ArticlePDF Available

Detection of mink astrovirus in Poland and further phylogenetic comparison with other European and Canadian astroviruses

Authors:

Abstract and Figures

Mink astrovirus infection remains a poorly understood disease entity, and the aetiological agent itself causes disease with a heterogeneous course, including gastrointestinal and neurological symptoms. This paper presents cases of astrovirus infection in mink from continental Europe. RNA was isolated from the brains and intestines of animals showing symptoms typical of shaking mink syndrome (n = 6). RT-PCR was used to amplify astrovirus genetic material, and the reaction products were separated on a 1% agarose gel. The specificity of the reaction was confirmed by sequencing fragment coding RdRP protein (length of sequencing product 170 bp) from all samples. The presence of astrovirus RNA was detected in each of the samples tested. Sequencing and bioinformatic analysis indicated the presence of the same variant of the virus in all samples. Comparison of the variant with the sequences available in bioinformatics databases confirmed that the Polish isolates form a separate clade, closely related to Danish isolates. The dissimilarity of the Polish variant to those isolated in other countries ranged from 2.4% (in relation to Danish isolates) to 7.1% (in relation to Canadian isolates). Phylogenetic relationships between variants appear to be associated with the geographic distances between them. To our knowledge, this work describes the first results on the molecular epidemiology of MAstV in continental Europe. The detection of MAstV in Central Europe indicates the need for further research to broaden our understanding of the molecular epidemiology of MAstV in Europe.
This content is subject to copyright. Terms and conditions apply.
Vol:.(1234567890)
Virus Genes (2021) 57:258–265
https://doi.org/10.1007/s11262-021-01834-z
1 3
ORIGINAL PAPER
Detection ofmink astrovirus inPoland andfurther phylogenetic
comparison withother European andCanadian astroviruses
AndrzejJakubczak1 · MarekKowalczyk2· IlonaMazurkiewicz1· MarcinKondracki1
Received: 21 October 2020 / Accepted: 26 March 2021 / Published online: 15 April 2021
© The Author(s) 2021
Abstract
Mink astrovirus infection remains a poorly understood disease entity, and the aetiological agent itself causes disease with a
heterogeneous course, including gastrointestinal and neurological symptoms. This paper presents cases of astrovirus infection
in mink from continental Europe. RNA was isolated from the brains and intestines of animals showing symptoms typical
of shaking mink syndrome (n = 6). RT-PCR was used to amplify astrovirus genetic material, and the reaction products were
separated on a 1% agarose gel. The specificity of the reaction was confirmed by sequencing fragment coding RdRP protein
(length of sequencing product 170bp) from all samples. The presence of astrovirus RNA was detected in each of the sam-
ples tested. Sequencing and bioinformatic analysis indicated the presence of the same variant of the virus in all samples.
Comparison of the variant with the sequences available in bioinformatics databases confirmed that the Polish isolates form
a separate clade, closely related to Danish isolates. The dissimilarity of the Polish variant to those isolated in other coun-
tries ranged from 2.4% (in relation to Danish isolates) to 7.1% (in relation to Canadian isolates). Phylogenetic relationships
between variants appear to be associated with the geographic distances between them. To our knowledge, this work describes
the first results on the molecular epidemiology of MAstV in continental Europe. The detection of MAstV in Central Europe
indicates the need for further research to broaden our understanding of the molecular epidemiology of MAstV in Europe.
Keywords Mink astrovirus· Molecular diagnostics· Molecular polymorphism· Phylogenetics· Shaking mink syndrome
Introduction
Astroviruses are pathogens that infect a wide range of hosts
belonging to various species. Two genera are distinguished
within the family AstroviridaeMamastrovirus and Avas-
trovirus, which include astroviruses that infect mammals and
birds, respectively. The virus has been detected in represent-
atives of mammals inhabiting both terrestrial environments
(e.g. pigs and cattle) and aquatic environments (dolphins), as
well as in birds and fish [1, 2]. A pathogen from this group
was diagnosed for the first time in children with diarrhea in
the mid-1970s. Today, alongside rotaviruses, it is one of the
main causes of viral gastrointestinal infections [3]. Astro-
viruses cause gastrointestinal diseases in humans (HastVs
1–8), sheep (OAstV), cattle (BoAstV), mink (MiAstV), pigs
(PoAstV), cats (FeAstV), dogs (CaAstV) and marine mam-
mals. In turkeys (TAstVs) and chickens (CAstV), they cause
nephritis and gastrointestinal diseases [3, 4]. Some strains of
astroviruses in humans and animals, such as mink, cattle and
sheep, can bypass the gastrointestinal tract, showing tropism
for nervous tissue. They then cause infections of organs of
the central nervous system (CNS), especially the brain [4].
Mink astrovirus (MiAstV) is known to play a major role in
mink pre-weaning diarrhea, and some of viruses detected
in mink faeces such as rotavirus C and hepatitis E virus
(HEV) are considered as zoonotic agents. These viruses are
not monitored in commercial mink, and the role of these
viral infections in mink health is not well understood [5].
The genetic material of astroviruses is single-stranded
RNA with positive polarization. The 6.8 to 7.9kb genome
contains a 5UTR untranslated region followed by three
Edited by Zhen F. Fu.
* Andrzej Jakubczak
andrzej.jakubczak@up.lublin.pl
1 Institute ofBiological Basis ofAnimal Production, Faculty
ofAnimal Sciences andBioeconomy, University ofLife
Sciences inLublin, Lublin, Poland
2 Institute ofQuality Assessment andProcessing ofAnimal
Products, Faculty ofAnimal Sciences andBioeconomy,
University ofLife Sciences inLublin, Lublin, Poland
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
259Virus Genes (2021) 57:258–265
1 3
open reading frames—ORF1a, ORF1b and ORF2, a 3UTR
region, and a poly(A)tail [3, 4, 6]. ORF1a and ORF1b code
for non-structural protein precursors, while ORF2 codes for
a structural protein precursor [4, 7]. ORF1 encodes protease
and RNA-dependent polymerase [3]. The subgenomic RNA
of the astrovirus, derived from ORF2, encodes a single, large
structural capsid protein (CP). Depending on the strain of the
virus, this capsid polyprotein precursor contains from about
775 to 785 amino acid residues, and also has a molecular
weight of 87–90 kilodaltons (kDa) [4]. The CP is an external
structural barrier that not only surrounds the nucleic acids,
but also interacts with the host, influencing cell tropism and
mediating entry into the cell. Furthermore, it is an antigen
that induces an immune response in the host [8].
Phylogenetic and genomic analyses indicate high homol-
ogy between astroviruses infecting humans and those iso-
lated from mink. There is evidence indicating the zoonotic
potential of astroviruses. After observing the occurrence of
diseases in people living near infected farms, Quan etal.
suggested that the pathogen may flow between mink and
humans [9]. The zoonotic potential of astroviruses is also
suggested by the results of Meliopoulos etal. [10], who con-
firmed the presence of antibodies against turkey astrovirus
in humans.
The aetiological factor for astrovirus infections in mink
is the MAstV-1 virus. Mink astrovirus infection is a disease
with a heterogeneous course and a diverse clinical picture.
When the pathogen colonizes the nervous system, shaking
mink syndrome (SMS) develops [11, 12]. Other clinical pic-
tures of the disease, often treated as one disease entity due
to the similarity of the symptoms, are pre-weaning diarrhea
[13] and wet mink syndrome (WMS).
In the case of WMS, viraemia results in increased activ-
ity of the apocrine glands, especially in the neck and tail
area, where a sticky, greasy secretion appears, to which the
disease owes its name. Affected mink kits display diarrhea
with concomitant excessive secretions from the cervical
apocrine glands, and exudate on the skin surface, the tail,
and the claws. Moreover, dehydration may ultimately lead
to the death of the affected kits [14]. The secretions may
cause deterioration in the quality of the fur, which takes
on a wavy structure. As the disease develops, alopecia may
occur at the site of excessive secretion of apocrine glands
[4]. The syndrome includes a characteristic symptom of
astrovirus infections, i.e. diarrhea, lasting up to 10days, usu-
ally foamy and yellowish, and often with an admixture of
undigested milk [13]. Animal faeces are infectious material
through which the virus can spread. In many cases, diarrhea
and fever result in dehydration and an overall decrease in
immunity, which is conducive to complications caused by
bacterial co-infections. The clinical picture may also include
behavioural changes in mink; sick individuals often make
sounds that resemble meowing. Pre-weaning diarrhea is a
syndrome affecting farm-raised neonatal mink kits. Apart
from diarrhea it causes greasy skin exudation, dehydration,
and distressed behavior. Moreover, dehydration may ulti-
mately lead to the death of the affected kits [15].
In view of the relatively little-known aetiology and epide-
miology of mink astrovirus infection, as well as the lack of
research on this subject in continental Europe, the aim of the
study is to examine molecular variation in MAstV on farms
in Poland and the relationship between the variants obtained
and previously isolated variants.
Materials andmethods
The study covered two farms in north-western Poland with
more than 10,000 mink. Symptoms typical of the neurologi-
cal form of astrovirus infection—shaking mink syndrome—
were observed in animals on these farms, such as tremors,
an unsteady gait, and awkward movements. Cases of wet
mink syndrome were also noted on the farms. The study
material comprised brains (n = 6) and intestines (n = 6) of
six mink that had died from the disease (3 mink from each
of the tested farm). The age of the tested animals ranged
from 4 to 7weeks. Tissues were collected and fixed in RNA
later reagent.
Viral RNA isolation
Isolation was carried out in two independent replicates using
the RNeasy Mini Kit (Qiagen) and the Total RNA Mini kit
(A&A Biotechnology). The tissues were suspended in lysis
buffers: 600μl RLT buffer was used in the case of the RNe-
asy Mini Kit, while the tissues were suspended in 800μl
fenozol for isolation with the Total RNA Mini Kit. Then the
samples were homogenized in a Tissue Lyser for 40s at a
frequency of 20Hz. The remainder of the isolation proce-
dure was carried out according to the recommendations of
the kit manufacturers. Genetic material was eluted in 50μl
of nuclease-free water.
Reverse transcription andPCR
Reverse transcription was performed using the QuantiTect
Reverse Transcription Kit (Qiagen). The gDNA Wipeout
Buffer reagent was used to remove DNA residue. Reverse
transcription was carried out according to the manufacturer’s
protocol and included a reaction with oligo-dT primers and
random hexameters to obtain total cDNA, with a recom-
mended incubation time of 42°C for 15min and 3min of
enzyme inactivation at 95°C.
PCR was carried out for all samples isolated from the
intestines and brains using both of the kits and transcribed
into cDNA (n = 24: 6 intestinal samples and 6 brain samples
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
260 Virus Genes (2021) 57:258–265
1 3
for the Qiagen kit and 6 intestinal samples and 6 brain sam-
ples for the A&A Biotechnology kit). The primers used were
MA17 (reverse, 5 GAG GAG TTT CAG ACA GAT G 3) and
MA15 (forward 5 CAA ATG CCT GGA AGA ACA C 3),
proposed by Mittelholzer etal. [7]. Primers targeted a part
of RNA-dependent RNA-polymerase region (RdRp). The
reaction mixture contained 3μl DNA and 1 U Taq poly-
merase (AmpliTaq Gold 360 DNA Polymerase, Applied
Biosystems) in the manufacturer’s buffer, adjusted to a final
concentration of 2.5mM MgCl2; 0.8mM of each dNTP; and
1.2mM of each primer—25 μL total volume. The reaction
took place under the following conditions: 95°C for 10min,
40 cycles of 95°C for 45s, 54°C for 45s, 72°C for 45s,
and 72°C for 10min in a Labcycler Thermocycler (Senso-
Quest). The reaction products were separated on a 1% aga-
rose gel with ethidium bromide at 80V. Visualization and
archiving of the gel was carried out in Scion Image software.
Sequencing andbioinformatics analysis
The samples were purified with an EPPiC Fast kit (A&A
Biotechnology) and subjected to sequencing with the same
primers as in the standard reaction, using the BigDye Ter-
minator v3.1 Cycle Sequencing Kit (Applied Biosystems).
Sequencing PCR products were purified using the Extermi-
nator kit (A&A Biotechnology). The purified samples were
suspended in formamide, denatured, and then separated
on an ABI PRISM 3100 Avant Genetic Analyzer (Applied
Biosystems).
Sequencing results were assembled into contigs in DNA
Baser software to obtain fragments of 170bp. Specificity
was confirmed using the Blast application, and sequences
were compared with the NCBI bioinformatics database.
Sequences obtained during the analyses were compared
with sequences from the GenBank database in MEGA7 soft-
ware. The similarity between isolates was determined using
BioEdit software. Analysis of polymorphisms and phyloge-
netic analysis were carried out in MEGA7. The evolutionary
history was inferred using the neighbor-joining (NJ) method.
The percentage of replicate trees in which the associated
taxa are clustered together in the bootstrap test (1000 repli-
cates) are shown next to the branches [16].
Results
The RT-PCR method confirmed the presence of the genetic
material of the virus in both brain and intestinal samples
isolated using both kits. In each of the tested samples the
product of 178bp was obtained. The specificity of the reac-
tion was confirmed by sequencing all PCR products. After
sequencing the fragment of 170bp was obtained and used
in further bioinformatic analysis. The results show 100%
similarity for the tested fragment between isolates from the
two farms.
The similarity of the nucleotide sequence of RdRP gene
of the mink astrovirus variants from Polish farms with Dan-
ish, Swedish and Canadian variants deposited in the NCBI
database was assessed as well (Table1). The Polish isolates
showed the highest similarity to the variants isolated from
Danish farms, ranging from 96.4 to 97.6% (97.2% on aver-
age). Lower similarity was noted in comparison to Swedish
isolates, ranging from 94.1 to 96.4% (95.64% on average).
The greatest variation in relation to Polish isolates was found
for the MAstV variants from Canada, differing in 12 nucleo-
tides, which translated into a more than 7% difference within
the analysed fragment. Bioinformatics analysis indicated a
unique G3674A polymorphism in the Polish isolates, which
has thus far not been detected in other sequences deposited
in databases.
All polymorphisms between Danish and Polish isolates
are transitions, while in the case of Swedish farms there
were two transversions—T3514A and T3616A, both in the
case of an isolate deposited under number GU985458.1,
isolated from an individual with SMS (shaking mink syn-
drome). There was also a transversion for one isolate from
Canada—C3514A.
Among the polymorphisms examined, two were non-syn-
onymous and were associated with differences in the amino
acid sequence. In the Polish isolates, adenine was present at
position 3515, as in the case of most isolates from databases.
A difference in this position occurs only in the sequence
from Danish isolate deposed in NCBI database in 2003
under number AY196095.1, with guanine at position 3515.
The polymorphism involves a change at amino acid position
301—methionine (present in most of the tested sequences)
to valine (appearing only in sequence AY196095.1). The
other amino acid change (L317P) occurs in the sequence
deposited under number AY196100.1, where thymine is
found in nucleotide position 3564 (the codon encodes leu-
cine), and cytosine in the remaining sequences (the codon
encodes proline).
Phylogenetic analysis was performed to determine the
phylogenetic relationships between Polish isolates and
isolates from Denmark, Sweden and Canada (Fig.1). Five
main groups were distinguished, three of which form one
relatively closely related clade. The first group (the Danish
group) was formed by Danish isolates, whose percentage
similarity within the group is over 99% (Table2).
The bootstrap value obtained for the node is 88. The same
group includes clades grouping together the Polish isolates
and some of the Swedish isolates. Polish isolates, like the
Danish ones, were characterized by high homogeneity,
which was manifested by the presence of the same genetic
variant in all samples. Isolates from Sweden were signifi-
cantly less homogeneous. Based on phylogenetic analysis,
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
261Virus Genes (2021) 57:258–265
1 3
two heterogeneous groups were distinguished—Swed-
ish I—which included two closely related clades (98.2%
similarity) and Swedish II, which included, among others,
the GU985458.1 isolate, causing shaking mink syndrome
(SMS). This variant differed from representatives of the
Swedish I group by over 6% (differences from 6.5 to 7.1%).
The second isolate within the Swedish II group was depos-
ited under number AY196104.1. In relation to the Swedish
Table 1 Polymorphic nucleotides differentiating Polish isolates from variants from the NCBI database
Differences between database isolates and isolates from Polish farms are given (First letter—nucleotide or amino acid position in the database
variant, number—polymorphism location, second letter—nucleotide or amino acid in the Polish isolate. Bolded polymorphisms—nonsynony-
mous substytutuions
Country Accession number Polish iso-
late [%]
Polymorphic nucleotides
Denmark AY196095.1 96.4 G3515A > V301M, G3574A*, T3598C, G3604A, T3628C, C3631T
AY196096.1 97.0 G3574A*, T3598C, G3616A, T3628C, C3631T
AY196097.1 97.6 G3574A*, T3598C, T3628C, C3631T
AY196098.1 97.6 G3574A*, T3598C, T3628C, C3631T
AY196099.1 97.6 G3574A*, T3598C, T3628C, C3631T
AY196100.1 97.0 T3564C > L317P, G3574A*, T3598C, T3628C, C3631T
Sweden AY196101.1 95.8 C3508T, T3529C, C3547T, A3550G, G3574A*, T3598C, C3631T
AY196102.1 96.4 C3508T, C3547T, A3550G, G3574A*, T3583C, C3631T
AY196103.1 95.8 C3508T, T3529C, C3547T, A3550G, G3574A*, T3598C, C3631T
AY196104.1 95.2 T3514A, A3523G, T3529C, C3547T, G3559A, G3574A*, T3577C, T3583C
AY196105.1 95.8 C3508T, T3529C, C3547T, A3550G, G3574A*, T3598C, C3631T
NC_004579.1 96.4 C3508T, C3547T, A3550G, G3574A*, T3583C, C3631T
GU985458.1 94.1 T3514A, T3529C, C3547T, G3559A, G3574A*, T3577C, G3604A, C3613T, T3616A, T3628C
Canada MH282878.1 92.9 C3514A, C3520T, C3532T, C3547T, G3559A, G3574A*, T3583C, A3586G, T3589C, T3595C,
T3598C, C3607T
MH282880.1 92.9 T3514A, C3520T, C3532T, C3547T, G3559A, G3574A*, T3583C, A3586G, T3589C, T3595C,
T3598C, C3607T
Fig. 1 Phylogenetic analysis of the Polish MAstv isolate in relation to sequences deposited in the NCBI database. The tree was constructed using
the NJ method with a bootstrap value of 1000. Analysis based on 170bp fragment coding RdRp (RNA-dependent RNA polymerase) gene
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
262 Virus Genes (2021) 57:258–265
1 3
I group it showed 94.7–95.2% similarity. The similarity
between the two sequences assigned to the Swedish II group
was 96.4%.
A separate group was a homogeneous clade consisting of
Canadian isolates (99.4% similarity within the group), which
showed significant differences in relation to variants of the
virus isolated in Europe. Compared to representatives of the
Danish group, the differences ranged from 7.1 to 8.3%. A
similar level of similarity was found in relation to most of
the Swedish isolates (average differences of 7–8%), except
for isolate AY196104.1, which showed over 94% similar-
ity to one of the Canadian isolates. Expanded phylogenetic
analysis, including astroviruses infecting representatives of
other species, confirmed that the mink astrovirus is the most
closely related to astroviruses infecting humans (Fig.2).
Discussion
Mink astrovirus infection remains a poorly characterized
disease entity. In 2002, Englund etal. investigated the rela-
tionship between the presence of astrovirus in the mink
intestines and faeces and the occurrence of pre-weaning
diarrhea in mink. The researchers confirmed the presence of
the pathogen in both types of material and indicated a pos-
sible causal relationship between the astrovirus and the onset
of disease [13]. The results obtained by Englund etal. were
based on histopathological examination and observations of
viral particles under an electron microscope. Therefore, it is
difficult to state conclusively whether the pathogens present
in the set of samples examined by that team included MAstV
or another member of the Astroviridae family.
In our own research, the presence of MAstV genetic
material was confirmed in both the brain and the intestines.
However, in contrast to Englund’s study [13], we observed a
different set of symptoms, much more similar to the shaking
mink syndrome described by Blomström etal. 2010 [12].
An increasing body of research indicates that astroviruses
may be responsible for the development of disease entities
with a diverse clinical picture. Most reports confirm gastro-
intestinal symptoms resulting from replication of the virus
in the intestines [17, 18], or in the case of avian astroviruses
in the liver as well [19, 20]. An increasing number of stud-
ies confirm the link between astroviruses and neurological
symptoms detected in pigs [21], cattle [22], or mink [12]. In
our research as well, the symptoms pointed to the neurologi-
cal form of the disease, and the presence of MAstV genetic
material was also confirmed in the brain of the animals.
Table 2 Comparison of the
RdRp (RNA-dependent RNA
polymerase) gene sequence
between variants available in
the NCBI database (similarity
was expressed as %)
Sweden Denmar
kC
anada
Country
Accession
number
Poland
AY196101.1
AY196102.1
AY196103.1
AY196104.1
AY196105.1
NC_004579.1
GU985458.1
AY196095.1
AY196096.1
AY196097.1
AY196098.1
AY196099.1
AY196100.1
MH282878.1
MH282880.1
Sweden
AY196101.1 95.8ID 98.2100 94.7100 98.2 93.5 95.8 96.4 97.0 97.0 97.0 96.4 92.3 92.3
AY196102.196.498.2 ID 98.295.298.2 100 92.9 95.2 95.8 96.4 96.4 96.4 95.8 92.9 92.9
AY196103.195.810098.2 ID 94.7100 98.2 93.5 95.8 96.4 97.0 97.0 97.0 96.4 92.3 92.3
AY196104.1 95.294.7 95.294.7 ID 94.7 95.2 96.4 92.9 93.5 94.1 94.1 94.1 93.5 93.5 94.1
AY196105.1 95.810098.2100 94.7 ID 98.2 93.5 95.8 96.4 97.0 97.0 97.0 96.4 92.3 92.3
NC_004579.196.498.2 100 98.295.298.2 ID 92.9 95.2 95.8 96.4 96.4 96.4 95.8 92.9 92.9
GU985458.194.193.5 92.993.596.493.5 92.9 ID 94.1 94.1 94.1 94.1 94.1 93.5 91.1 91.7
Denmark
AY196095.1 96.495.8 95.295.892.995.8 95.2 94.1 ID 98.2 98.8 98.8 98.8 98.2 91.7 91.7
AY196096.197.096.4 95.896.493.596.4 95.8 94.1 98.2 ID 99.4 99.4 99.4 98.8 92.3 92.3
AY196097.197.697.0 96.497.094.197.0 96.4 94.1 98.8 99.4 ID 100100 99.4 92.9 92.9
AY196098.197.697.0 96.497.094.197.0 96.4 94.1 98.8 99.4 100 ID 100 99.4 92.9 92.9
AY196099.1 97.697.0 96.497.094.197.0 96.4 94.1 98.8 99.4 100100 ID 99.4 92.9 92.9
AY196100.197.096.4 95.896.493.596.4 95.8 93.5 98.2 98.8 99.4 99.4 99.4 ID 92.3 92.3
Canada
MH282878.1 92.992.3 92.992.393.592.392.991.191.792.392.992.992.992.3 ID 99.4
MH282880.1 92.992.3 92.992.394.192.392.991.091.792.392.992.992.992.399.4 ID
Green colour—similarity within Swedish isolates, Blue colour—similarity within Danish isolates, Yellow
colour—similarity within Canadian isolates
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
263Virus Genes (2021) 57:258–265
1 3
Fig. 2 Analysis of phylogenetic relationships between MAstV and
astroviruses infecting humans, bats, pigs, rodents, dolphins, cattle,
dogs, cats, and sea lions. The analysis was carried out using the NJ
method, with a bootstrap value of 1000. Analysis based on 170bp
fragment coding RdRp (RNA-dependent RNA polymerase) gene
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
264 Virus Genes (2021) 57:258–265
1 3
The results confirm the diagnostic effectiveness of the
primers proposed by Mittelholzer etal. [7]. In addition,
the presence of genetic material of the astrovirus was con-
firmed for the first time in continental Europe. Analysis of
the sequenced fragment indicates the presence of the same
variant of the virus in all samples tested, obtained from two
separate farms. Similarly, Mittelholzer etal. [7] reported
high similarity in a study conducted on Danish and Swedish
farms, which showed 96.7–100% similarity of the isolates.
The researchers observed high similarity of the virus isolates
within each of the countries studied, but at the same time
variation between them. Analysis of polymorphisms by Mit-
telholzer enabled clear differentiation between Danish and
Swedish strains [7].
The high similarity observed by the Mittelholzer team
and in our research may be due to both the relatively short
length of the analysed sequence and the conserved nature of
the fragment coding RdRp, which can be used for diagnos-
tic purposes. However, molecular characterization remains
a very important element in the study of diseases caused
by astroviruses, including understanding of their underly-
ing cause and the mechanisms of their onset and develop-
ment. The usefulness of this type of analysis is confirmed by
research conducted on mink with diarrhea symptoms from
Chinese farms. Sun etal. confirmed the presence of genetic
material of astroviruses, but interestingly, none of them was
a representative of MAstV, and additional bioinformatic
analysis indicated the possibility of mink infection with
astrovirus from birds [23]. The researchers formulated an
interesting hypothesis regarding transmission of the patho-
gen via feed obtained from infected birds and the possibil-
ity of interspecies infection. The possibility of interspecies
transmission is also indicated by Quan [9], who detected
encephalitis in a boy with X-linked agammaglobulinaemia.
The authors suggest that one of the potential causes was
infection from mink from a nearby farm, but due to the com-
plicated history of the disease, as well as immunosuppres-
sion in the patient, conclusive determination of the source
of infection is not possible.
The phylogenetic analysis carried out in our study points
to a relationship between the genetic variant of the virus and
the country where the samples were isolated, which also
confirms the observations of Mitlleholzer [7] for Swedish
and Danish isolates. The differences detected may indicate
multiple and independent MAstV infections originating in
different primary outbreaks.
In addition to the study by Mitlleholzer etal. [7] men-
tioned above, MAstV phylogenetics has been studied by
Blomström [12], who obtained a tree similar to the one in
the present study. Isolate GU985458.1 presented by the
researchers formed a separate clade in relation to most
of the isolates associated with the occurrence of pre-
weaning diarrhea, which could indicate the presence of
polymorphisms influencing the tropism of the pathogen
for nerve tissue. However, the results of the present study
group the Polish isolate obtained from individuals with
the neurological form of the disease together with iso-
lates associated with pre-weaning diarrhea. Therefore,
the ORF1b region containing the RdRp gene seems not
to affect tropism or the clinical picture of the infection,
especially as the polymorphisms between GU985458.1
and most sequences are synonymous.
Bearing in mind the potential for cross-species infec-
tions caused by astroviruses, as well as their widespread
dissemination, it seems reasonable to analyse the RdRp
fragment to confirm infection and as a fragment ena-
bling preliminary assessment of the diversity of isolates
obtained in relation to the global pool of the virus.
Conclusions
Molecular diagnostics and epidemiology are increasingly
used as a tool to understand the spread and evolution of
infectious agents. The subject of mink astrovirus infec-
tion remains poorly understood, as confirmed by both the
small number of sequences deposited in bioinformatics
databases and the small number of studies in the PubMed
database.
The study confirmed the presence of MAstV astrovi-
rus genetic material in the mink brain and intestines with
a clinical picture indicative of shaking mink syndrome.
To our knowledge, this work describes the first results of
molecular epidemiology of MAstV in continental Europe.
Previous analyses have concerned Scandinavia (Sweden and
Denmark), Canada, and China. The detection of MAstV in
Central Europe indicates the need for further research that
will not only enable a better understanding of the aetiology
of such disease entities as pre-weaning diarrhea and shak-
ing mink syndrome, but also broaden current knowledge of
MAstV molecular epidemiology in Europe.
Author contributions AJ and MK conceived and planned the experi-
ments, methodology, AJ, MK—laboratory and bioinformatic analy-
sis, AJ, MK and IM—preparing of manuscript, AJ, MK, IM and M
Kon. All authors have read and agreed to the published version of the
manuscript.
Declarations
Conflict of interest The authors declare that there is no conflict of in-
terest.
Ethical approval Approval for these tests was obtained from the 2nd
Local Ethics Committee for Experiments on Animals at the University
of Life Sciences in Lublin (approval no. 83/2009).
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
265Virus Genes (2021) 57:258–265
1 3
Open Access This article is licensed under a Creative Commons Attri-
bution 4.0 International License, which permits use, sharing, adapta-
tion, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in
the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a
copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
References
1. Geoghegan JL, Di Giallonardo F, Cousins K, Shi M, Williamson
JE, Holmes EC (2018) Hidden diversity and evolution of viruses
in market fish. Virus Evol. https:// doi. org/ 10. 1093/ ve/ vey031
2. Shi M, Lin XD, Chen X, Tian JH, Chen LJ, Li K etal (2018) The
evolutionary history of vertebrate RNA viruses. Nature 556:197.
https:// doi. org/ 10. 1038/ s41586- 018- 0012-7
3. De Benedictis P, Schultz-Cherry S, Burnham A, Cattoli G (2011)
Astrovirus infections in humans and animals—molecular biology,
genetic diversity and interspecies transmissions. Infect Genet Evol
11:1529–1544. https:// doi. org/ 10. 1016/j. meegid. 2011. 07. 024
4. Bidokhti MRM, Ullman K, Hammer AS, Jensen TH, Chriel M,
Byrareddy SN etal (2019) Immunogenicity and efficacy evalua-
tion of subunit astrovirus vaccines. Vaccines. https:// doi. org/ 10.
3390/ vacci nes70 30079
5. Xie XT, Macdonald RE, Tapscott B, Nagy E, Turner PV (2018)
Detection of astrovirus. Rotavirus C. and hepatitis E viral RNA
in adult and juvenile farmed mink (Neovison vison). Front Vet Sci
5:132. https:// doi. org/ 10. 3389/ fvets. 2018. 00132
6. Schlottau K, Schulze C, Bilk S, Hanke D, Hoper D, Beer M
etal (2016) Detection of a novel bovine astrovirus in a cow with
encephalitis. Transbound Emerg Dis 63:253–259. https:// doi. org/
10. 1111/ tbed. 12493
7. Mittelholzer C, Englund L, Hedlund KO, Dietz HH, Svensson L
(2003) Detection and sequence analysis of Danish and Swedish
strains of mink astrovirus. J Clin Microbiol 41:5192–5194. https://
doi. org/ 10. 1128/ JCM. 41. 11. 5192- 5194. 2003
8. Mendez E, Aguirre-Crespo G, Zavala G, Arias CF (2007) Asso-
ciation of the astrovirus structural protein VP90 with membranes
plays a role in virus morphogenesis. J Virol 81:10649–10658.
https:// doi. org/ 10. 1128/ jvi. 00785- 07
9. Quan PL, Wagner TA, Briese T, Torgerson TR, Hornig M, Tash-
mukhamedova A etal (2010) Astrovirus encephalitis in boy with
X-linked agammaglobulinemia. Emerg Infect Dis 16:918–925.
https:// doi. org/ 10. 3201/ eid16 06. 091536
10. Meliopoulos VA, Kayali G, Burnham A, Oshansky CM, Thomas
PG, Gray GC etal (2014) Detection of antibodies against Turkey
astrovirus in humans. PLoS ONE. https:// doi. org/ 10. 1371/ journ
al. pone. 00969 34
11. Gavier-Widen D, Brojer C, Dietz HH, Englund L, Hammer
AS, Hedlund KO etal (2004) Investigations into shaking mink
syndrome: an encephalomyelitis of unknown cause in farmed
mink (Mustela vison) kits in Scandinavia. J Vet Diagn Investig
16:305–312
12. Blomstrom AL, Widen F, Hammer AS, Belak S, Berg M (2010)
Detection of a novel astrovirus in brain tissue of mink suffering
from shaking mink syndrome by use of viral metagenomics. J Clin
Microbiol 48:4392–4396. https:// doi. org/ 10. 1128/ jcm. 01040- 10
13. Englund L, Chriel M, Dietz HH, Hedlund KO (2002) Astrovi-
rus epidemiologically linked to pre-weaning diarrhea in mink.
Vet Microbiol 85:1–11. https:// doi. org/ 10. 1016/ s0378- 1135(01)
00472-2
14. Birch JM, Agger JF, Dahlin C, Jensen VF, Hammer AS, Struve T
etal (2017) Risk factors associated with diarrhea in Danish com-
mercial mink (Neovison vison) during the pre-weaning period.
Acta Vet Scand 59:43. https:// doi. or g/ 10. 1186/ s13028- 017- 0312-1
15. Mathiesen R, Birch JM, Chriél M, Jensen HE, Agger JF, Heegaard
PMH etal (2018) Mink (Neovison vison) kits with pre-weaning
diarrhea have elevated serum amyloid A levels and intestinal
pathomorphological similarities with New Neonatal Porcine Diar-
rhea Syndrome. Acta Vet Scand 60:48. https:// doi. org/ 10. 1186/
s13028- 018- 0403-7
16. Kumar S, Stecher G, Tamura K (2016) MEGA7: molecular evolu-
tionary genetics analysis version 7.0 for bigger datasets. Mol Biol
Evol 33:1870–1874. https:// doi. org/ 10. 1093/ molbev/ msw054
17. Kumthip K, Khamrin P, Saikruang W, Kongkaew A, Vachi-
rachewin R, Ushijima H, etal (2018) Detection and genetic char-
acterization of porcine astroviruses in piglets with and without
diarrhea in Thailand. Arch Virol 163:1823–1829. https:// doi. org/
10. 1007/ s00705- 018- 3806-x
18. Martella V, Catella C, Capozza P, Diakoudi G, Camero M, Lanave
G etal (2020) Identification of astroviruses in bovine and buffalo
calves with enteritis. Res Vet Sci 131:59–68. https:// doi. org/ 10.
1016/j. rvsc. 2020. 04. 010
19. Wu W, Xu R, Lv Y, Bao E (2020) Goose astrovirus infection
affects uric acid production and excretion in goslings. Poult Sci
99:1967–1974. https:// doi. org/ 10. 1016/j. psj. 2019. 11. 064
20. Long KE, Ouckama RM, WeiszA BML, Ojkic D (2018) White
chick syndrome associated with chicken astrovirus in Ontario,
Canada. Avian Dis 62:247–258. https:// doi. org/ 10. 1637/
11802- 012018- Case.1
21. Arruda B, Arruda P, Hensch M, Chen Q, Zheng Y, Yang C etal
(2017) Porcine astrovirus type 3 in central nervous system of
swine with polioencephalomyelitis. Emerg Infect Dis 23:2097–
2100. https:// doi. org/ 10. 3201/ eid23 12. 170703
22. Deiss R, Selimovic-Hamza S, Seuberlich T, Meylan M (2017)
Neurologic clinical signs in cattle with astrovirus-associated
encephalitis. J Vet Intern Med 31:1209–1214. https:// doi. org/ 10.
1111/ jvim. 14728
23. Sun N, Yang Y, Wang GS, Shao XQ, Zhang SQ, Wang FX etal
(2014) Detection and characterization of avastrovirus associated
with diarrhea isolated from minks in China. Food Environ Virol
6:169–174. https:// doi. org/ 10. 1007/ s12560- 014- 9155-3
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Phylogenetic studies of ORF2-encoded capsid protein sequences showed that these divergent MLB and VA/HMO clades are more closely related to animal astroviruses than to classical HAstVs. For example, HAstV-VAs are most closely related to ovine and mink astroviruses, resulting in the naming of the Human Mink Ovine (HMO) clade [13][14][15][16][17][18][19][20]. Notably, many of these related animal astroviruses are found to be associated with neurological disease in ovine, minks, bovine, porcine, alpacas, and muskox [13,20,21], and HAstV-VAs are increasingly being associated with neurological disease. ...
... In differential diagnosis of central nervous system infections, including encephalitis and meningitis, human astroviruses from the VA/HMO clade are increasingly associated with infections, particularly in immunocompromised patients, with high rates of mortality. Neurological diseases in domesticated mammals, such as in mink, ovine, bovine and porcine, are also associated with infection by VA/HMO astroviruses [13,14,17,20,[64][65][66]. To understand how After an initial baseline step, histidine-tagged HAstV-VA1 gastro spike protein (blue) or HAstV-VA1 neuro spike protein (red) or no protein (cyan) were loaded onto Anti-Penta-His biosensors, followed by another baseline step. ...
Article
Full-text available
Human astrovirus (HAstV) is a known cause of viral gastroenteritis in children worldwide, but HAstV can cause also severe and systemic infections in immunocompromised patients. There are three clades of HAstV: classical, MLB, and VA/HMO. While all three clades are found in gastrointestinal samples, HAstV-VA/HMO is the main clade associated with meningitis and encephalitis in immunocompromised patients. To understand how the HAstV-VA/HMO can infect the central nervous system, we investigated its sequence-divergent capsid spike, which functions in cell attachment and may influence viral tropism. Here we report the high-resolution crystal structures of the HAstV-VA1 capsid spike from strains isolated from patients with gastrointestinal and neuronal disease. The HAstV-VA1 spike forms a dimer and shares a core beta-barrel structure with other astrovirus capsid spikes but is otherwise strikingly different, suggesting that HAstV-VA1 may utilize a different cell receptor, and an infection competition assay supports this hypothesis. Furthermore, by mapping the capsid protease cleavage site onto the structure, the maturation and assembly of the HAstV-VA1 capsid is revealed. Finally, comparison of gastrointestinal and neuronal HAstV-VA1 sequences, structures, and antigenicity suggests that neuronal HAstV-VA1 strains may have acquired immune escape mutations. Overall, our studies on the HAstV-VA1 capsid spike lay a foundation to further investigate the biology of HAstV-VA/HMO and to develop vaccines and therapeutics targeting it.
... Simultaneously, this is the first report on astrovirus prevalence in foxes and small wild animals like racoons, martens, and rats in Poland. Studies on astrovirus screening in animals in Poland were previously performed only in bats found dead and collected during passive rabies surveillance and in farmed animals like poultry and mink [16][17][18]. ...
... Previous studies on AstVs in farmed and companion animals have demonstrated that turkeys, ducks, chickens, guinea fowl, pigeons, geese, mink, sheep, cattle, pigs, and cats are hosts of astroviruses [3,16,17]. In the population of wild animals, AstVs were mostly detected in bat samples, with several studies performed in wild boars in Slovakia and Hungary [29,30]. ...
Article
Full-text available
Citation: Kuczera, K.; Orłowska, A.; Smreczak, M.; Frant, M.; Trębas, P.; Rola, J. Prevalence of Astroviruses in Different Animal Species in Poland. Viruses 2024, 16, 80. https://
... All brain and intestinal samples were positive. The obtained amplicon sequences were 100% identical to each other and >95% identical to MiAstV (MAstV 10) and SMS-AstV [34], suggesting that in the case of SMS, the virus is not exclusively targeting the nervous tissue but also the GIT. Unfortunately, a pathological examination of these kits was not reported, so whether the neurological signs correlated with encephalitis or lesions in other organs remained undetermined. ...
... There is evidence for NT-AstV being transmitted fecal-orally, as is known from enteric astroviruses. NT-AstV of bovines, pigs, ovines and minks were detected in intestinal/fecal samples of diarrhetic or healthy animals [34,37,40,77]. Possibly, NT-AstV invade the CNS from the GIT retrograde along the peripheral nerves or the circulatory system, since there are cases in which NT-AstV infection induced dorsal root ganglionitis [9,48,53] or could be detected by ISH in the myenteric plexus and mesenteric lymph nodes [40]. ...
Article
Full-text available
Astrovirus infections are among the main causes of diarrhea in children, but their significance for animal health has remained underestimated and largely unknown. This is changing due to the increasing amount of newly identified neurotropic astroviruses in cases of nonsuppurative encephalitis and neurological disease in humans, pigs, ruminant species and minks. Neurological cases in ruminants and humans usually occur sporadically and as isolated cases. This contrasts with the situation in pigs and minks, in which diseases associated with neurotropic astroviruses are endemic and occur on the herd level. Affected animals show neurological signs such as mild ataxia to tetraplegia, loss of orientation or trembling, and the outcome is often fatal. Non-suppurative inflammation with perivascular cuffing, gliosis and neuronal necrosis are typical histological lesions of astrovirus encephalitis. Since astroviruses primarily target the gastrointestinal tract, it is assumed that they infect the brain through the circulatory system or retrograde following the nerves. The phylogenetic analysis of neurotropic astroviruses has revealed that they are genetically closely related, suggesting the presence of viral determinants for tissue tropism and neuroinvasion. In this review, we summarize the current knowledge on neurotropic astrovirus infections in animals and propose future research activities.
... Animals infected with NT-AstV in the brain show neurological signs such as tremors and limb paralysis and generally have non-suppurative encephalitis [12]. Although the mechanism by which neurological symptoms are initiated in NT-AstV infection remains unclear, it is likely via the fecal-oral route, similar to enteric AstV, which is detected in the gastrointestinal tract and feces [11,13,14]. Based on their phylogenetic analysis NT-AstVs are divided into two clades, the HMO (human-mink-ovine/bovine) clade and the MLB (Melbourne) clade [12]. ...
Preprint
Full-text available
Astroviruses (AstVs) have been detected in a wide range of animal species, including mammals and birds. Recently, a novel AstVs associated with neurological symptoms has been detected in the brain of some mammals. Raccoon dog AstV has been reported recently in China. However, there have been no reports in South Korea. Therefore, the present study aimed to detect and genetically characterize AstVs in intestine and brain tissues of 133 wild raccoon dogs collected in Korea between 2017 and 2019. Seven wild raccoon dogs were positive for AstV, four of which were also detected in brain tissue. Analysis of the capsid protein amino acid sequences of raccoon dog AstVs detected in Korea revealed a high similarity to canine AstVs, suggesting possible interspecies transmission between raccoon dogs and dogs. Phylogenetic and capsid protein amino acid sequence analysis of raccoon dog AstVs detected in brain 17-148B strain belonged to the HMO clade and exhibited conserved sequences found in neurotropic AstVs (NT-AstVs), indicating their potential as NT-AstVs. However, the pathogenicity and transmission routes of the raccoon dog AstV detected in Korea have not yet been elucidated, so further research and continued surveillance for AstV in wild raccoon dogs are needed.
Article
Full-text available
Simple Summary Goose astrovirus (GAstV) is a novel emerging pathogen that causes significant economic losses in waterfowl farming. In this study, we developed a droplet digital polymerase chain reaction (ddPCR) system for the sensitive and accurate quantification of GAstV using the conserved region of the ORF2 gene. The detection limit of ddPCR was 10 copies/µL, ~28 times greater sensitivity than quantitative real-time PCR (qPCR). The specificity of the test was determined by the failure of amplification of other avian viruses. The ddPCR test showed good repeatability and linearity, and the established ddPCR method had high sensitivity and good specificity to GAstV. Clinical sample test results showed that the positive rate of ddPCR was higher than that of qPCR. A convenient, sensitive, and specific detection method for GAstV in field samples is important in order to effectively control GAstV. Abstract (1) Goose astrovirus (GAstV) is a novel emerging pathogen that causes significant economic losses in waterfowl farming. A convenient, sensitive, and specific detection method for GAstV in field samples is important in order to effectively control GAstV. Droplet digital polymerase chain reaction (ddPCR) is a novel, sensitive, good-precision, and absolute quantitation PCR technology which does not require calibration curves. (2) In this study, we developed a ddPCR system for the sensitive and accurate quantification of GAstV using the conserved region of the ORF2 gene. (3) The detection limit of ddPCR was 10 copies/µL, ~28 times greater sensitivity than quantitative real-time PCR (qPCR). The specificity of the test was determined by the failure of amplification of other avian viruses. Both ddPCR and qPCR tests showed good repeatability and linearity, and the established ddPCR method had high sensitivity and good specificity to GAstV. Clinical sample test results showed that the positive rate of ddPCR (88.89%) was higher than that of qPCR (58.33%). (4) As a result, our results suggest that the newly developed ddPCR method might offer improved analytical sensitivity and specificity in its GAstV measurements. The ddPCR could be widely applied in clinical tests for GAstV infections.
Article
Full-text available
Astroviruses (AstVs) have been detected in a wide range of animal species, including mammals and birds. Recently, a novel AstV associated with neurological symptoms has been detected in the brains of some mammals. Raccoon dog AstV has been reported recently in China. However, there have been no reports in South Korea. Therefore, the present study aimed to detect and genetically characterize AstVs in the intestine and brain tissues of 133 wild raccoon dogs collected in Korea between 2017 and 2019. Of the seven raccoon dogs, AstVs were detected in six intestinal tissues and four brain tissues. Analysis of the capsid protein amino acid sequences of raccoon dog AstVs detected in Korea revealed a high similarity to canine AstVs, suggesting possible interspecies transmission between raccoon dogs and dogs. Phylogenetic and capsid protein amino acid sequence analysis of raccoon dog AstVs detected in the brain the 17-148B strain belonging to the HMO clade and exhibiting conserved sequences found in neurotropic AstVs (NT-AstVs), indicating their potential as NT-AstVs. However, the pathogenicity and transmission routes of the raccoon dog AstV detected in Korea have not yet been elucidated, so further research and continued surveillance for AstV in wild raccoon dogs are needed.
Article
At a dairy farm in Gunma Prefecture, Japan, a specific gene fragment of astrovirus (AstV) was detected in the cerebrum of a cow with nonsuppurative encephalitis. The cow laid down, lactation decreased, lost its appetite, and the prognosis was poor. No gross abnormalities were detected during the necropsy. Histopathological examination of the cerebrum, diencephalon, midbrain, pons, and medulla oblongata revealed multiple vessels cuffed by numerous lymphocytes and glial nodules, which were more severe in the brain stem. No pathogenic bacteria were isolated from the cerebrum and other organs, and no other viruses were detected. The nucleotide sequence of the detected AstV gene fragment had a 95% coincidence with the non-structural protein region of neurotropic AstV reported in the first confirmed case in Japan. AstV may have been involved in this case of encephalitis, which is considered to be the second reported case in Japan. This study suggests the existence of neurotropic AstV in multiple regions of Japan and infers the possibility that AstV is prevalent in Japan.
Article
While blocking inflammation is an effective way to ease the symptoms of gout disease in humans, the treatment and prevention of gout in goslings infected with goose astrovirus (GAstV), a recently emergent condition, remain unclear. In this study, we investigated the reprogramming of the host genes as a result of GAstV infection by combining analysis of the global transcriptome and metabolic network pathways in the kidneys of goslings infected with GAstV. We showed that as GAstV replication increased in vivo, the regulation of key enzymes in the host metabolism progressively increased, flowing metabolites into the purine/pyrimidine biosynthesis pathways. Furthermore, we found that GAstV: 1) inhibits the host oxidation-reduction response by inhibiting the expression of the catalase gene; 2) activates the Toll-like receptor 2 pathway to enhance the immune inflammatory response; and 3) activates the key enzyme in lactic acid synthesis to produce lactate accumulation which inhibits the host's antiviral response, so as to facilitate the replication of the virus itself. This study provided the first insight into the overall metabolic requirements of GAstV for replication in vivo by combining transcriptome with metabolic network pathway information.
Article
A novel neurological disorder, shaking mink syndrome (SMS), emerged in Denmark and Sweden since 2000. SMS has seldom been reported in China, but the causative agent has not been detected in the country. SMS outbreaks occurred in multiple provinces in 2020. A total of 44 brain samples from minks associated with SMS were collected from Heilongjiang, Liaoning, and Shandong provinces, of which 28 samples (63.3%) were SMS‐Astrovirus (SMS‐AstV) positive by reverse transcription PCR. Histopathological examination revealed non‐suppurative encephalitis in 3 minks. Moreover, the complete coding region sequences (CDS, 6559 bp) of a sample collected from a two‐month‐old mink (termed SMS‐AstV‐H1, GSA accession No. SAMC816786) were amplified by PCR and Sanger sequenced. The complete CDS and ORF2 sequences of SMS‐AstV‐H1 were 94.3% and 96.4% identical to a SMS‐AstV strain (GenBank accession number: GU985458). Phylogenetically, SMS‐AstV‐H1 was closely related to an SMS‐AstV strain (GU985458). Based on the above results, we describe SMS‐AstV‐associated encephalitis in farmed minks in China. Future studies need to focus on epidemiology, virus isolation, and potential interspecies transmission of SMS‐AstV. This article is protected by copyright. All rights reserved
Article
Full-text available
We present the latest version of the Molecular Evolutionary Genetics Analysis (MEGA) software, which contains many sophisticated methods and tools for phylogenomics and phylomedicine. In this major upgrade, MEGA has been optimized for use on 64-bit computing systems for analyzing bigger datasets. Researchers can now explore and analyze tens of thousands of sequences in MEGA. The new version also provides an advanced wizard for building timetrees and includes a new functionality to automatically predict gene duplication events in gene family trees. The 64-bit MEGA is made available in two interfaces: graphical and command line. The graphical user interface (GUI) is a native Microsoft Windows application that can also be used on Mac OSX. The command line MEGA is available as native applications for Windows, Linux, and Mac OSX. They are intended for use in high-throughput and scripted analysis. Both versions are available from www.megasoftware.net free of charge.
Article
Full-text available
In 2018, a new goose astrovirus (GAstrV) was reported in China, which causes 2 to 20% deaths in 4- to 16-day-old goslings causing great damages to the livestock industry. Gout is the typical feature of GAstrV infection in goslings. However, the mechanism of gout formation remains unclear. In the present study, 2-day-old goslings were infected intramuscularly with GAstrV for 14 D. One quarter of the infected goslings died, and typical gout pathological changes were found in the dead infected goslings. Pathological changes were observed in the morphology of the kidney and liver, such as degeneration, necrosis, and inflammatory cell infiltration. Accordingly, a high virus load was found in both organs. The serum level of uric acid in the inoculated goslings was higher, whereas no differences were found in levels of creatinine, calcium, and phosphorus. Moreover, the xanthine dehydrogenase (XOD) and adenosine deaminase (ADA) activities and the mRNA levels of xanthine dehydrogenase, adenosine deaminase, phosphoribosyl pyrophosphate amidotransferase, and phosphoribosyl pyrophosphate synthetase 1 in livers increased, wheres the multidrug resistance–associated protein 4 mRNA level and Na-K-ATPase activity in the kidneys decreased. These results showed that GAstrV infection could cause lesions on the liver and kidney and then increase the expression or activity of enzymes related to uric acid production in the liver and decrease renal excretion function, which contribute to hyperuricemia and gout formation.
Article
Full-text available
A full understanding of the immune response to astrovirus (AstV) infection is required to treat and control AstV-induced gastroenteritis. Relative contributions of each arm of the immune system in restricting AstV infection remain unknown. In this study, two novel subunit AstV vaccines derived from capsid protein (CP) of mink AstV (MAstV) such as CPΔN (spanning amino acids 161–775) and CPΔC (spanning amino acids 1–621) were evaluated. Their immunogenicity and cytokine production in mice, as well as protective efficacy in mink litters via maternal immunization, were studied. Truncated CPs induced higher levels of serum anti-CP antibodies than CP, with the highest level for CPΔN. No seronegativity was detected after booster immunization with either AstV CP truncates in both mice and mink. All mink moms stayed seropositive during the entire 104-day study. Furthermore, lymphoproliferation responses and Th1/Th2 cytokine induction of mice splenocytes ex vivo re-stimulated by truncated CPs were significantly higher than those by CP, with the highest level for CPΔN. Immunization of mink moms with truncated CPs could suppress virus shedding and clinical signs in their litters during a 51-day study after challenge with a heterogeneous MAstV strain. Collectively, AstV truncated CPs exhibit better parameters for protection than full-length CP.
Article
Full-text available
Aquaculture is the fastest growing industry worldwide. Aquatic diseases have had enormous economic and environmental impacts in the recent past and the emergence of new aquatic pathogens, particularly viruses, poses a continuous threat. Nevertheless, little is known about the diversity, abundance and evolution of fish viruses. We used a meta-transcriptomic approach to help determine the virome of seemingly healthy fish sold at a market in Sydney, Australia. Specifically, by identifying and quantifying virus transcripts we aimed to determine (i) the abundance of viruses in market fish, (ii) test a key component of epidemiological theory that large and dense host populations harbour a greater number of viruses compared to their more solitary counterparts and (iii) reveal the relative roles of virus–host co-divergence and cross-species transmission in the evolution of fish viruses. The species studied comprised both shoaling fish—eastern sea garfish (Hyporhamphus australis) and Australasian snapper (Chrysophrys auratus)—and more solitary fish—eastern red scorpionfish (Scorpaena jacksoniensis) and largetooth flounder (Pseudorhombus arsius). Our analysis identified twelve potentially novel viruses, eight of which were likely vertebrate-associated across four viral families and that exhibited frequent cross-species transmission. Notably, the most solitary of the fish species studied, the largetooth flounder, harboured the least number of viruses while eastern sea garfish, a densely shoaling fish, had the highest number of viruses. These results support the emerging view that fish harbour a large and largely uncharacterised virome.
Article
Full-text available
Background: Pre-weaning diarrhea (PWD) is a syndrome affecting farm-raised neonatal mink kits. Apart from diarrhea it causes greasy skin exudation, dehydration, and distressed behavior and can ultimately lead to death. No specific causative agents have been identified and the syndrome is regarded as multifactorial. The aim of the present study was to investigate a possible inflammatory state in mink kits with PWD, as indicated by raised serum concentrations of the acute phase protein serum amyloid A (SAA) and by changes in intestinal pathomorphology and intestinal contents of bacteria. Samples collected from 20 diarrheic mink kits with PWD and 20 age-matched non-diarrheic control mink kits from two commercial Danish farms during the pre-weaning period (April-May) in 2016 were analyzed. Results: Concentrations of SAA in serum samples from mink kits with PWD were significantly higher (up to 1000-fold) compared to non-diarrheic control mink kits. Significant features of enterocytic vacuolization, atrophy and fusion of villi in jejunum and mucosal atrophy of the colon of kits with PWD were found. Moreover, attachment of coccoid bacteria to enterocytes was more often found in kits suffering from PWD, while intra-cytoplasmic eosinophil bodies were more frequently observed in control kits. Cellular infiltrations with mononuclear and neutrophil leukocytes were not associated with disease status. Bacteria from the Staphylococcus intermedius group, such as Staphylococcus delphini, were more frequently cultivated from control mink kits, whereas Enterococcus spp. dominated in mink kits with PWD. Escherichia coli was cultivated from both control and mink kits with PWD, but with a higher frequency from mink kits with PWD. Conclusion: A significant increase in circulating concentrations of SAA was found in PWD affected mink kits from 6 to 23 days old compared to controls. The histopathological changes in PWD mink kits suggest that the type of diarrhea is secretory. Attachment of coccoid bacteria, therefore, might be responsible for an enterotoxic effect causing a loss of balance in movements of ions and water leading to the vacuolization and swelling of the enterocytes. The slight to moderate infiltrations of neutrophils irrespectively of diarrheic status and the attachment of coccoid bacteria to enterocytes are comparable to observations found in piglets suffering from New Neonatal Porcine Diarrhea Syndrome. Mechanisms behind the correlation between increased SAA levels and the observed pathological intestinal features remain obscure.
Article
Full-text available
Mink astrovirus (MiAstV) is known to play a major role in mink pre-weaning diarrhea, and rotavirus and hepatitis E virus (HEV) are both considered potentially zoonotic agents. These viruses are not monitored in commercial mink, and the role of these viral infections in mink health is not well understood. This study assessed the prevalence of mink astrovirus, rotavirus C, mink HEV and swine HEV in 527 pooled healthy adult female mink and mink kit fecal samples from 50 Canadian mink farms in two seasons over 4 years. Viral RNA was extracted and amplified in RT-PCR to detect mink astrovirus and HEV RdRp genes, swine HEV ORF2, and rotavirus C VP6 gene. At least 26% of all positive samples for each virus was sequenced for phylogenetic analysis. Fourteen percent of samples were astrovirus positive, while 3 and 9% of samples were rotavirus C and mink HEV positive, respectively. One adult female sample was found to be positive by PCR for swine HEV. A significantly higher number of kit samples were astrovirus- and HEV-positive compared to adult female samples (p = 0.01 and p < 0.0001, respectively). Astrovirus was detected in significantly more summer samples from adult females compared to winter samples from adult females (p = 0.001). The detected sequences were closely related to previously reported MiAstV, swine rotavirus C, and mink and swine HEV strains. Two astrovirus sequences were distantly related to all other detected sequences as well as previously reported MiAstVs. These results demonstrate low to moderate prevalence of the three viruses in feces from clinically healthy Canadian commercial mink, and suggest that further monitoring of these viruses may provide a better understanding of how these potentially zoonotic agents may play a role in mink enteritis and overall productivity.
Article
Full-text available
Our understanding of the diversity and evolution of vertebrate RNA viruses is largely limited to those found in mammalian and avian hosts and associated with overt disease. Here, using a large-scale meta-transcriptomic approach, we discover 214 vertebrate-associated viruses in reptiles, amphibians, lungfish, ray-finned fish, cartilaginous fish and jawless fish. The newly discovered viruses appear in every family or genus of RNA virus associated with vertebrate infection, including those containing human pathogens such as influenza virus, the Arenaviridae and Filoviridae families, and have branching orders that broadly reflected the phylogenetic history of their hosts. We establish a long evolutionary history for most groups of vertebrate RNA virus, and support this by evaluating evolutionary timescales using dated orthologous endogenous virus elements. We also identify new vertebrate-specific RNA viruses and genome architectures, and re-evaluate the evolution of vector-borne RNA viruses. In summary, this study reveals diverse virus-host associations across the entire evolutionary history of the vertebrates.
Article
Full-text available
Porcine astrovirus (PAstV) is widely distributed and highly prevalent among pigs, nevertheless its clinical significance remains unclear as it can be detected in both diarrheic and in healthy pigs. Information about the prevalence, clinical significance and molecular characterization of PAstV in Thailand is not available. This study investigated the prevalence of PAstV in 488 fecal samples collected from piglets with and without diarrhea in 28 pig farms in northern and central parts of Thailand using RT-PCR. The overall prevalence of PAstV infection was 6.5% (32/488), of which 21/251 (8.4%) were in diarrheic and 11/237 (4.6%) were in healthy pigs. Of 32 positive samples, 46.9% were positive for PAstV alone whereas 53.1% were co-infected with porcine group A rotavirus (PRVA). A phylogenetic analysis of the partial RNA-dependent RNA polymerase/capsid genes revealed two lineages of PAstV strains detected in this study. PAstV4 was the most dominant genotype (92%), followed by PAstV2 (8%). This study revealed for the first time that PAstV4 and PAstV2 were circulating in Thailand with PAstV4 as the most dominant genotype in pig herds in northern and central parts of Thailand.
Article
Astroviruses (AstVs) have been identified in the stools of calves with enteritis and in the brain tissues of bovines with encephalitis but their pathogenic role has not been clarified. In this study, we report the detection and characterization of bovine and water buffalo AstV strains identified in young bovine and buffalo calves with enteritis in Italy between 2012 and 2015. By negative staining transmission electron microscopy (TEM) observation, AstV-like particles were identified in the stools of the animals and AstV RNA was confirmed molecularly. The sequence (~3.2-kb) at the 3′ end of the genome was determined for two bovine and two buffalo AstVs. Sequence and phylogenetic analysis on the partial ORF1b and full-length ORF2 revealed a marked genetic diversity although the viruses were distantly related to other AstV identified from ruminants. Gathering sequence information on ruminant AstVs is important to understand the extent of inter-species circulation and for the development of reliable, specific diagnostic tools.
Article
Sixty-four cases of white chick syndrome (WCS) in broiler breeders producing affected progeny were reported from seven hatcheries in Ontario, Canada, between 2009 and 2016, with 43 of those originating from two hatcheries owned by a single company. WCS cases were identified by the presence of typical chicks in the hatchery that were generally weak with pale to white down, enlarged abdomens, and occasionally brown wiry fluff on the dorsum of the neck. Affected embryos and chicks had characteristic gross and histologic liver lesions, and livers were positive for chicken astrovirus (CAstV) RNA by real-time reverse transcriptase PCR. Affected broiler breeder flocks experienced egg production drops of 0% to 21% and hatchability drops of 0% to 68.4%. The amino acid sequence of the region encoding the capsid gene of WCS viruses demonstrated all Ontario CAstV to be in Group B, Subgroup Bii.