection of bovine papillomavirus in lesions, intact skin and peripheral blood mononuclear cells of horses affected by
hoof canker. Equine Vet. J. 2011,43, 202–209. [CrossRef]
27.
Silva, M.A.; Silva, K.M.; Jesus, A.L.; Barros, L.O.; Corteggio, A.; Altamura, G.; Borzacchiello, G.; Freitas, A.C. The presence and
gene expression of bovine papillomavirus in the peripheral blood and semen of healthy horses. Transbound. Emerg. Dis.
2014
,61,
329–333. [CrossRef]
28.
Armbruster-Moraes, E.; Ioshimoto, L.M.; Leão, E.; Zugaib, M. Presence of human papillomavirus DNA in amniotic fluids of
pregnant women with cervical lesions. Gynecol. Oncol. 1994,54, 152–158. [CrossRef]
29.
Rombaldi, R.L.; Serafini, E.P.; Mandelli, J.; Zimmermann, E.; Losquiavo, K.P. Perinatal transmission of human papilomavirus
DNA. Virol. J. 2009,6, 83. [CrossRef]
30.
Freitas, A.C.; Mariz, F.C.; Silva, M.A.; Jesus, A.L. Human papillomavirus vertical transmission: Review of current data. Clin.
Infect. Dis. 2013,56, 1451–1456. [CrossRef] [PubMed]
31.
Roperto, S.; Borzacchiello, G.; Esposito, I.; Riccardi, M.; Urraro, C.; Lucà, R.; Corteggio, A.; Tatè, R.; Cermola, M.; Paciello, O.; et al.
Productive infection of bovine papillomavirus type 2 in the placenta of pregnant cows affected with urinary bladder tumors.
PLoS ONE 2012,7, 33569. [CrossRef] [PubMed]
32.
Roperto, S.; Russo, V.; De Falco, F.; Taulescu, M.; Roperto, F. Congenital papillomavirus infection in cattle: Evidence for
transplacental transmission. Vet. Microbiol. 2019,230, 95–100. [CrossRef] [PubMed]
33.
Roperto, S.; Russo, V.; Corrado, F.; De Falco, F.; Munday, J.S.; Roperto, F. Oral fibropapillomatosis and epidermal hyperplasia of
the lip in newborn lambs associated with bovine Deltapapillomavirus. Sci. Rep. 2018,8, 13310. [CrossRef] [PubMed]
34.
Silva, M.A.R.; Batista, M.V.A.; Pontes, N.E.; Santos, E.U.D.; Coutinho, L.C.A.; Castro, R.S.; Balbino, V.Q.; Freitas, A.C. Comparison
of two PCR strategies for the detection, of bovine papillomavirus. J. Virol. Methods 2013,192, 55–58. [CrossRef]
35.
Foresta, C.; Patassini, C.; Bertoldo, A.; Menegazzo, M.; Francavilla, F.; Barzon, L.; Ferlin, A. Mechanism of human papillomavirus
binding to human spermatozoa and fertilizing ability of infected spermatozoa. PLoS ONE 2011,6, e15036. [CrossRef]
36.
Bogaert, L.; Martens, A.; Van Poucke, M.; Ducatelle, R.; De Cock, H.; Dewulf, J.; De Baere, C.; Peelman, L.; Gasthuys, F. High
prevalence of bovine papillomaviral DNA in the normal skin of equine sarcoid-affected and healthy horses. Vet. Microbiol.
2008
,
129, 58–68. [CrossRef] [PubMed]
37.
Pratscher, B.; Hainisch, K.; Sykora, S.; Brandt, S.; Jindra, C. No evidence of bovine papillomavirus type 1 or 2 infection in healthy
equids. Equine Vet. J. 2019,51, 612–616. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
Article
Bovine Papillomaviruses (BPVs) constitute a diverse group within the Papillomaviridae family, playing a crucial role in bovine health and economic considerations. This study investigates the dynamics of vertical transmission of BPV in cattle, focusing on five cows and their reproductive tissues, as well as three gravid cows and their fetuses. DNA and RNA samples were extracted from the warts, fetal skin, placenta, uterus, ovary, and blood of cows, as well as the skin and blood of fetuses. Polymerase Chain Reaction (PCR) targeted BPV types 1–6 and 8–14, was assessed in both cows and fetuses. Additionally, Reverse Transcription PCR (RT-PCR) examined BPV-2 E5 oncogene expression in the skin and reproductive sites of mother cows and fetuses. Our findings unveil a rich diversity of BPV types, including BPV-2, 3, 9, 10, 12, and 14, present in both maternal and fetal tissues. Intriguingly, certain types, namely BPV-4, 6, 8, and 11, were exclusively identified in maternal tissues A higher diversity of BPVs was observed in cow warts, followed by cow blood, fetal blood, and fetal skin. Strikingly similar BPV types in gravid cow blood and fetuses suggest primary dissemination through the bloodstream and transmission via the placenta, though detected in lower numbers in cow uterus and ovary. Histopathological analysis revealed no abnormalities in the reproductive tissues despite the presence of BPV. However, in one bladder sample from a cow that did not consume bracken fern, urothelial neoplasia in situ was observed. The study extends beyond detection, exploring the expression of the BPV-2 E5 oncogene in fetal tissues, providing insights into potential cell implications. Comparative analyses with previous studies highlight the uniqueness of our investigation, encompassing a broader array of BPV types in the gravid cows and their fetuses. The findings not only establish a foundation for further investigations into the mechanisms of vertical transmission but also highlight the need for targeted interventions and surveillance strategies to mitigate potential health risks associated with specific BPV types.
Article
Full-text available
Papillomaviruses (PVs) are small, non-enveloped viruses, ubiquitous across the animal kingdom. PVs induce diverse forms of infection, such as cutaneous papillomas, genital papillomatosis, and carcinomas. During a survey on the fertility status of a mare, a novel Equus caballus PV (EcPV) has been identified using Next Generation Sequencing, and it was further confirmed with genomewalking PCR and Sanger sequencing. The complete circular genome 7607 bp long shares 67% average percentage of identity with EcPV9, EcPV2, EcPV1, and EcPV6, justifying a new classification as Equus caballus PV 10 (EcPV10). All EcPV genes are conserved in EcPV10, and phylogenetic analysis indicates that EcPV10 is closely related to EcPV9 and EcPV2, genus Dyoiota 1. A preliminary EcPV10 genoprevalence study, carried out on 216 horses using Real Time PCRs, suggested a low incidence of this isolate (3.7%) compared to EcPVs of the same genus such as EcPV2 and EcPV9 in the same horse population. We hypothesize a transmission mechanism different from the one observed in the closely related EcPV9 and EcPV2 that particularly infect Thoroughbreds. This horse breed is usually submitted to natural mating, thus indicating a possible sexual diffusion. No differences were detected for breeds in terms of susceptibility to EcPV10. Further studies are needed to investigate the molecular mechanisms behind the host and EcPV10 infection to explain the reduced viral spread.
Article
Full-text available
Papillomaviruses (PVs) are a family of small DNA tumor viruses that can induce benign lesions or cancer in vertebrates. The observation that animal PV capsid-proteins spontaneously self-assemble to empty, highly immunogenic virus-like particles (VLPs) has led to the establishment of vaccines that efficiently protect humans from specific PV infections and associated diseases. We provide an overview of PV-induced tumors in horses and other equids, discuss possible routes of PV transmission in equid species, and present recent developments aiming at introducing the PV VLP-based vaccine technology into equine medicine.
Article
Full-text available
A proliferative cauliflower lesion was excised from the udder of a sheep. Histological investigation confirmed the macroscopic classification of the lesion as a papilloma, without any fibroblastic proliferation. PCR revealed the presence of bovine papillomavirus (BPV), which was further confirmed by the identification of a Deltapapillomavirus 4 by Next Generation Sequencing analysis. This was subsequently classified as bovine papillomavirus type 1. Negative staining electron microscopy (EM) analyses produced negative test results for papillomavirus particles. RNA in situ hybridization (ISH) confirmed the presence of BPV-1. The results further confirm the ability of BPVs belonging to the Deltapapillomavirus genus to infect distantly related species and to cause lesions that are different from sarcoids.
Article
Full-text available
Human papillomavirus (HPV) infection is the cause of a growing percentage of head and neck cancers (HNC); primarily, a subset of oral squamous cell carcinoma, oropharyngeal squamous cell carcinoma, and laryngeal squamous cell carcinoma. The majority of HPV-associated head and neck cancers (HPV + HNC) are caused by HPV16; additionally, co-factors such as smoking and immunosuppression contribute to the progression of HPV + HNC by interfering with tumor suppressor miRNA and impairing mediators of the immune system. This review summarizes current studies on HPV + HNC, ranging from potential modes of oral transmission of HPV (sexual, self-inoculation, vertical and horizontal transmissions), discrepancy in the distribution of HPV + HNC between anatomical sites in the head and neck region, and to studies showing that HPV vaccines have the potential to protect against oral HPV infection (especially against the HPV types included in the vaccines). The review concludes with a discussion of major challenges in the field and prospects for the future: challenges in diagnosing HPV + HNC at early stages of the disease, measures to reduce discrepancy in the prevalence of HPV + HNC cases between anatomical sites, and suggestions to assess whether fomites/breast milk can transmit HPV to the oral cavity.
Article
Full-text available
Papillomaviruses (PVs) have been identified in a wide range of animal species and are associated with a variety of disease syndromes including classical papillomatosis, aural plaques, and genital papillomas. In horses, 13 PVs have been described to date, falling into six genera. Using total RNA sequencing (meta-transcriptomics) we identified a novel equine papillomavirus in semen taken from a thoroughbred stallion suffering a genital lesion, which was confirmed by nested RT-PCR. We designate this novel virus Equus caballus papillomavirus 9 (EcPV9). The complete 7656 bp genome of EcPV9 exhibited similar characteristics to those of other horse papillomaviruses. Phylogenetic analysis based on concatenated E1-E2-L2-L1 amino acid sequences revealed that EcPV9 clustered with EcPV2, EcPV4, and EcPV5, although was distinct enough to represent a new viral species within the genus Dyoiotapapillomavirus (69.35%, 59.25%, and 58.00% nucleotide similarity to EcPV2, EcPV4, and EcPV5, respectively). In sum, we demonstrate the presence of a novel equine papillomavirus for which more detailed studies of disease association are merited.
Article
Full-text available
Despite the characteristic species specificity of Papillomaviruses (PVs), the bovine papillomavirus (BPV) types 1, 2, and—more rarely—13, can cross-infect equids, where they are involved in the pathogenesis of sarcoid neoplasms. Sarcoids are locally invasive fibroblastic skin tumors that represent the most common skin neoplasms in horses worldwide. The transmission mechanism of BPV is still controversial in horses. Thus far, direct and indirect routes have been implicated, while vertical transmission has been suggested after the detection of viral DNA in the semen of healthy stallions. Testing of the blood and placenta of non-sarcoid baring mares and their respective foals revealed that the equine placenta can harbor BPV DNA, leading us to speculate a possible prenatal vertical DNA transmission in equids.
Article
Brief summary A 7-year-old Friesian stallion with a history of oesophageal stenosis, weight loss, inappetence, and recurrent hyperthermia was referred for gastroscopy. The stomach mucosa surrounding the oesophageal opening showed a large, necrotic, and ulcerated mass. On post-mortem examination, a very large, cauliflower-like neoplasm was seen, affecting non-glandular gastric mucosa. Nodular lesions were observed, scattered on the omentum, the spleen and the liver. Microscopic findings allowed the diagnosis of gastric squamous cell carcinoma with abdominal metastasis. Biomolecular investigations demonstrated the presence of EcPV-2 genes in neoplastic lesions, thus supporting the role of EcPV-2 in the ethiology of equine gastric cancer.
Chapter
Foals may need to be restrained for physical exams, treatments, diagnostic procedures, and sometimes for minor interventions. The veterinarian can use standing restraint while completing a physical exam or a handler can hold the foal while treatment is administered. Placing a foal in lateral recumbency is often used to perform abdominal ultrasound, to place a catheter, or to complete other procedures under sedation. Veterinarians can also help owners learn proper restraint and care. In addition, they can guide owners in the use of imprinting techniques which can facilitate socialization of the foal with humans, learning to halter and lead, and getting used to several routine procedures including picking up feet, tolerating touch during a physical exam, and accepting use of clippers. Foals that have undergone imprint training get used to common procedures and may become easier to handle and less fractious compared to untrained foals.
Article
Cervical cancer is by far the most common HPV-related disease. About 99.7% of cervical cancer cases are caused by persistent genital high-risk human papillomavirus (HPV) infection. Worldwide, cervical cancer is one of the most common cancers in women with an estimated 528,000 new cases reported in 2012. Most HPV infections clear spontaneously but persistent infection with the oncogenic or high-risk types may cause cancer of the oropharynx and anogenital regions. The virus usually infects the mucocutaneous epithelium and produces viral particles in matured epithelial cells and then causes a disruption in normal cell-cycle control and the promotion of uncontrolled cell division leading to the accumulation of genetic damage. There are currently two effective prophylactic vaccines against HPV infection, and these comprise of HPV types 16 and 18, and HPV types 6, 11, 16 and 18 virus-like particles. HPV testing in the secondary prevention of cervical cancer is clinically valuable in triaging low-grade cytological abnormalities and is also more sensitive than cytology as a primary screening. If these prevention strategies can be implemented in both developed and developing countries, many thousands of lives could be saved.