You are on page 1of 52

446

13

Viral Infections of Waterfowl

­Introduction
Simone T. Stoute

Interests in viral infections of waterfowl are more diverse of ducks and geese), Hepadnaviridae (duck hepatitis B
than for poultry and focus on diseases associated with virus), Parvoviridae (MDPV and GPV), and Polyomaviridae
wild waterfowl, problems associated with raising birds (GHPV).
commercially and problems created by migratory water­ Oncogenic viruses of waterfowl are not included in
fowl in the transmission of infectious diseases to this chapter.
­commercially reared waterfowl and poultry. The objec­
tive of this chapter is to bring together all of the viral Avian Paramyxoviruses
diseases affecting waterfowl. Avian paramyxoviruses (APMV) serotypes 1, 4, 6, 8, and
This chapter has detailed subchapters on the following 9 have been associated with infections in waterfowl.
diseases: Infections by these viruses have been demonstrated
through virus isolation or serology in various waterfowl
●● Duck hepatitis, a major disease of young ducklings attrib­
species. However, most of these APMV serotypes are
uted to at least five different viruses. Three genotypes of
generally considered to be apathogenic in waterfowl
duck hepatitis A virus (DHAV), duck atrovirus type 1
species.
(DAstV‐I), and duck astrovirus type 2 (DAstV‐II).
While waterfowl are considered to be natural reser­
●● Duck virus enteritis (DVE), caused by a herpesvirus
voirs of APMV‐1, there are several reports of APMV‐1
poses a major threat to both commercially reared and
strains associated with pathogenicity in waterfowl. Wen
wild waterfowl.
et al. reported a virulent APMV‐1 genotype III causing
●● Parvovirus infections of geese and Muscovy ducks.
respiratory and neurological disease in duck layers in
Although Muscovy duck parvovirus (MDPV) and
China (54). Zou et al. (56), and Jinding et al. (22) reported
goose parvovirus (GPV) are closely related, they are
an APMV type 1 causing high mortality in geese in
genotypically distinct. MDPV has been recognized in
China. Shi et al. (42) reported an APMV‐I isolated from
the United States.
a Muscovy duck that was pathogenic for Muscovy and
●● Goose hemorrhagic polyomavirus (GHPV) is the
Pekin ducks.
cause of hemorrhagic nephritis enteritis of geese
APMVs are dealt with in more detail in Chapter 3.
(HNEG).
Other virus infections associated with waterfowl will Avian Metapneumovirus
be covered in this introduction. Avian metapneumoviruses (aMPV), members of the
RNA viruses associated with waterfowl include family Paramyxoviridae, were first isolated in 1980s.
Picornaviridae (DHA, duck picornavirus [DPV]), They have been of most concern to the turkey industry
Astroviridae (DastV‐1 and DAstV‐2), Paramyxoviridae and were not detected in the United States until 1996.
(avian metapneumovirus, avian paramyxoviruses), US turkey isolates of aMPV are designated aMPV type
Orthomyxoviridae (avian influenza), Flaviviridae (West C, which is different from the types A and B found in
Nile virus), and Reoviridae (Duck reovirus, Muscovy duck Europe. Isolates from ducks in France have been
reovirus, and goose reovirus); DNA viruses include assigned to subgroup C but European and American
Herpesviridae (DVE, goose herpesvirus), Adenoviridae subgroup C viruses belong to different genetic lineages
(duck adenovirus), Circoviridae (circovirus‐like infection (51). Ducks often play a role as nonclinical carriers of

Diseases of Poultry, Fourteenth Edition. Editor-in-chief David E. Swayne.


© 2020 John Wiley & Sons, Inc. Published 2020 by John Wiley & Sons, Inc.
Chapter 13  Viral Infections of Waterfowl 447

aMPV and can serve as a potential source of infection Avian Influenza Virus
for domestic turkeys (43). Turpin et  al. (53) surveyed Until relatively recently waterfowl typically do not expe­
wild birds for aMPV type C in Georgia, South Carolina, rience significant disease problems due to avian influ­
Arkansas, and Ohio. Avian metapneumovirus type C enza viruses (AIVs), but infections in these birds are
was isolated from oral swabs of American coots and widespread. Avian influenza viruses can be recovered
Canada geese, demonstrating that wild waterfowl can from migratory waterfowl, particularly ducks; at least 30
serve as aMPV reservoirs. of 149 species of ducks, geese, and swans have yielded
In 1999, Toquin et al. (51) reported the isolation of a virus but natural infections are usually considered
pneumovirus from 42‐week‐old Muscovy ducks exhib­ asymptomatic (50).
iting coughing and decreased egg production. Mortality The picture changed dramatically in late 2002 when
was about 2%. Lesions of general congestion, spleno­ HPAI (H5N1) occurred in geese, ducks, and swans,
megaly and tracheitis were identified. The isolate was among other avian species, at two waterfowl parks in
confirmed as a pneumovirus by RT‐PCR on the N Hong Kong (13). The range of pathological changes pre­
gene. Using monospecific aMPV antisera, the Muscovy sent in the various waterfowl examined in this outbreak
duck virus isolates reacted most strongly with the resembled those reported generally for HPAI viruses in
aMPV Colorado (type C) antiserum when compared chickens (50). High mortality in ducks and geese was
with antisera against aMPV A, B, and non‐A and non‐B also reported in H5N1 outbreaks in India in 2011,
types. Indonesia in 2012 and Bangladesh in 2013 (12, 16, 43).
Avian metapneumovirus RNA has been isolated from Since the outbreak in 2002, H5N1 HPAI has spread
the nasal turbinates or swabs of mallard ducks, wild through Asia, Europe, and parts of Africa. The role of
geese, and sentinels captured in the north central United migratory waterfowl in the spread of HPAI H5N1 has
States. (44). The aMPV M gene from wild birds had more been investigated. Chen et al. (9) described an outbreak
than 96% predicted amino acid identity with the MN/2 A in bar‐headed geese (Anser indicus) at Qinghai Lake in
turkey aMPV isolate. McComb et al. reported the detec­ Western China in May 2005; more than 1,500 birds died.
tion of aMPV viral RNA in choanal–tracheal swabs Hulse‐Post et al. (20) reported that the H5N1 HPAI can
collected from 8‐week‐old sentinel mallard ducks
­ revert to non‐pathogenic forms in ducks. Thus, wild
allowed to mingle with wild waterfowl in central waterfowl may appear uninfected by the H5N1, but still
Minnesota (32). The authors also detected aMPV viral may continue to circulate the virus. Wild waterfowl
RNA in choanal swabs from Canada geese, blue winged migration was also implicated in the rapid global expan­
teal, and snow geese (32). sion of HPAI intercontinental group A (icA) H5N8 clade
In Minnesota, United States, aMPV‐negative mallard 2.3.4.4 in poultry in Asia in 2014 with subsequent spread
ducks placed next to a turkey farm experiencing a severe through Europe, Asia, and North America by 2015 (27).
outbreak of aPMV infection did not develop clinical dis­ There are multiple reports regarding the role of water­
ease but infectious aMPV was recovered from choanal fowl in avian influenza virus transmission and additional
swabs after two weeks and anti‐aMPV antibodies were details of this virus are covered in Chapter 6.
detected after four weeks. (43).
Bennett et  al. demonstrated aMPV in wild Canada West Nile Virus
geese (Branta canadensis) and blue winged teal (Anas West Nile virus (WNV) is a member of the Japanese
discors) by reverse transcriptase‐polymerase chain reac­ encephalitis virus antigenic complex of arthropod‐borne
tion (RT‐PCR) (5). Using Canada geese isolates, Bennett flaviviruses (Flaviviridae) that are transmitted through
et al. (5) investigated the genomic structure of the virus. mosquitoes to a variety of mammals and birds (25). The
All but one of the eight genes were similar to those of a notoriety of this virus has increased since 1999 when an
turkey aMPV isolate in terms of size, sequence identity, epizootic causing death in wild American crows (Corvus
and genome organization. This virus replicated in the brachyrhynchus) began in New York (36). This was the
upper respiratory tract of experimentally challenged first time that WNV had been detected in North
domestic turkeys without eliciting clinical signs and America. At the same time, WNV‐positive cases
could be horizontally transmitted to naïve turkeys and occurred in a number of wild bird species, humans, and
elicit aMPV specific antibody production. The authors horses, and at zoological collections of mammals and
suggested that this virus may be a safe and effective vac­ birds in New York (25, 26, 48).
cine for commercial turkeys (5). Outbreaks of WNV involving ducks and geese have
In 2014 Sun et al. reported a subgroup C aMPV circu­ been reported from Israel and Romania (26, 35, 40). In
lating in Muscovy duck breeders in South China (49). the New York outbreak, Steele et al. (48) examined birds
Upper respiratory disease, decreased egg production, from two wildlife facilities, which had either died or were
thin‐shelled eggs and mortality exceeding 10% was noted euthanized after suspected of being infected with WNV.
in some of the affected Muscovy flocks. Lesions in emaciated Anseriformes included cerebral
448 Section II  Viral Diseases

hemorrhages, massive necrotizing splenitis, splenomeg­ authors consider GRV and DRV to belong to a species
aly, nephritis, and congestion in the kidneys. One mal­ distinct from others established within the subgroup 2 of
lard and two bronze‐winged ducks were involved with orthoreoviruses.
this study. In the mallard, abundant antigen was demon­ Hollmen et al. (17) reported the isolation of a reovirus
strated by immunohistochemistry in the brain, heart, from common eider ducks (Somateria mollissima) in
liver, kidney, and pancreas, and to a lesser extent in the Finland. The virus was isolated from the bursa of
adrenal gland and intestine. Virus in excess of 102 Fabricius, inoculated into Muscovy duck embryo fibro­
pfu/0.2 ml was isolated from brain, heart, spleen, liver, blasts. The relationship of this reovirus to DRV and GRV
and kidney; virus also was demonstrated by RT‐PCR in has not been reported.
these same tissues. Only immunohistochemistry was A reovirus causing duck viral swollen head hemor­
performed on the tissues from the bronze‐winged ducks, rhagic disease in Pekin ducks has been reported from
but results obtained were similar to those in the China (28), and an indirect immunoperoxidase assay was
mallard. developed for detection of the virus. Liu et al. (29) report
In the 1999 outbreak in New York, an Abyssinian blue‐ the isolation and characterization of a reovirus causing
winged goose (Cyanochen cyanopterus), a Rosybill duck spleen necrosis in Pekin ducklings. DNA sequencing
(Netta peposcaca), and a domestic goose (Anser anser) revealed that the isolate was closely related to Muscovy
showed asymptomatic seroconversion to WNV. In the duck reoviruses.
domestic goose and trumpeter swan (Cygnus cygnus
buccinator) morbidity and recovery were recorded. No Goose Herpesvirus (GHV)
deaths were recorded in any of the birds. The New York In Australia, a herpesvirus has been implicated in a pera­
isolates of WNV yielded an E gene nucleotide sequence cute disease of domestic geese that caused 97% mortality
that was closely homologous to that from the WNV over a 24‐day period. Clinical signs and gross pathology
­isolated from a goose in Israel in 1998 and also from a were similar to those seen with DVE infections.
1996 Romanian isolate. Histologically, button ulcers and large plaques overlying
Bird to bird transmission of West Nile virus in geese lymphocyte aggregates were present on the small intesti­
has been proposed and some studies have reported direct nal mucosa. Focal necrosis and hemorrhages were seen
(non‐vector) transmission of WNV in geese (1, 3). Banet‐ in the livers. Numerous intranuclear hepatic inclusion
Noach et al. concluded that horizontal transmission can bodies were observed. A herpesvirus was isolated in var­
occur in commercial flocks and may be aggravated by ious primary chicken and duck embryo cell cultures.
cannibalism and feather picking of sick birds (3). This virus was not neutralized by DVE antiserum. In
Detection of WNV neutralizing antibodies has been experimental transmission studies, the virus caused
reported in wild migratory ducks in Japan (39) and South 100% mortality in adult domestic geese, 50% mortality in
Korea (55) and domestic (23) and wild ducks (33) in 1‐day‐old commercial ducklings, and 25% mortality in 4‐
India. to 6‐week‐old ducklings (24). However, Pekin ducks are
not susceptible to GHV (15).
Duck, Muscovy Duck, and Goose Reoviruses Using cross‐neutralization tests in cell cultures, GHV
Muscovy duck reovirus infections have been described. was compared with five other avian herpesviruses. No
In France, it is considered to be a major virus disease of significant cross neutralization was reported, confirming
Muscovy ducks. In 2‐ to 4‐week‐old ducks, the disease is that GHV is antigenically distinct from DVE viruses (15).
acute, morbidity is high, and mortality can reach 10%.
Clinical signs include apathy, diarrhea and difficulty Adenoviruses
moving. Lesions include fibrinous pericarditis, splenic Duck adenovirus 1 (DAdV‐1) (previously known as
necrosis, and hepatic necrosis and mononuclear hepati­ group 3 avian adenovirus, egg drop syndrome‐1976 virus
tis and exudative synovitis of leg tendons. Palya et al. (37) [EDS], avian adenovirus EDS, and egg drop syndrome
reported reovirus causing disease in young geese. The virus) is a member of the genus Atadenovirus, species
disease was characterized by splenitis and hepatitis with Duck adenovirus A. Ducks and geese are assumed to be
miliary necrotic foci during the acute phase, and epicar­ the natural hosts of this virus, but there is sparse evi­
ditis, arthritis, and tenosynovitis during the subacute/ dence of respiratory disease in waterfowl associated with
chronic phase. this virus. Ivanics et  al. (21) reported in 2001 an acute
Cross‐neutralization tests have demonstrated that respiratory disease in goslings in Hungary attributable to
Muscovy duck reovirus is antigenically distinct from EDS virus.
chicken reovirus S1133. Banyai et al. (4) investigated the Aviadenoviruses have also been isolated from mallards
genetic variability among goose reoviruses (GRV). The and a Muscovy duck (7). In Muscovy ducks, mortality
S4 genome segment of 5 GRVs shared substantial struc­ occurred, and the isolated adenovirus was tentatively
tural similarity with Muscovy duck reovirus (DRV). The named Duck adenovirus 2.
Chapter 13  Viral Infections of Waterfowl 449

Unclassified Adenoviruses  Hollmen et  al. (18) reported Smyth et al. (45) investigated a circovirus infection in
the isolation of an adenovirus from long‐tailed ducks geese by in situ hybridization using a GoCV DNA probe
(Clangula hyemalis) collected during a die‐off in the that showed that circovirus DNA could be demonstrated
Beaufort Sea off the north coast of Alaska. The authors in the cloacal bursa, spleen, thymus, bone marrow, liver,
reproduced the disease experimentally in long‐tailed kidney, lung, and heart.
ducks; no mortality was recorded but clinical signs Diagnostic tests for GoCV and DuCV are mostly PCR
included watery feces and blood in the feces. Challenged based (8). Liu et al. (30) have since reported the develop­
ducks seroconverted. The virus could not be neutralized ment of an indirect enzyme‐linked immunosorbent
by reference antisera to group 1, 2, or 3 avian adenoviruses assay (ELISA) for the detection of DuCV infection in
and may represent a new serotype. flocks using recombinant capsid protein as antigen.
Hollmen et  al. (19) also reported on an adenovirus Glavits et al. (14) investigated an outbreak of West Nile
associated with impaction of the posterior small intes­ virus infection in a goose flock in Hungary. Histological
tine with mucosal necrosis and the cause of death in 10 lesions suggestive of WNV and circovirus were identi­
male common eider ducks (Somateria mollissima) in the fied and both virus infections were diagnosed by molec­
northern Baltic Sea near Finland. The adenovirus iso­ ular diagnostic tests.
lated from cloacal swans could not be neutralized with A duck circovirus (DuCV) was detected by PCR in
reference antisera to group 1, 2, or 3 avian adenoviruses. bursal and thymic samples from Pekin ducks from New
The virus caused clinical signs of illness and gastrointes­ York. The birds exhibited bursal and thymic atrophy and
tinal pathology in an experimentally infected mallard arthritis caused by Staphylococcus aureus (2). This is the
duckling. first report involving Pekin ducks. Genetic diversity of
Cheng et  al. (10) reported attempts to characterize a DuCV has been reported.
virus causing enteritis in goslings in China. They identi­
fied an adenovirus and named the virus new gosling viral Miscellaneous Viral Infections
enteritis virus (NGVEV). Tembusu viral infection is an emerging flavivirus infec­
For further references on characterization, see previ­ tion reported in ducks and geese in Asia. Infection has
ous editions of Diseases of Poultry. For more information been associated with anorexia, diarrhea, decreased egg
on adenoviruses see Chapter 9. production, ataxia and paralysis. Tembusu virus infec­
tion has been associated with lesions of hepatomegaly,
Circovirus‐Like Infection of  Ducks and  Geese  Soike et  al. meningeal congestion, myocardial hemorrhage, and
(47) first reported in 1999 the presence of circovirus‐like ­pulmonary edema (31).
particles, approximately 15 nm in diameter, in cloacal Tsai et  al. (52) reported that 77.3% of 611 ducks and
bursa, splenic, and thymic tissues from a flock of Czech 70.9% of 542 geese in Taiwan were positive for antibodies
hybrid geese with a history of increased losses and to Japanese encephalitis virus.
runting. Since then a virus has been isolated from two Bidin et  al. (6) reported the first evidence of avian
female mulards showing characteristic signs of circovirus nephritis virus (ANV) in ducks. The report from Croatia
infection (46). This duck circovirus (DuCV) has been confirmed the diagnosis by RT‐PCR; high nucleotide
shown to be closely related phylogenetically to goose and amino acid identities were shared with other ANV
circovirus (GoCV), but is still distinct. polymerase (ORF 1b) genes.
Avian circovirus infections which occur in the first Avian bornavirus (ABV) associated with non‐suppura­
months of life are characterized by developmental and/ tive inflammation in the central peripheral and auto­
or feathering disorders. The virus invades the lymphoid nomic nervous systems has been detected in wild,
tissues and leads to immunosuppression, growth retar­ free‐ranging Canada geese and trumpeter swans (Cygnus
dation, and an increased probability of secondary infec­ buccinator) in Ontario, Canada. The virus was detected
tions. In geese the only apparent gross lesions in 2‐week‐old in brain by immunohistochemistry and RT‐PCR (11).
and 9‐week‐old birds were a cloudiness of the air sacs ABV also has been detected in healthy Canada geese by
(47). Lesions included lymphocytic depletion and histio­ RT‐PCR; the virus was propagated in duck embryo fibro­
cytosis of lymphoreticular tissue; and was most apparent blast cells and detected by RT‐PCR after two passages
in the cloacal bursa. Basophilic globular intracytoplas­ and indirect fluorescent antibody assay after three pas­
mic inclusions were found in the bursa follicular and epi­ sages antibody (38).
thelial cells. In naturally infected commercial Muscovy,
mule, Pekin ducks, and White Roman geese in Taiwan,
4‐ to 6‐week‐old birds had clinical signs of loss of wing ­Acknowledgement
and body feathers, necrosis of feather follicles, and
stunted growth. The most common lesion was polyse­ The author is greatly indebted to Dr. P.R. Woolcock for
rositis (8). his contribution to earlier editions of this subchapter.
450 Section II  Viral Diseases

­Duck Hepatitis
Hsiang-Jung Tsai

Summary DHAV‐3, isolated in South Korea (65) and China (38),


also belong to the Avihepatovirus genus. Both DHV type
Agent, Infection, and Disease.  Duck hepatitis is an acute, 2 and DHV type 3 viruses now belong to the genus
highly fatal, and contagious viral disease of young Avastrovirus in the family Astroviridae, and are referred
ducklings. The disease is characterized by a short to as duck astrovirus type 1 (DAstV‐1) and type 2
incubation period, sudden onset, opisthotonos, high (DAstV‐2), respectively (11). For other reviews of DHV
mortality, and characteristic liver lesions. Three distinct types 1 and 3, see Calnek (12), and for DHV type 2, see
types of duck hepatitis virus (DHV) have been identified. Gough and Stuart (47).
The most internationally distributed and economically In addition to the five viruses that are etiologically
important is DHV type I, a picornavirus. Three associated with liver disease in ducks, a member of the
genotypes, and probably also serotypes, of DHV type I hepadnavirus group (duck hepatitis B virus; DHBV) can
have been identified and designated as duck hepatitis A also be found in wild and domestic ducks. Within the
virus (DHAV) types 1, 2, and 3. While DHV type 2 and Picornaviridae family, three novel duck picornaviruses
DHV type 3 viruses are two distinct astrovirus. have also been found in domestic ducks. One belonged
to the genus Sapelovirus (115), another to the genus
Diagnosis.  A presumptive diagnosis can be made on the Megrivirus (74), and the third a newly proposed genus,
basis of the characteristic disease pattern in the flock and Aalivirus (120). Also, a novel duck astrovirus has been
gross pathological lesions in the liver, but isolation and detected in newly hatched ducklings, and the nucleic
identification of the causative agent are necessary to acid sequence indicates that it is genetically different
confirm the diagnosis. The disease caused by all DHV from DAstV‐1 and ‐2 viruses (78). All of these novel
types in ducklings is indistinguishable, and molecular duck picornaviruses, novel duck astroviruses, and
diagnostic methods are commonly used for rapid DHBVs are not known to be associated with hepatitis in
identification and differentiation of the etiological DHV ducklings.
types.
Economic Significance and Public Health
Intervention.  Resistance against DHV may be conferred Significance
to ducklings by active immunization of ducklings with
live vaccine, or by injecting ducklings with immune Duck hepatitis is of economic importance to all duck‐
serum or yolk, or by maternal antibodies derived from growing farms because of the high potential mortality if
breeding stock immunized with live or inactive vaccines. not controlled. All three DHV types are not known to
have any public health significance.

Introduction History
Definition and Synonyms Duck Hepatitis Virus Type 1
Duck hepatitis (DH) or duck viral hepatitis (DVH) is a DHV type 1 was first observed in young White Pekin
highly fatal, rapidly spreading, viral infection of young Ducks on Long Island in 1945 (72). Since then, DHV
ducklings characterized primarily by hepatitis. The dis­ type 1 has been reported in duck‐raising areas world­
ease can be caused by at least five different viruses. wide (137). The causative pathogen, DHAV‐1, was first
Traditionally, viruses causing DH were classified into isolated in chicken embryos in 1950 (71), and was
three serotypes: DHV type 1, DHV type 2, and DHV type ­originally classified as an enterovirus (124) until the
3. The most pathogenic and widespread is DHV type 1 complete genome was determined in 2006 (68).
(6, 12, 28, 68, 70). According to the Ninth Report of the Presently, it is classified in the new genus Avihepatovirus
International Committee on Taxonomy of Viruses, DHV in the family Picornaviridae (113). In 2007, two anti­
type 1 has been renamed duck hepatitis A virus type 1 genically and genetically distinct viruses were identi­
(DHAV‐1) and is classified in the newly proposed genus fied: DHAV‐2, isolated in Taiwan (113), and DHAV‐3,
Avihepatovirus in the Picornaviridae family (141). Two isolated in South Korea (66). For historical details of the
newly described DHAV genotypes (which may also be disease, please refer to the previous edition of Diseases
serotypes), DHAV‐2, isolated in Taiwan (114), and of Poultry (126).
Chapter 13  Viral Infections of Waterfowl 451

Duck Hepatitis Virus Type 2 and Type 3 Chemical Composition


Duck hepatitis virus types 2 and 3 were recognized as Duck Hepatitis Virus Type 1
separate entities because they induced hepatitis in DHV The complete genome of DHAV‐1 isolates from Korea
type 1‐immune ducklings (6, 12, 37, 44, 45, 47, 52, 84, 109, (68), Taiwan (113), and China (28, 75) have been deter­
111). Duck hepatitis virus type 2 was first isolated from mined and shown to comprise a positive‐sense, single‐
an outbreak of hepatitis in ducklings in Norfolk, England stranded ribonucleic acid (RNA) of about 7,800
in 1965 by Asplin (6). The disease disappeared from com­ nucleotides in length, with a poly (A) tail at the 3′ end.
mercial flocks by 1969, but reappeared from 1983 to 84 The genomic organization of DHAV‐1 is typical of a
on three farms, again in Norfolk, England (44). DHV‐2 picornavirus, with a single, long, open reading frame
was originally regarded as a picornavirus, but later was (ORF) flanked by 5′ and 3′ untranslated regions (UTRs).
characterized as an astrovirus by morphology in 1984, The ORF can be translated into a large polyprotein,
and later renamed as duck astrovirus 1 (DAstV‐1) in 1984 about 2,200 amino acids in size, which appears to be
(42, 45, 84). Since the outbreaks in the mid‐1980s, there cleaved into 12 mature products, in order from the 5′ to
have been no additional outbreaks of the disease in that 3′ end, forming its structural (VP0, VP3, and VP1) and
area (43). Outside of the UK, outbreaks of DVH caused by nonstructural proteins (2A1, 2A2, 2A3, 2B, 2C, 3A, 3B,
DAstV‐1 have been reported in China in 2008 and 2012 3C, and 3D). Three in‐tandem 2A genes are unique fea­
(17, 37). Duck hepatitis type 3 was first reported in 1969 tures of DHAV‐1. Among them, 2A1 protein is an aph­
by Toth (111), and last observed in 1975 (53) on Long thovirus‐like 2A protein, and 2A3 protein is a human
Island in the United States. Duck hepatitis type 3 was parechovirus‐like 2A protein, whereas 2A2 protein is not
originally thought to be a picornavirus and later was related to any known picornavirus proteins (113). The
­characterized as an astrovirus in 2009 (109). 2A2 protein of DHAV‐1 was shown to induce apoptosis
in primary cell culture (14).
The DHAV genes for VP0, VP1, and VP3 encode cap­
Etiology sid proteins, which could be the main viral antigen
epitopes that confer specific antigenicity. Among them,
Classification the VP1 protein has been shown to probably play a vital
role in receptor binding, virulence, immunogenicity, and
Duck Hepatitis Virus Type 1 protection against DHAV (75). Sequence analysis results
Duck hepatitis A virus (DHAV) belongs to the genus have shown that mutations were distributed mainly in
Avihepatovirus in the Picornaviridae family. As the only the gene encoding VP1 (17, 75, 118, 122, 138). Li et al.
species in its genus, DHAV consists of three distinct gen­ (72) also showed that VP1 of DHAV‐1 is a target of neu­
otypes and probably also serotypes. They are designated tralizing antibodies and involved in receptor‐binding
DHAV‐1, DHAV‐2 and DHAV‐3 (109). activity. The gene encoding the 3D RNA‐dependent
RNA polymerase, which is responsible for the synthesis
Duck Hepatitis Virus Type 2 and 3 of the viral RNA, is highly conserved among DHV‐1
Duck hepatitis virus type 2 belongs to the genus strains (105). DHAV‐1 has no hemagglutination activity
Avastrovirus in the Astroviridae family, and has been (34, 108).
renamed duck astrovirus 1 (DAstV‐1) (42, 85, 109). Duck
hepatitis virus type 3 was originally classified as a picor­
navirus (53), but more recently it was reclassified as also Duck Hepatitis Virus Type 2 and 3
an astrovirus and was named duck astrovirus type 2 Astroviruses are non‐enveloped, single‐stranded, posi­
(DAstV‐2) (109). tive‐sense RNA viruses. The genome arrangement is
ORF1a and ORF1b at the 5′ end, encoding the viral pro­
tease and the RNA‐dependent RNA polymerase, and
Morphology
ORF2 at the 3′ end encoding the precursor capsid
Duck hepatitis virus type 1 viruses are of typical picor­ ­protein (11).
navirus morphology; naked with an icosahedral capsid The complete genome of DAstV‐1 is about 7,752
and estimated to be 20–40 nm in size (94, 95, 108). Duck nucleotides (nt) in length with a 30 nt poly(A) tail, a
hepatitis virus type 2 viruses have an astrovirus‐like 7,483 nt coding region, a 22 nt short 5′ UTR, and a 247 nt
morphology, and a diameter of 28–30 nm, as seen by 3′ UTR. The coding region includes three overlapping
electron microscopy (EM), has been reported (46). Duck ORFs (ORF1a, ORF1b, and ORF2), which encode poly­
hepatitis virus type 3 viruses have a cubic symmetry, peptides of 1,240; 516; and 731 amino acids, respectively.
and are about 30 nm in diameter as observed in cyto­ The ORF2 of DAstV was not found to be in the same
plasmic crystalline arrays of infected cultured duck reading frame as either ORF1a or ORF1b, which was dis­
­kidney (DK) cells by EM (52). tinct from all other astroviruses (17, 37).
452 Section II  Viral Diseases

Virus Replication formalin, pH 3.0, and 1 M Mg2+ treatment at 56°C and
60°C. Also, both viruses were sensitive to these tempera­
Adamiker (2) reported on the in vivo replication of
tures in the absence of the cation, with DHAV‐2 being
DHAV‐1. Six‐day‐old ducklings were infected intrana­
more resilient.
sally and intramuscularly with DHAV‐1 and killed 1–24
hours later; their livers were examined by EM. Virus‐like
Duck Hepatitis Virus Type 2 and 3
particles were detected at 1 hour and 18–20 hours
DAstV‐1 is resistant to chloroform, pH 3.0, trypsin treat­
postinfection (PI) (2). In another study, one‐day‐old
ment, and heating at 50°C for 60 minutes. Formaldehyde
ducklings were subcutaneously injected with a virulent
fumigation and standard disinfection procedures
or an attenuated strain of DHAV‐1 and killed 36 hours
have eliminated the infection from contaminated prem­
later. The results indicated that both virus strains repli­
ises (47).
cated well in the liver, spleen, and intestine; however, the
DAstV‐2, as an RNA virus, is also insensitive to iodo­
virulent strain replicated more efficiently than the atten­
deoxyuridine (IUdR). It has been found to be resistant to
uated strain (104).
chloroform and pH 3.0, but sensitive to 50°C irrespective
Yao et al. (140) studied the replication cycle of DHAV‐1
of the presence of 1 M MgCl2 (52).
in duck embryonic hepatocytes by monitoring the
dynamic changes of the relative DHAV‐1 gene expres­
sion. The results suggested that the absorption of Strain Classification
DHAV‐1 progressive increase from 15–90 minutes and
Duck Hepatitis Virus Type 1
reached a peak at 90 minutes PI, and replication of
The diseases caused by all the different DHV types can­
DHAV‐1 gradually increases and reached a peak at
not be differentiated based on clinical manifestation and
11–13 hours PI, and the release of DHAV‐1 was in steady
pathology. But DHV type 1, 2, and 3 are genetically and
state after 32 hours PI.
serologically distinct from each other. Among DHV type
1, both DHAV‐2 and DHAV‐3 have also been shown to
be distinct genetically and serologically from DHAV‐1
Susceptibility to Chemical and Physical
(38, 65, 113). It is unknown whether DHAV‐2 is serologi­
Agents
cally distinct from DHAV‐3. However, the nucleic acid
Duck Hepatitis Virus Type 1 sequence differences that have been found in capsid‐
DHAV‐1 is resistant to lipid solvents, such as ether and coding regions (approximately 30% divergence in nucle­
chloroform; relatively heat stable; and capable of survival otides) suggest that they may belong to different
for long periods of time under usual environmental con­ serotypes (118).
ditions. DHAV‐1 has been shown to resist treatment A variant strain of DHAV‐1, named DHAV‐1a, has
with ether or fluorocarbon (88), chloroform, pH 3 and been described (98). Partial cross‐protection between
trypsin (108), and 30% methanol or ammonium sulfate types 1 and 1a has been demonstrated in vitro and in vivo
(54), 2% lysol or 0.1% formalin (8). Complete inactivation (98, 135). Viruses differing or serologically distinct from
of DHAV‐1was reported with 1% formaldehyde or 2% DHAV‐1 have been recognized as causes of hepatitis in
caustic soda within two hours at 15–20°C, and 2% cal­ ducklings in India (92) and Egypt (100). The Indian iso­
cium hypochlorite within three hours at 15–20°C (91). It late is known to be distinct from DHAV‐l, but its rela­
was reported that most of DHVA‐1 was inactivated after tionships to the other DHV types are not known.
30 minutes at 56°C (54) and that heat stability was unaf­
fected by a 1 M solution of divalent cations (Mg2+) (110). Duck Hepatitis Virus Type 2 and 3
Using cell culture‐grown DHAV‐1, the half‐life of the DAstV‐1 has been compared with astrovirus isolates
virus was determined to be 48 minutes at 50°C. However, from chickens and turkeys in cross‐protection and trans­
in the presence of NaCl, Na2SO4, MgCl2, or MgSO4, the mission studies, and found to be antigenically distinct
virus was protected from inactivation at that tempera­ (47). Phylogenetic analyses have shown that DAstV‐2 is
ture (24). It has been shown that DHAV‐1 can survive for more closely related to turkey astrovirus type 2 than
21 days at 37°C (90). Under more natural environmental DAstV‐1 (109).
conditions, the virus can survive at least 10 weeks in
uncleaned, infected brooders, and for longer than 37
Laboratory Host Systems
days in moist feces stored in a cool shed (8). At 4°C, the
virus has been shown to survive more than 2 years (8, 32) Duck Hepatitis Virus Type 1
and at – − 20°C for as long as 9 years (53). DHAV‐1 is readily propagated in chicken and duck
Tseng and Tsai (114) demonstrated that DHAV‐2 is embryos (70). Levine and Fabricant (70) were the first to
similar to DHAV‐1 in several physical and chemical propagate the virus in the allantoic sac of 9‐day‐old
characteristics. Both viruses are resistant to chloroform, chicken embryos. Ten percent to 60% of the embryos
Chapter 13  Viral Infections of Waterfowl 453

died by the fifth or sixth day PI and were stunted or A description of other attempts to grow and assay
edematous (Figure 13.1). Death of the chicken embryos DHAV‐1 in other host systems and cell cultures of vari­
inoculated with the attenuated strain was found to occur ous origins can be found in the previous edition (136).
as early as the third or fourth day PI (Tseng and Tsai,
unpublished data). Hwang and Dougherty (61) passaged Duck Hepatitis Virus Type 2
a DHAV‐1 strain as two lines in 10‐day‐old chicken DAstV‐1 has been replicated in embryonating chicken
embryos. The serially passaged lines became nonpatho­ eggs following several blind passages in the amniotic sac,
genic for newly hatched ducklings after the twentieth and attenuation of pathogenicity was found to occur
and twenty‐sixth transfers. The virus titer in chicken after serial passage in chicken embryos (44). DAstV‐1
embryos was 1–3  log10 lower than when grown in has not been propagated efficiently in various duck and
ducklings. chicken cell cultures (44, 136); therefore, production of
Golubnichi et al. (41) reported successful growth and enough antigen for diagnostic tests is difficult.
an extensive cytopathic effect (CPE) in duck embryo
fibroblasts inoculated with chick embryo‐adapted Duck Hepatitis Virus Type 3
DHAV‐1. Woolcock (127) reported that primary duck DAstV‐2 has been grown successfully in embryonating
embryo liver (DEL) cells are particularly sensitive to eggs of ducks, but not of chickens (52, 136). During the
DHAV‐1, with a CPE characterized by cell rounding and first passages embryo deaths were erratic and did not
necrosis. A plaque assay has also been developed for occur until the eighth or ninth day PI, but this was reduced
DHAV‐1 in primary monolayers of duck embryo kidney with higher passages. In severely affected embryos, the
(DEK) cells (132) and DEL cells (127). Kaleta (62) CAMs were discolored and the surface of the affected
described a microneutralization assay using attenuated areas had a dry crusty or cheesy appearance. Underneath,
DHAV‐1 in primary DEK cells, which was used to the CAM was edematous and thickened up to 10 times
­monitor immune responses to vaccines (129). normal. Embryo lesions included stunting, edema, skin
Recently, Wang et  al. (119) described the establish­ hemorrhages, flaccid appearance, gelatinous fluid accu­
ment of a duck embryo epithelial cell line and suggested mulations and enlargement of liver, kidneys, and spleen.
its use for DHAV‐1 propagation and vaccine development. Attenuation of pathogenicity for ducklings, accompanied
by increased pathogenicity for duck embryos, was found
to occur following serial passage in embryonating duck
eggs inoculated by the CAM route (51).
Liver and kidney cell cultures of duck embryo or duck­
ling origin were shown to support replication of the virus
(52). Woolcock (127) reported that DAstV‐2 failed to pro­
duce plaques in primary DEK and DEL cell monolayers.

Pathogenicity
Until recently, DHAV‐I infection had only been associ­
ated with acute hepatitis in ducklings; however, it has
now been reported to cause pancreatitis and encepha­
litis in Muscovy ducks (49, 50). After 20 or more pas­
sages in chicken embryo passages, DHV‐1 was found
to have lost its pathogenicity for ducklings (5, 56, 58,
93). Hwang and Dougherty (61) reported that chicken
embryo‐passaged strains, while nonpathogenic for
ducklings, multiplied in the tissues, but at lower titers.
Field strains were found in high concentrations in
duckling brain, while chicken embryo‐passaged strains
could not be detected or were present in low titers. A
similar attenuation of pathogenicity has been reported
when DHAV‐1 was passaged in duck embryos (10).
Embryo passage‐attenuated DHAV‐1 strains are still
capable of causing very mild and transitory histologic
Figure 13.1  Normal 15‐day‐old chick embryo (right). Fifteen‐day‐
old chicken embryo inoculated six days previously with duck changes after inoculation (97, 107), and reversion to
hepatitis A virus (DHAV) type 1 (left). Note small size, hemorrhage, virulence occurs after back‐passage in young ducklings
and edema. (133, 134).
454 Section II  Viral Diseases

Pathobiology and Epidemiology The flocks showed about 20–40% morbidity and less
than 5% mortality (76).
Incidence and Distribution
Duck Hepatitis Virus Type 2 and Type 3
Duck Hepatitis Virus Type 1 Ducks appear to be the only species affected by DAstV‐1,
DHAV‐1 is worldwide in distribution (125, 128). DHAV‐2 and no wildlife reservoirs or vectors have been detected.
has been reported in Taiwan (114) and DHAV‐3 has been All recorded outbreaks have initially involved ducks kept
reported in South Korea (65), China (37), and Viet Nam on open fields; therefore, wildfowl, gulls, and other wild
(28). Recent studies indicated a higher prevalence rate of birds have been suspected as being vectors (41).
DHAV‐3 infection than DHAV‐1 infection in Korean, DAstV‐2 has been found to have low pathogenicity for
Vietnamese, and Chinese duck farms (29, 80, 103). ducklings experimentally infected, and only ducklings
appear to be affected by the virus. Subcutaneous (SC) or
Duck Hepatitis Virus Type 2 and 3 intramuscular (IM) inoculation of liver homogenate
Previously, DHV type 2 had only been reported in from infected ducklings into susceptible day‐old duck­
England. Recently, outbreaks in commercial duck flocks lings is unreliable. Intravenous inoculation may increase
of DH caused by DAstV‐1 have also been reported in the effectiveness (13).
China (17, 37). Disease caused by DHV type 3 is only
known to have occurred in the United States.
Age of Host Commonly Affected
In naturally occurring outbreaks, all three DHV types
Natural and Experimental Hosts
have been found only in young ducklings. The ducklings
Duck Hepatitis Virus Type 1 have been found to gradually become more resistance as
Young ducks and geese are susceptible to DHAV‐1 infec­ they grow older. Adult breeders on infected premises
tions. Experimental infections in goslings (4) and mal­ have not been found to become clinically ill and have
lard ducklings (36) have been reported. In experimentally been found to continue in full production.
exposed birds, no mortality occurred in chicks, Muscovy
ducklings, or pigeon squabs; low mortality occurred in
Transmission, Carriers, and Vectors
young turkeys and quail, while high mortality occurred
in young pheasants, geese, and guinea fowl. All exposed Duck hepatitis A virus is excreted in the feces from
birds became infected with DHAV‐1 (60). In another infected ducklings and is transmitted horizontally by
experimentally exposed study, Tsiay duck (Anas platy- direct contact between birds or through fomites, such as
rhynchos var. domesticus) and Cherry Valley ducklings brooders, water, feed, equipment. Under field condi­
were highly sensitive, and Pekin ducklings and hybrid tions, DHAV spreads rapidly to all susceptible ducklings
ducklings of Tsiay duck and Pekin duck were relatively in the flock. Although high mortality and rapid spread of
sensitive. Muscovy ducklings were least sensitive, the disease on farms indicate extreme contagiousness,
whereas mule ducklings showed intermediate sensitivity occasional exceptions have been observed. In one pen,
to infection (Tseng and Tsai, unpublished data). 65% of the ducks died, while in an adjoining pen which
Field observations have indicated that all duck breeds was separated only by a 14 inch curb, mortality was
are susceptible to DHAV‐1 infection, while chickens and ­negligible (135).
turkeys were resistant. Asplin (8) reported that young Field experience with DHAV‐1 has indicated that egg
chickens can contract an inapparent infection and pass it transmission does not take place. Newly hatched ducklings
on through contact with other chicks. Rahn (91), how­ produced by breeders on infected premises have remained
ever, found that day‐old and week‐old poults exposed to well when housed where no ducks were being kept (5).
DHAV‐1 developed signs, lesions, and neutralizing Ducklings may also become infected by respiratory or oral
­antibody. Poults, after either oral or intraperitoneal expo­ routes (54, 90). The portal of entry may be the pharynx or
sure, had mottled livers and enlarged gall bladders and upper respiratory tract, because the virus administered in
spleens. DHAV‐1 was isolated from livers up to 17 days a capsule failed to produce infection (112).
after oral exposure of day‐old poults. Schoop et al. (199) Recovered ducks may excrete virus in feces for up to
and Reuss (94) failed to infect chickens experimentally. eight weeks after infection (94). Asplin (8) reported that
In 2004, DHAV‐2 was isolated in Taiwan from 1‐week‐ there is strong field evidence to incriminate wild birds as
old goslings in a white Roman goose flock experiencing mechanical carriers of the virus over short distances. He
mortality exceeding 70%. The affected goslings displayed also suggested the possibility that an unknown host
liver hemorrhage typical of DH (114). Recently DHAV‐3 ­acting as a healthy carrier might be responsible for new
has been reported to cause a new disease in overfed outbreaks at great distances. However, no serologic
geese in China characterized by hemorrhagic hepatitis. ­evidence of DHAV‐1 infection was found in 520 wild
Chapter 13  Viral Infections of Waterfowl 455

aquatic fowl of 6 species (7), or in 4 species of 36 wild


ducks taken from ponds where DHAV type 1 had
occurred in domestic ducks (116). In addition, all 153
wild duck embryonating eggs from an infected area were
susceptible to experimental infection.
Of possible significance in the epizootiology of the
­disease is a report indicating that brown rats (Rattus nor-
vegicus) could act as a reservoir host of DHAV‐1 (27).
Ingested virus remained alive up to 35 days and the virus
was excreted 18–22 days postinoculation (PI). Serum
antibodies were also present 12–24 days PI. However,
there has been no evidence of the involvement of any
vectors in transmission of DHAV‐1.

Incubation Period
The incubation period for DHAV‐1 is 18–48 hours (130).
For DAstV‐1 and DAstV‐2 infection, the incubation
period is 2–4 days (44, 111).

Clinical Signs
Duck Hepatitis Virus Type 1
Onset and spread of DHAV‐1 are very rapid, with practi­ Figure 13.2  Duckling dead from infection with duck hepatitis A
cally all mortality occurring within 3–4 days. Affected virus (DHAV) type 1. Note typical opisthotonos.
ducklings at first fail to keep up with the brood. Within a
short time, they stop moving and squat down with
Morbidity and Mortality
eyes  partially closed. Birds fall on their sides, kick
­spasmodically with both legs, and die with heads drawn Duck Hepatitis Virus Type 1
back (Figure  13.2). Death occurs within an hour or so Morbidity is 100% and mortality in young ducklings
after signs are noted. During the height of severe out­ infected with DHAV‐1 varies according to age. In some
breaks, ducklings can die very rapidly. The clinical signs broods less than one week old, mortality may reach 95%.
caused by all three DHAV genotypes are similar. In 1‐ to 3‐week‐old ducklings, mortality may be 50% or
less. In 4‐ to 5‐week‐old ducklings, morbidity and mor­
Duck Hepatitis Virus Type 2 tality are low or negligible (136).
The clinical course of DAstV‐1 infection is similar to that
of DHAV‐1 and can be seen in ducklings immune to Duck Hepatitis Virus Type 2 and Type 3
DHAV‐1 infection. Deaths occur within 1–4 days, usu­ Mortality due to DHV type 2 varies between 10 and
ally within 1–2 hours after the appearance of clinical 25% in 3‐ to 6‐week‐old birds, and can be up to 50% in
signs, which include polydipsia with loose droppings, 6‐ to 14‐day‐old birds (44). DHV type 3 is less severe
excessive urate excretion, and sometimes convulsions than DHV type 1 and 2, and mortality rarely exceeds
and acute opisthotonos (47). Affected ducks usually die 30% (111).
in good condition, and the time of death and mortality
rate (10–50%) depend on the age of the ducks (44).
Pathology
Survivors excrete virus for at least one week after infec­
tion (47) and rear normally, with little evidence of Gross
retarded growth (44). Mature ducks are refractory to the Duck Hepatitis Virus Type 1.  Principal lesions due to
disease (44). DHAV are found in the liver, which is enlarged and
displays distinct punctuate or ecchymotic hemorrhagic
Duck Hepatitis Virus Type 3 foci (Figure  13.3). Frequent reddish discoloration or
Ducklings dying from DAstV‐2 infection show the typi­ mottling of the liver surface is seen. The spleen is
cal clinical signs of DHV type 1 infection such as out­ sometimes enlarged and mottled. In numerous cases,
stretched legs and opisthotonos. Mortality rarely exceeds the kidneys are swollen and renal blood vessels
30%, but gross pathologic changes are similar to those congested. The lesions caused by all three DHAV
caused by DHV type 1 (111). genotypes are similar.
456 Section II  Viral Diseases

Figure 13.3  Enlarged liver with diffuse petechial and ecchymotic


hemorrhages caused by duck hepatitis A virus (DHAV) type 1
infection. Figure 13.5  Microscopic lesions in liver of duckling dead from
infection with duck hepatitis A virus (DHAV) type 1. Note chronic
lesions show extensive bile duct proliferation.

muscle of ducks infected with DHAV‐1 by EM. The


spleen showed regressive changes at 6 hours PI and
became necrotic by 24 hours. There were degenerative
changes in the nuclei of plasma cells that may have been
caused by the virus. Virus particles were not identified.
Only slight changes were seen in muscles.

Duck Hepatitis Virus Type 2 and 3.  Lesions of DAstV‐1 and


DAstV‐2 infections are similar to those of DHAV infection.
Microscopic changes in the acute case are characterized by
extensive necrosis of the hepatocyte cytoplasm and bile
duct hyperplasia is widespread.

Pathogenesis of the Infectious Process


Figure 13.4  Microscopic lesions in liver of duckling dead from
infection with duck hepatitis A virus (DHAV) type 1. Note acute Duck hepatitis A virus infection causes hepatic necrosis
lesions show massive hepatocyte necrosis and hemorrhage. and apoptosis, resulting in liver injury, which plays an
important role in disease pathogenesis (102). Ahmed
Duck Hepatitis Virus Type 2 and Type 3.  Lesions caused by et al. (3) reported that, in clinical cases of DHAV‐1 infec­
all three types of DHV are similar. Occasionally, small tion, there were lower serum levels of total protein and
hemorrhages are seen in the intestinal wall and on the albumen, and elevated levels of alkaline phosphatase,
heart fat of DAstV‐1‐infected ducklings (44). glutamic pyruvic transaminase (GPT), bilirubin, and
creatinine. The serum levels of GPT and glutamic
Microscopic and Ultrastructural oxaloacetic transaminase were increased in relation to
Duck Hepatitis Virus Type 1.  Microscopic changes in severity of infection (84).
DHAV infection consist of necrosis of hepatic cells and Song et al. (104) reported that the main target organs of
varying degrees of inflammatory cell infiltration and both a virulent (SH) and an attenuated (FC64) strain of
hemorrhage (Figure 13.4); survivors with more chronic DHAV‐1 were liver, spleen, and intestine. Infection with
lesions have shown regeneration of liver parenchyma the SH strain was lethal to the 1‐day‐old ducklings at
and widespread bile duct hyperplasia (Figure 13.5) (33). 36  hours PI, and apoptosis and visible lesions were
Peng showed by EM that DHAV invades many tissues in demonstrated in the liver. Interferon‐gamma (IFN‐γ),
­
the duckling and causes swelling, hemorrhage, and interleukin 2 (IL‐2), inducible nitric oxide synthase
necrosis of the liver, spleen, kidneys, and pancreas. (iNOS), and nitric oxide (NO) production were
Pathological changes have also been seen in the central strongly  upregulated by SH infection, which may
nervous system and the bursa of Fabricius in infected ­contribute to the ­pathogenicity of SH. However, the FC64
ducklings (87). Adamiker (1) examined spleen and strain was  nonpathogenic. The induction of type I IFNs,
Chapter 13  Viral Infections of Waterfowl 457

IFN‐­stimulated genes, and IL‐6 by FC64 infection was ●● Inoculation, SC or IM, of the isolate into 1‐ to 7‐day‐
several hundred‐fold greater than that of SH infection. old DHAV susceptible ducklings. The characteristic
The intensive induction of cytokines by FC64 may be clinical disease should follow, with deaths often occur­
involved in restriction of virus replication and stimula­ ring within 24 hours. Ducklings should show the gross
tion of ­adaptive immune responses. pathology attributable to DHAV. The virus should be
reisolated from the livers to confirm the diagnosis.
Immunity (Active, Passive) ●● Inoculation of serial dilutions of the liver homogenate
into the allantoic sacs of embryonating duck eggs (aged
Duck Hepatitis Virus Type 1 10–14 days) from a DHAV‐free flock, or chicken eggs
Recovery from DHAV‐1 infection results in solid immu­ (aged 8–10 days). DHAV‐infected duck embryos
nity and neutralizing antibodies in serum. Active immu­ should die within 24–72 hours; chicken embryos are
nity can be induced in adult ducks by injection of certain more variable and erratic in their response and usually
strains of the virus (5). Some strains require repeated take 5–8 days to die. The allantoic fluid is opalescent
injections to obtain high levels of antibody (85). Virus‐ or a pale greenish‐yellow. Gross pathological changes
neutralizing activity was revealed in both immunoglobu­ in the embryos include stunting and SC hemorrhages
lin M (IgM) and IgG classes of sera of actively‐immunized over the whole body, with edema, particularly of the
ducks (112). Davis and Hannant (26) reported that neu­ abdominal and hind limb regions. The embryo livers
tralizing antibody was present 4 days post vaccination of may be swollen, red, and yellowish in color, and show
2‐day‐old ducklings. Song et  al. (104) reported that in necrotic foci. The liver lesions and embryo stunting
ducklings inoculated with an attenuated strain of become more apparent in embryos that take longer to
DHAV‐1, high levels of neutralizing antibodies were die. Histological changes include: proliferation of
produced and maintained for 45 days. granulocytes in various organs, focal necrosis of the
Passive immunity can be conferred to ducklings by liver, bile duct hyperplasia, and SC edema. Inclusion
injection of serum from recovered or immunized birds. bodies should not be found (35).
Passive antibodies may also be transferred through yolk ●● Inoculation of primary cultures of DEL cells (127). Serial
to hatched ducklings to protect them. Toth and Norcross dilutions of the liver homogenate containing DHAV‐1
(112) have shown that it is IgG, and not IgM, that is trans­ should cause a CPE that is characterized by cell round­
ferred from the dam to the newly hatched ducklings. ing and necrosis. When overlaid with a maintenance
medium containing 1% agarose (wt/vol), the CPE gives
Duck Hepatitis Virus Type 2 and 3 rise to plaques approximately 1 mm in diameter.
Gough and Stuart (47) found that survivors of DAstV‐1
infection were immune to further infection. Detectable A rapid and accurate diagnosis of DHAV‐1 can be
antibody levels following infection were shown to be low, made using the direct fluorescent antibody (FA) tech­
using a virus‐constant serum neutralization test in nique on livers of naturally occurring cases or inoculated
embryonating chicken eggs (42). An active immune duck embryos (82, 117). Virus isolation and identifica­
response to DAstV‐2 infection can be stimulated in adult tion procedures have been reviewed (51, 110).
ducks by inoculation of attenuated virus. This immunity
may be passively transferred via the yolk to progeny. Duck Hepatitis Virus Type 2
Traditionally the most reliable diagnostic method for
DHV type 2 has been EM examination of liver homogen­
Diagnosis ates for the detection of astrovirus‐like particles. Now,
molecular diagnostic methods are more sensitive and are
Isolation and Identification widely used.
of the Causative Agent The virus can only be isolated, with difficulty, follow­
Although a presumptive diagnosis can be made on the ing repeated passage in the amniotic sac of embryonat­
basis of the characteristic disease pattern in the flock and ing chicken or duck eggs. However, these are difficult
gross pathological lesions in the liver, isolation and iden­ and expensive processes because the embryos may
tification of the causative agent are necessary to confirm respond erratically only after four passages and no deaths
the diagnosis. Liver specimens can be collected at post­ may be seen during earlier passages. Few embryos have
mortem for virus identification. been shown to die in less than seven days, but infected
embryos have appeared stunted and had greenish
Duck Hepatitis Virus Type 1 necrotic livers in which astrovirus‐like particles could be
The presence of DHAV may be confirmed by molecular demonstrated by EM (44). Inoculation of susceptible
diagnostic methods, or one or more of the following pro­ ducklings with virus gives a variable response; mortality
cedures (125, 128): up to 20% may occur within 2–4 days PI. (44, 125, 128).
458 Section II  Viral Diseases

Duck Hepatitis Virus Type 3 This assay was considerably more sensitive than assays
Duck hepatitis virus type 3 may be tentatively identified in eggs. A microneutralization assay in DEK cells has
by inoculation of liver suspension onto the CAM of 10‐ been developed as a more practical, rapid, and econom­
day‐old embryonating duck eggs if embryo lesions and ical alternative to other tests for the diagnosis of
mortality patterns develop as above‐described (125, DHAV‐1 (62). This assay has been adapted to monitor
128). Alternatively, virus may be isolated and identified the serum neutralization antibody responses of ducks
in DK or DEK cultures examined by immunofluores­ to vaccines in field and laboratory trials, and a titer of
cence 48–72 hours PI using DAstV‐2‐specific antibody. less than 4 log2 is considered to be negative (129).
A direct FA test in duckling livers and DEK or DK cells An indirect ELISA using the whole DHAV virus parti­
has been described for DAstV‐2 (125, 128). cle as a coating antigen has been shown comparable to
the neutralization test (141). Recently, indirect ELISAs
using the VP1 or VP3 proteins of DHAV‐1 as coating
Molecular Diagnostic Methods
antigens have also been described (77, 101). For reviews
Several reverse transcriptase‐polymerase chain reac­ of other serologic tests such as the passive hemagglutina­
tions (RT‐PCR) have been developed and are useful for tion (HA) test, indirect HA test, and agar gel diffusion
identifying DHAV‐1 infections (22, 38, 66). Chen et  al. precipitin test, please see the previous edition of this
(22) reported the detection limit of RT‐PCR DHAV‐1 textbook (136).
RNA as 3 pg/10 µl. They also demonstrated that RT‐PCR
was the most sensitive when the detection rates were Duck Hepatitis Virus Type 2
compared on 185 clinically suspected DHAV‐1‐infected An indirect ELISA has been developed based on the
liver tissues by RT‐PCR, enzyme‐linked immunosorbent expressed C‐terminal ORF2 protein of DAstV‐1 (121).
assay (ELISA), and virus isolation methods.
Reverse transcriptase‐loop‐mediated isothermal ampli­
Differential Diagnosis
fication (RT‐LAMP) assays for the detection of DHAV
have also been developed, and it has been shown that the The sudden onset, rapid spread, and acute course of dis­
assay is as sensitive as RT‐PCR (72, 105). Duplex and mul­ ease caused by DHAV‐1 are characteristic. Hemorrhagic
tiplex RT‐PCR have been developed for the simultaneous lesions in the liver of ducklings up to three weeks of age
detection and differentiation of DHAV types (55, 67, 123). are pathognomonic. Since all three DHV types cause
Yang et al. (139) reported the development of a one‐step similar disease outbreaks, differential diagnosis is needed
real‐time RT‐PCR assay (rRT‐PCR) based on primers to a because immunization is serotype‐specific and does not
conserved region in the 3D gene for the detection of confer protection against infection with heterologous
DHAV‐1. The detection limit of this assay was 10 viral type and serotypes.
genomic copies per reaction. An astrovirus‐specific RT‐ Possible synergistic effects of DHAV‐1 with
PCR assay based on the ORF1b region of astroviruses Chlamydophyla psittaci (15) and influenza virus (48)
(109) has been reported for use to detect DAstV‐1, have been suggested. Other potential causes of acute
DAstV‐2 and other duck astroviruses. Multiplex RT‐PCR mortality in ducklings include avian influenza, duck viral
has been developed for the simultaneous detection of enteritis, Pasteurella anatipestifer infection, salmonello­
DHAV‐1, DHAV‐3, and DAstV‐1 (16). sis, coccidiosis, and aflatoxicosis. Aflatoxicosis may
cause ataxia, convulsions, and opisthotonos as well as
microscopic lesions of the bile duct with hyperplasia
Serology
suggestive of DHV, but does not cause the same charac­
Duck Hepatitis Virus Type 1 teristic liver hemorrhages.
Due to the short course of the disease, serologic tests
have not been useful in diagnosing acute outbreaks of
DHAV‐1. However, the neutralization test is useful for
virus identification, titration of serologic response to
Intervention Strategies
vaccination, and epidemiologic surveys.
Management Procedures
Both in vivo and in vitro neutralization tests have
been developed for DHV type 1. Hwang (57) first Duck hepatitis virus type 1 can be prevented by strict
described an accurate, reproducible DHAV‐1 neutrali­ isolation, particularly during the first 4–5 weeks of age.
zation test in chicken embryos. Modifications of this In areas where the disease is prevalent, however, it is very
procedure have been described (46, 112), including difficult to obtain the necessary degree of isolation.
those adapted to duck embryos or ducklings (51) and Contact with wild waterfowl should be prevented and,
tissue culture (41). Woolcock et al. (135) first described since rats may act as a reservoir host of the virus, pest
a plaque‐reduction test for neutralization antibody. control is needed. Panikar (86) as well as Kaszanyitzky
Chapter 13  Viral Infections of Waterfowl 459

and Tanyi (63) demonstrated the feasibility of eradicat­ The application of inactivated vaccines has also been
ing DHAV‐1 in selected areas where isolation can be investigated. Multiple inoculations of inactivated vac­
achieved. In both studies, vaccination of breeder ducks cine are needed to provide passive immunity to progeny
was used as part of the program. for a complete laying cycle. A single inoculation of inac­
tivated vaccine given to breeder ducks, which has been
previously primed with live DHAV‐1, at the time before
Vaccination
the birds come into lay, also provides passive immunity
Type of Vaccine to progeny ducklings for a complete laying cycle (46,
Duck Hepatitis Virus Type 1.  Resistance against DHAV‐1 129). For example, Gough and Spackman (46) reported
may be conferred to ducklings by three methods: that effective levels of duckling protection can be secured
injection of immune serum or yolk, as described under by administering three doses of inactivated, oil‐emulsion
Treatment (see below); immunization of breeding stock vaccine. They also reported that live DHAV‐1 vaccine
to ensure high levels of passively transferred antibody in administered at 2–3 days of age, followed by inactivated
the hatched ducklings; and direct active immunization of vaccine at 22 weeks, producing significantly higher
ducklings with live avirulent strains of DHAV‐1. virus‐neutralizing (VN) antibody levels than did three
Attenuated DHAV‐1 strains suitable for vaccine use doses of inactivated vaccine. Finally, they reported that
have been produced by passage in chicken embryos (5, inactivated vaccine prepared from virus grown in duck
39, 40, 99) or duck embryos (10, 96). Up to this time, eggs gave a better antibody response than virus grown in
various strains of chicken embryo‐passaged DHAV‐1 chicken eggs. Woolcock (129) also investigated the use of
have been used most frequently as vaccines. Davis (25) inactivated DHAV‐1 vaccines in breeder ducks. He
reported that triple plaque‐purified strains of DHAV‐1 showed that ducks primed with modified live virus at
used for vaccines could revert to virulence as readily as 12 weeks of age, and boosted with inactivated DHAV‐1
non‐cloned virus. This finding has been confirmed with vaccine at 18 weeks of age, developed VN antibody titers
various egg‐passaged levels of DHAV‐1 (131). Davis that were 16‐fold higher than those in ducks that received
­suggested rapid passage as a method to increase genetic only the MLV priming. This level of immunity was suf­
stability. ficient to protect ducklings hatched through a complete
Recently, live attenuated DHAV‐3 vaccine has been laying cycle (eight months), as demonstrated by chal­
developed in South Korea (64). Also, a live attenuated lenging progeny with virulent DHAV‐1. He also showed
duck enteritis virus vector vaccine, which was designed that only ducks given multiple inoculations of inacti­
to express VP1 of both DHAV‐1 and DHAV‐3, has shown vated vaccine developed titers of 6 log2 or greater, which
protection to challenge of virulent strains of DHAV‐1 was considered the minimum protective level.
and DHAV‐3 (142).
Ducklings.  Chicken embryo‐attenuated strains of
Duck Hepatitis Virus Type 2 and  Type 3.  An experimental DHAV‐1 have been shown to induce a considerable
DAstV‐1 live attenuated virus vaccine has been shown to degree of protection in one‐day‐old ducklings inoculated
protect ducklings from challenge with virulent virus, but by the SC, IM, intranasal (IN), or foot‐web route (5, 23,
this vaccine has never been produced commercially (47). 40, 93, 143). Effective mass vaccination of ducklings by
Also, a DAstV‐2 live attenuated vaccine has been used aerosol and drinking water routes have also been
experimentally in breeder ducks to confer passive reported (54, 69, 85, 86). However, Sung et  al. (106)
immunity to ducklings, but this vaccine has not been reported that IM administration of attenuated DHAV‐1
available commercially (136). vaccine showed more efficient protection than by the
oral or eye drop route of administration. Newly hatched
Field Vaccination Protocols and Regimes ducklings injected intramuscularly with an attenuated
Breeders.  It is generally suggested that two or three DHAV‐1 rapidly developed resistance in 3 days (59).
doses of attenuated virus vaccine be given in order to Oral exposure required up to six days for protection to
achieve satisfactory levels of protection of progeny (30, occur. There was evidence that vaccination would be of
59, 62, 93). Rispens (96) recommended that two doses benefit even at the start of an outbreak.
of attenuated virus vaccine be administered to breeders Results from studies on the effects of maternal anti­
at least six weeks apart; passive immunity was bodies on vaccination of ducklings with live attenuated
transmitted to progeny for about nine months after the DHAV‐1 have not been consistent (9, 79). However, field
second vaccination. Woolcock (130) recommended experience has indicated that successful practical duck­
administering the attenuated virus vaccine ling vaccination depends on the absence of maternal
subcutaneously at the neck of breeder ducks at 16, 20, antibodies, and is influenced by time and severity of
and 24 weeks of age and every 12 weeks thereafter, exposure to virulent virus. Vaccination is also less effec­
throughout the laying period. tive when ducklings are exposed to virulent virus early in
460 Section II  Viral Diseases

life, especially in endemic areas and on heavily infected breeder ducks. At the DRL, Long Island, New York, this
premises. Judicious application of good hygiene and san­ procedure has been modified by substituting yolk from
itation methods can help with this problem (136). eggs produced by specific pathogen free chicken hyper­
Kim et  al. (64) reported on the development of a immunized with DHAV‐1. For immunization, yolk anti­
chicken embryo‐attenuated strain of DHAV‐3. One‐day‐ body preparations with a minimum neutralizing index of
old ducklings vaccinated intramuscularly with this virus 103, as determined by the constant‐yolk/varying–virus
were fully protected from challenge with pathogenic method, would be considered to be satisfactory (125).
virus at 2–3 days post vaccination. Recently, Chinese herb medicine has been studied
with respect to treatment and effect on DHAV‐1. Bush
Sophora Root polysaccharide and its sulfate (19–21),
Treatment
astragalus polysaccharide and its sulfate (18), icariin and
Duck Hepatitis Virus Type 1 its phosphorylated structural modification (137), and a
As soon as the cause and nature of DHAV‐1 infection flavone/polysaccharide‐containing prescription drug
were recognized (70), it became apparent that ducklings (31) have shown some antiviral activities against DHAV.
might be protected by administration of serum from Also, phosphorylated Codonopsis pilosula polysaccha­
immune ducks. This procedure proved to be highly suc­ ride has been shown to reduce the replication of DHAV‐1
cessful in laboratory experiments and in the field. For in duck embryonic hepatocytes, probably through a
many years, the Duck Research Laboratory (DRL) at reduction in expression of IFN‐β (83).
Cornell University at Eastport, Long Island, New York,
kept a bank of antiserum processed from blood collected Duck Hepatitis Virus Type 2 and Type 3
at the time of slaughter from recovered birds. Inoculation of susceptible ducklings with convalescent
Intramuscular injection of 0.5 ml DHAV‐1 antiserum serum obtained from DHV type 2‐infected ducks has
into each duckling of a brood at the time of the first been used successfully to control the disease in the field
deaths in an outbreak was an effective control method. (47). Also, convalescent sera obtained from DHV type
Rispens (96) has suggested passive immunization by 3‐infected ducks have been used effectively in the field to
injection of yolk from eggs produced by hyperimmune control outbreaks.

­Duck Virus Enteritis (Duck Plague)


Samia A. Metwally and Anchun Cheng

Summary Introduction
Agent, Infection, and Disease.  Duck virus enteritis (DVE), Definition and Synonyms
also called duck plague, is one of the major contagious
Duck virus enteritis (DVE) is an acute, contagious her­
and fatal diseases of ducks, geese, and swans. The agent
pesvirus infection of ducks, geese, and swans, character­
can spread via horizontal and/or vertical transmission
ized by vascular damage, tissue hemorrhages, digestive
and causes death, vascular damage, and subsequent
mucosal eruptions, lesions of lymphoid organs, and
internal hemorrhage, lesions in lymphoid organs,
degenerative changes in parenchymatous organs.
digestive mucosal eruptions, severe diarrhea and
Synonyms for the disease are duck plague, eendenpest
degenerative lesions in parenchymatous organs of
(Dutch), peste du canard (French), Entenpest (German),
infected birds. Huge economic losses are caused by high
and duck virus enteritis. Although Bos (5) first used the
morbidity and mortality, decreased egg production, and
term duck plague, it was proposed as the official name by
hatchability. Duck virus enteritis is worldwide in
Jansen and Kunst in 1949 (29). Subsequently, DVE, based
distribution.
on principal features of the disease and to distinguish it
from fowl plague, has become the preferred term.
Diagnosis.  The diagnosis of DVE includes virus
isolation, serological, and molecular tests in combination
with clinical manifestations and histopathology. Economic Significance
In duck‐producing areas of the world where the disease
Intervention.  Vaccination of live‐attenuated or inactivated has been reported, DVE has produced significant eco­
vaccines can prevent disease in broiler and breeder nomic losses in domestic and wild waterfowl due to mor­
ducks. tality, condemnations, and decreased egg production.
Chapter 13  Viral Infections of Waterfowl 461

The first outbreak in the United States was in 1967 and ­ iameter of 150–300 nm and located in cytoplasmic vac­
d
caused losses in excess of $1 million during a one‐year uoles (Figure 13.6).
period for the small, but concentrated, duck industry of Study on the morphogenesis of DEV in infected DEF
Long Island, New York (42). cells demonstrated that the attached DEV probably
­penetrated the cell membrane by direct fusion between
the viral envelop and the plasma membrane. Progeny
Public Health Significance
nucleocapsids assembled in the nucleus and exited from
Duck virus enteritis is primarily a disease of waterfowl. this compartment into the cytoplasm by budding
No known risk to human health has been reported. through the inner nuclear leaflet into the interconnected
perinuclear cisterna and cisternae of endoplasmic retic­
ulum. DEV tegumentation occurred in cisternae of
History endoplasmic reticulum and the tegumented progeny
nucleocapsids obtained their final envelop by budding
In 1923, Baudet (4) reported an outbreak of an acute, into cytoplasmic vesicles. Intravesicular mature DEV
hemorrhagic disease of domestic ducks in the particles were discharged from the cell through exocyto­
Netherlands. Bos (5) suggested that the disease was sis of the cytoplasmic vesicles by cells, or through rup­
caused by a new distinct viral disease of ducks, which he ture of the virus‐containing vesicles (20). Intracytoplasmic
termed “duck plague.” These observations were further inclusion bodies and intranuclear inclusion bodies could
supported by more detailed studies on virus propaga­ be observed respectively in the cytoplasm and nucleus of
tion, incidence and distribution, pathology, and immu­ infected DEF. With the appearance of progeny DEV in
nity (28, 67) DEF, some densely electron‐stained, virus‐related struc­
After that, serious outbreaks in migratory waterfowl, tures which were rod‐shaped, U‐shaped, or of circle,
zoos, and game farm flocks have been reported (17). The semicircle, or concentric circle in appearance, could be
latest outbreak was reported in Germany (2007) in ducks observed in the cytoplasm of DEF.
and geese kept in captivity to prevent spread of highly
pathogenic avian influenza (H5N1) (34). Duck virus
enteritis was delisted from the OIE Terrestrial Animal
Health Code in 2010 due to its limited spread across
country borders.
For detailed historical details of DVE, see prior edi­
tions of this book.

Etiology
Classification
The causative agent of DVE is a herpesvirus (Anatid her­
pesvirus 1), belonging to the Mardivirus genus of
Alphaherpesvirinae subfamily (55). Duck enteritis virus
is non‐hemagglutinating and non‐hemadsorbing (13).

Morphology
Ultrastructure, Size, and Density
Ultrathin sectioning and transmission electron micros­
copy (TEM) of infected duck embryo fibroblasts (DEF)
were employed to investigate the morphology of duck
enteritis virus (DEV) (23). The nucleic acid of DEV was
round in shape with diameter of 35‐45 nm and was often
in a cluster in the nucleus of DEF. The nucleocapsid of Figure 13.6  The morphology and distribution of mature duck
DEV was round in shape with diameter of 90–100 nm enteritis virus (DEV) particles in host cells. Microscopically, liver
shows focal areas of necrosis filled with fibrin from duck virus
and could be observed both in nucleus and cytoplasm of enteritis (DVE). Intranuclear inclusion bodies can be seen in
DEF. The mature DEV which had the structures of hepatocytes near areas of necrosis. Arrow: The mature particles
envelop and tegument was spherical in shape with located in the cytoplasmic vacuoles. N: nucleus, C: cytoplasm.
462 Section II  Viral Diseases

Chemical Composition have been noted, all appear to be immunologically iden­


tical and antigenically related (66). The virus is immuno­
The virion contains DNA (33). RNase treatment on thin
logically distinct from other avian viruses, including fowl
sections had no effect on ultrastructural morphology of
plague, Newcastle disease, duck hepatitis (13), and other
the virus, and exposure to DNase led to the removal
herpesviruses (59).
of  the central core without affecting the envelope.
A herpesvirus was isolated from domestic geese in
Fluorescence of intranuclear inclusion bodies in cell
Australia showing gross pathological and histopatho­
­cultures stained with acridine orange also was consistent
logical changes similar to those seen in DVE (36). The
with the presence of DNA. Inactivation by ­pancreatic
virus isolate was antigenically and genomically dis­
lipase indicates that the virions contain an essential lipid.
tinct from DEV as shown by protection, serological
analysis, and genetic characterization by restriction
Virus Replication endonuclease digestion and polymerase chain reaction
(PCR) assays (19).
Development of the virus in cell cultures was studied by
electron microscopy (EM) and growth curves of intracel­
Genetic or Molecular Characteristics
lular and extracellular virus (21, 22, 70). Examination of
Duck enteritis virus is currently grouped into the
thin sections revealed development forms only in the
Mardivirus genus of the Alphaherpesvirinae subfam­
nucleus 12 hours postinoculation (PI). By 24 hours, in
ily. Complete genomic analysis of the CHv strain of
addition to viral forms in the nucleus, larger particles
DEV demonstrated that the genome comprises of
with an envelope were observed in the cytoplasm. Virus
162,175 nucleotides encoding 78 putative proteins
titrations of similar cell cultures demonstrated new cell‐
(81). Further analysis showed DEVs have a typical type
associated virus 4 hours PI, with maximum titer at 48
D herpesvirus genome arrangement pattern (UL‐IRS‐
hours. Extracellular virus was first detected 6–8 hours PI
US‐TRS) which are consistent with genomic organiza­
and reached maximum titer at 60 hours (33). Increased
tion of the members of Alphaherpesvirinae. Like most
incubation temperatures of tissue cultures (39.5–41.5°C)
members of them, the genes in the UL region are well
favored viral replication, especially of less virulent strains
conserved, while the genomic arrangement of IRS‐US
(7). Viral glycoprotein C plays an active role in the adsorp­
is similar to that of Marek’s disease virus and equine
tion of DEV over DEF to enhance the infectivity and
herpesvirus 1. Moreover, comparative genomic analy­
hence blocking the gC can be a critical strategy in pre­
sis of DEVs demonstrated that, although similar to
venting the viral establishment in the host cells (25, 32).
other herpes viruses, the DEV genome also show vari­
In a susceptible host, virus replicates primarily in the
ation (83). For example, the LORF3 segment of the
mucosa of the digestive tract, especially in the esophagus,
European strain (2085) was 33 bp shorter than that of
and then spreads to the cloacal bursa, thymus, spleen, and
Asian DEV strains (CHv, VAC). A 181 amino acid
liver. The epithelial cells, lymphocytes, and macrophages
domain present in virulent strains was absent in atten­
of these organs are the principal sites of viral replication
uated strain.
(60, 86). Since DEV can replicate quickly in many cell
types and tissues, it is considered as a pantropic virus that
leads to pathological lesions in many different organs (54). Laboratory Host Systems
Duck enteritis virus can be propagated in duck embryo
Susceptibility to Chemical and Physical fibroblasts, duck embryo liver or kidney primary cells,
Agents and the chorioallantoic membrane (CAM) of 9‐ to 14‐
The virus was found to be sensitive to ether and chloro­ day‐old embryonating duck eggs. The virus can be
form. Exposing virus for 18 hours at 37°C to trypsin, adapted to grow in embryonating chicken eggs and
­chymotrypsin, and pancreatic lipase markedly reduced chicken embryo cell cultures (13). Moreover, continu­
or inactivated it, and papain, lysozyme, cellulase, DNase, ous cell line CCL‐141 and Vero cell were also demon­
and RNase had no effect. Besides, heating, calcium strated suitable for DEV growth in recent years (2, 53).
­chloride drying and exposure to extreme pH resulted in However, they are unsatisfactory for primary isolation.
rapid inactivation (50). The virus produces a cytopathogenic effect in inocu­
For susceptibility details of DEV to chemical and lated cell cultures (13), and intranuclear inclusions
­physical agents, see prior editions of this book. have been observed in infected chicken and duck
embryo cell cultures (24) (Figure  13.7). Plaque assays
have been used to measure virus and neutralizing
Strain Classification
­antibody titers (13). In the presence of complement,
Antigenicity, Immunogenicity, or Protective Characteristics antibodies to DEV are capable of lysing infected duck
Although differences in virulence among DEV strains embryo fibroblasts (38).
Chapter 13  Viral Infections of Waterfowl 463

(A) (B)

1000×

Figure 13.7  Inclusion bodies in duck embryo fibroblasts infected with duck virus enteritis (DVE). (A) ×1000. (B) ×310,000.

Pathobiology and Epizootiology In addition to domesticated species, mallards (A.


platyrhynchos), Garganey teal (A. quer quedula), gad­
Incidence and Distribution wall (A. strepera), European widgeon (A. penelope),
wood ducks (Aix sponsa), shovelers (Spatula clypeata),
In addition to the Netherlands, DVE has been reported common pochards (Aythya ferina), common eiders
in China and confirmed in France, Belgium, India, (Somateria mollissima), white‐fronted geese (Anser
Thailand, England, Canada, Hungary, Denmark, Austria, albifrons), bean geese (A. fabalis), and mute swans
Viet Nam, Germany (34), Egypt (16), Poland (80), and (Cygnus olor) were susceptible to lethal infection. The
Bangladesh (1). Epidemiological investigations suggested first reported outbreaks of spontaneous DVE in wild
an association between the incidence of DVE in wild waterfowl on Long Island, New York (42) confirmed
birds and farmed poultry as samples from wild birds and DEV present in mallards, black ducks (A. rubripes), a
poultry were found to be positive/carrier, by various Canada goose (Branta canadensis), a bufflehead
virological and serological investigations. Higher flock (Bucephala albeola), a greater scaup (Aythya marila),
density is usually the main contributing factor for initia­ and a mute swan. Mallards were more resistant to lethal
tion of DVE outbreaks. Reports demonstrated that DVE infections and were considered a possible natural reser­
had a worldwide distribution that regularly occurred voir of infection (79).
throughout the year except in August and September, European teal (A. crecca) and pintails (A. acuta) were
and approximately 86% of these outbreaks have been resistant but produced antibodies against DEV as a
reported from March to June due to the physiological result of experimental exposure, but herring gulls
changes and breeding during spring season (15). (Larus argentatus) and black‐headed gulls (L. ridibun-
dus) of the order Charadriiformes were not susceptible
to experimental infection and failed to produce anti­
Natural and Experimental Hosts
bodies against DVE (74).An experimental study (78)
Although the virus can be adapted by serial passage to grow showed blue‐winged teal (A. discors) and Canada geese
in embryonating chicken eggs and chickens up to two weeks were extremely susceptible to DEV and experienced
of age (41), natural susceptibility to DVE has been limited to high mortalities. Blue‐winged teal had few gross lesions
members of the family Anatidae (ducks, geese, and swans) at necropsy.
of the order Anseriformes. Naturally occurring outbreaks A study on susceptibility of waterfowl to Lake Andes
have occurred in a variety of domestic ducks (Anas platy- strain of DEV showed that blue‐winged teals, wood
rhynchos) including white Pekin, Khaki Campbell, Muscovy ducks, and redheads were highly susceptible; Muscovy
ducks (Cairina moschata) (43), Indian runner, hybrids, and ducks and gadwalls were moderately susceptible; mal­
native ducks of mixed breeding, domestic geese (Anser lards and Canada geese were less susceptible; and pin­
anser), and mute swans (37) ranging from seven days of age tails were the least susceptible (17, 66). An outbreak in
to mature breeders. Gray call ducks have been found to be captive ducks and geese reported in Germany in 2005
resistant to lethal infections. Outbreaks of DVE in domestic identified 14 additional susceptible Anseriform species
ducks are frequently associated with aquatic environments that were collected by a hobbyist from different conti­
cohabited by wild waterfowl (43). nents (34).
464 Section II  Viral Diseases

Transmission, Carriers, and Vectors outstretched wings and head down suggesting weakness
and depression. Sick ducks forced to move may show
Duck virus enteritis can be transmitted by direct contact
tremors of head, neck, and body. Young ducklings 2–7
between infected and susceptible birds or indirectly by
weeks of age show dehydration, loss of weight, blue
contact with a contaminated environment. As the water­
beaks, conjunctivitis, lacrimation, nasal exudate, and
fowl is dependent on an aquatic environment, transmis­
often a blood‐stained vent.
sion through water seems to be a prime source. New
outbreaks of DVE can be prevented after limiting the
access of domestic ducks to open water bodies that are Morbidity and Mortality
cohabited by free‐flying waterfowl. However, once the
Total mortality in domestic ducks may range from
infection has been established, they may become carriers
5%–100%. Because the birds showing clinical signs usu­
and shed DEV into the environment, causing new foci of
ally die, morbidity closely approaches mortality. Adult
infection by movement into susceptible flocks or onto
breeder ducks tend to experience higher mortality than
bodies of water previously free of virus contamination.
young ducks. No differences in mortality rates are found
Experimentally, DVE can be transmitted via oral, intra­
in mallard and white Pekin ducks experimentally infected
nasal, intravenous, intraperitoneal, intramuscular, and
with DVE and Riemerella anatipestifer, indicating that
cloacal routes. Horizontal spread is the principal mode
these pathogens do not act synergistically (52). However,
of transmission, while vertical transmission has been
mallards immunosuppressed with cyclophosphamide
reported in persistently infected waterfowl (15).
and challenged with a sublethal dose of DVE may
The course and direction of the infection are depend­
increase mortality rate (18). Secondary bacterial infec­
ent on population density as well as rate of transmission
tions with Pasteurella multocida, R. anatipestifer, and E.
between infected and susceptible birds. Recovered birds
coli are often seen in a natural outbreak of a low virulent
become carriers and shed the virus periodically (9). Like
strain in young ducklings as a result of the immunosup­
other herpesviruses, DEV latency and reactivation have
pressive effect of the virus (61).
been blamed for precipitating outbreaks in domestic and
migrating waterfowl populations. The trigeminal gan­
glion, lymphoid tissues, and peripheral blood lympho­ Pathology
cytes have been shown to be the latency site for the virus
The specific pathologic response to DVE depends on
postinfection with DEV (62). According to the reports,
species affected (41), age, sex, stage of infection, and vir­
the US2 protein of DEV plays an active role in penetrat­
ulence and intensity of virus exposure (43).
ing the susceptible host cell and subsequent spread of
virus from one cell to another cell in the host and facili­
Gross
tates the establishment of DEV infection in susceptible
Lesions of DVE are associated with disseminated intra­
birds (76).
vascular coagulopathy and necrotic degenerative changes
in mucosa and submucosa of gastrointestinal tract in
Incubation Period lymphoid and parenchymatous organs. These collective
lesions, when present, are diagnostic of DVE.
In domestic ducks, the incubation period ranges from Petechial, ecchymotic, or larger extravasations of
3–7 days. After overt signs appear, death usually follows blood may be found on or in the myocardium and other
within 1–5 days. visceral organs and their supporting structures, includ­
ing the mesentery and serous membranes. On the epi­
cardium, especially within coronary grooves, closely
Clinical Signs
packed petechiae give the surface a red “paintbrush”
In domestic breeder ducks, sudden, high, persistent flock appearance (Figure  13.8). The latter lesion is observed
mortality is often the first observation. Mature ducks die more frequently in mature breeder ducks than in young
in good flesh. Prolapse of the penis may be evident in ducklings. When heart chambers are exposed, endocar­
dead mature males. In laying flocks, a marked drop in dial mural and valvular hemorrhages also may be
egg production may be noted during the period of high­ observed. Surfaces of liver, pancreas, intestine, lungs,
est mortality. and kidney may be covered with petechiae. In mature
As infection progresses within a flock, more signs are laying females, hemorrhages may be observed in
observed. Photophobia, associated with half‐closed deformed, discolored ovarian follicles, and massive hem­
pasted eyelids, loss of appetite, extreme thirst, droopi­ orrhage from the ovary may fill the abdominal cavity.
ness, ataxia, ruffled feathers, nasal discharge, soiled Lumina of intestines and gizzard are often filled with
vents, and watery diarrhea appear. Affected ducks are blood. The esophageal–proventricular sphincter appears
unable to stand; they maintain a posture with drooping as a hemorrhagic ring.
Chapter 13  Viral Infections of Waterfowl 465

Figure 13.10  Extensive hemorrhages lesions of bursa of Fabricius


with duck enteritis virus (DEV).
Figure 13.8  Petechial hemorrhages in the epicardium and
coronary heart fat of duck infected with duck virus enteritis (DVE)
virus.
openings of sublingual salivary gland ducts in chroni­
cally infected waterfowl (6). Meckel’s diverticulum may
be hemorrhagic and contain a fibrinous core (57).
In ceca, macular lesions are singular, separated, and
well‐defined between mucosal folds. The external sur­
face of affected ceca often presents a barred, congested
appearance. Rectal lesions are usually few in number
with greatest concentration at the posterior portion of
the rectum, adjacent to the cloaca. In the cloaca, macular
lesions are densely packed; initially, the entire mucosa
appears reddened. Later, individual plaque‐like eleva­
tions become green and form a continuous scale‐like
band lining the lumen of the organ.
All lymphoid organs are affected. The spleen tends to
be normal or smaller in size, dark, and mottled. The
bursa of Fabricius is intensely reddened during early
Figure 13.9  Petechial hemorrhages and ulceration of esophageal
mucosa from duck infected with duck enteritis virus (DEV) virus. infection (Figure  13.10). The exterior becomes sur­
Note the lack of inflammatory response and presence of rounded by clear, yellow fluid that discolors adjacent
intranuclear inclusion bodies. tissue of the pelvic cavity. When the lumen of the bursa
is opened, pinpoint yellow areas are found in an
Specific digestive mucosal lesions are found in the oral intensely hemorrhagic surface. Later, walls of the bursa
cavity, esophagus, ceca, rectum, and cloaca. Each of become thin and dark, and the bursal lumen is filled
these lesions undergoes progressive alterations during with white coagulated exudates. Intestinal annular
the course of the disease. Initially, macular surface hem­ bands appear as intensely reddened rings visible from
orrhages appear, which are later covered by elevated, yel­ external and internal surfaces. Yellow pinpoint areas
low‐white crusty plaques. Subsequently, the lesion can be observed on the mucosal surface. Later, the
becomes organized into a green superficial scab devoid entire band becomes dark brown and tends to separate
of its former hemorrhagic base. Lesions range in size at its margins from the mucosal surface. The multifocal
from approximately 1–10 mm in length. In the esopha­ necrosis of gut‐associated lymphoid tissue causes
gus and cloaca, lesions may become confluent; however, ulceration covered by fibrinous pseudomembranes
close inspection will often reveal their composite struc­ (Figure 13.11). The thymus is atrophied and has multi­
ture. In the esophagus, macules occur parallel to longitu­ ple petechiae (Figure 13.12) and necrotic focal areas on
dinal folds. When macular concentrations are numerous, the surface and cut section and is surrounded by clear,
small lesions may merge to form larger ones covered yellow fluid that infiltrates and discolors subcutaneous
with a patchy diphtheritic membrane (Figure  13.9). In tissues of the adjacent cervical region from the thoracic
young ducklings, individual lesions in the esophagus are inlet to the upper third of the neck. The latter lesion is
less frequent; sloughing of the entire mucosa is more of importance in meat inspection and is easily detected
common, and the lumen becomes lined with a thick yel­ when the opened neck of the carcass is observed on the
low‐white membrane. Oral erosions can be found at processing line.
466 Section II  Viral Diseases

(A)

(B)

Figure 13.11  (A) Multifocal necrosis of gut‐associated lymphoid tissue resulting in ulceration covered by fibrinous pseudomembranes of
duck enteritis virus (DEV). (B) Also note the reddened ring visible on the external surface of the intestine.

During early stages of infection, the entire liver surface and thymus, tissue hemorrhages and reproductive tract
has a pale copper color with an admixture of irregularly lesions predominate.
distributed pinpoint hemorrhages and white foci In geese, intestinal lymphoid disks are analogous to
(Figure  13.13), giving it a heterogeneous, speckled annular bands in ducks. In a single Canada goose, lesions
appearance. Late stages of infection are characterized by of the intestinal lymphoid disks resembled “button‐like
dark bronze or bile‐stained livers without hemorrhages; ulcers” (39). Similar intestinal lesions have been observed
the white foci are larger and appear more distinct on the in an outbreak of DVE in Canada and Egyptian geese. In
darker background. swans, diphtheritic esophagitis is a consistent lesion (37).
Although these lesions are consistent with DVE infec­ An outbreak caused by a low virulent strain of DVE in
tion, each age group responds distinctively. In duck­ commercial 2‐ to 6‐week‐old white Pekin ducklings
lings, tissue hemorrhages are less pronounced and ­produced atypical gross lesions, including diphtheritic
lymphoid lesions are more prominent. In mature membranes under the tongue and in nasal and infraorbi­
domestic ducks with naturally regressed cloacal bursa tal sinuses. Esophageal mucosa had a few necrotic
Chapter 13  Viral Infections of Waterfowl 467

Figure 13.12  The thymus is atrophied and has multiple petechiae.

Figure 13.13  Multiple irregular pale foci in liver with duck virus Figure 13.14  Microscopic appearance of esophageal ulcerations
enteritis (DVE). Microscopically, liver shows focal areas of necrosis of duck virus enteritis (DVE). Note the lack of inflammatory
filled with fibrin from duck virus enteritis (DEV). response and presence of intranuclear inclusion bodies.

plaques, and cloacal mucosa was covered with necrotic, Hemorrhages are especially pronounced in certain
greenish, diphtheritic membranes. No lesions were seen locations: interlobular venules of the proventriculus,
in the intestines including annular bands. Thymus and hepatic, and portal venules at the margins of liver lob­
bursa were atrophied and hemorrhagic, and the bursal ules; venules in the spaces between lung parabronchi;
lumen was filled with cheesy exudate. An experimental capillaries within intestinal villi; and star‐shaped
study showed that the bursal atrophy could last for at intralobular renal hemorrhages.
least 39 days PI; however, the thymus recovered after 10 As a result of vascular damage, affected tissues undergo
days of infection (61). progressive degenerative changes. Microscopic changes
can be found in any visceral organs including those
Microscopic ­without gross lesions.
The initial lesion occurs in the walls of blood vessels. Digestive lesions appear initially as hemorrhages of
Smaller blood vessels, venules, and capillaries, instead of capillary arcades of submucosal papillae or folds.
larger blood vessels, are more markedly involved. The Hemorrhages become larger and confluent, elevating
endothelial lining is disrupted, and connective tissue of and separating the overlying mucosa. The affected epi­
the wall becomes less compact, with visible separations at thelium above the hemorrhage becomes edematous,
points where extravasations of blood pass from the lumen necrotic, and raised into the lumen above normal adja­
through the thin ruptured wall into surrounding tissues. cent mucosal surfaces (Figure  13.14). Later, margins of
468 Section II  Viral Diseases

necrotic epithelium separate to define the borders of


elevated plaques. There is necrosis and degeneration of
stratified squamous epithelium of the esophagus and
cloaca (56). Eosinophilic intranuclear and cytoplasmic
inclusions have been seen in epithelial cells (70).
Hemorrhage from venules and capillaries fills lymphoid
tissue within intestinal annular bands or lymphoid disks
and lymphoid tissue of the esophageal–proventricular
sphincter and spleen. Lymphocytes undergo karyorrhexis
and pyknosis. Fragments of lymphocytes appear every­
where and are engulfed by phagocytes. In addition to
­cellular debris and hemorrhage within lymphoid follicles,
marked swelling of reticulum cells occurs, and their cyto­
plasm becomes subdivided and condensed into spherical
and oval pale‐staining bodies. Reticulum cells rupture Figure 13.15  Microscopically, liver shows focal areas of necrosis
filled with fibrin from duck virus enteritis (DVE). Intranuclear
and discharge their cytoplasmic contents into tissue
inclusion bodies can be seen in hepatocytes near areas of
spaces. An intranuclear inclusion body and delicate necrosis.
nuclear membrane and cell wall are the remaining ves­
tiges of reticulum cells.
Intestinal lymphoid lesions become large hemorrhagic Within necrotic foci in the liver, hepatic cords show a
infarcts. A layer of free blood separates lymphoid tissue variety of changes including detachment and disasso­
from the mucosa, which undergoes coagulation necrosis. ciation of hepatocytes from each other and their sur­
The necrotic mucosa forms a pseudomembrane higher rounding structure. A few necrotic liver cells become
than adjacent normal intestinal mucosa. swollen or subdivided and discharge their cytoplasmic
In the small intestines, sheets of epithelial cells are contents through a ruptured cell surface and are repre­
­displaced from the surface of villi, many of which are sented only by intranuclear inclusion bodies. Focal
broken and cast into the lumen. Abundant blood and areas of necrosis may be filled with fibrin (Figure 13.15).
­cellular debris fill the lumen. Similar, but more limited, changes occur in pancreas
Within the cloacal bursa, submucosal and interfollicular and kidney (40).
capillary hemorrhages are found. There is a severe deple­
tion of lymphocytes in the follicles, many of which have
Immunity
empty hollow cavities in the medulla. Corticomedullary
epithelial cells, capillary networks, and large phagocytic Active Immunity
cells containing fragmented lymphocytes form the cir­ Active immunity has been demonstrated following the
cumference around these cavities. Severe depletion of use of a modified live virus vaccine (27), inactivated tis­
lymphocytes in the follicles occurs, which is replaced by sue culture vaccine (63), and bivalent recombinant live
eosinophilic material mixed with heterophils. There are attenuated vaccine (48, 75, 85). It is assumed that both
occasional mononuclear cells that contain intranuclear humoral and cell‐mediated immunity are involved in
inclusions. Bursal epithelial cells are hypertrophied with protection (73). Field observations suggest that recov­
vacuolated cytoplasm and contain both intranuclear and ered birds are immune to re‐infection with DEV.
intracytoplasmic inclusions (61).
In the thymus, free blood fills interfollicular spaces. Passive Immunity
Coagulation necrosis of central medullary reticulum Maternal immunity has been reported in ducklings that
cells and destruction of cortical lymphocytes are declines fast and may interfere with response to live
pronounced. virus vaccines. Ducklings from those breeder ducks
In mature female breeder ducks, congestive, hemor­ that are vaccinated with a live‐attenuated vaccine are
rhagic, and necrotic alterations occur in the oviduct. fully susceptible. However, ducklings from breeders
Follicles may be misshapen and blood stained. In the that had been vaccinated and challenged with a virulent
ovary of immature female breeder ducks, focal intestinal virus were found protected at 4 days of age, and less
hemorrhages from capillaries and venules may be found. than 40% were protected at 13 days of age (72). In an
In mature breeder drakes, focal capillary hemorrhages experimental study (8), superinfection of persistently
occur in interstitial tissues between seminiferous tubules. infected mallard ducks resulted in death, indicating
In parenchymatous organs such as liver, pancreas, and that protection depends on the route of exposure, strain
kidneys, hemorrhages and focal necrosis are found sur­ leading to persistent infection, and strain of superin­
rounding blood vessels. fecting virus.
Chapter 13  Viral Infections of Waterfowl 469

Diagnosis and tissues from esophagus, liver, kidney, and spleen. By


comparison, qRT‐PCR gives an idea regarding the load of
Isolation and Identification of DEV DVE viral DNA in the body tissues of infected ducklings
which can further be correlated with the dissemination of
Duck enteritis virus mainly attacks the immune organs. virus and progression of disease in different organs (58).
Samples recommended for virus isolation are liver, In situ hybridization can detect the presence of DEV
spleen, bursa, kidneys, peripheral blood lymphocytes DNA through specific oligonucleotide probes in tissue
(PBL), and cloacal swabs. Virus isolation is carried out by sections of various organs and can provide information
inoculation of susceptible 1‐day‐old Muscovy ducklings, about localization of the viral DNA in case of quick diag­
white Pekin ducklings, or onto CAM of 9‐ to 14‐day‐old nosis of the viral infection (11). With advances in design­
embryonating duck eggs. Characteristic lesions and ing diagnostic procedures, LAMP‐based nucleic acid
mortality in inoculated ducklings are highly suggestive of amplification methods for DEV detection has proven to
DEV. Virus also can be isolated and propagated in white be a rapid, simple, accurate, specific, and sensitive method
Pekin or Muscovy duck embryo fibroblasts, primary cell for the diagnosis of DEV and has been considered to be a
culture of liver and kidney, and Vero cell lines, but inocu­ good choice for on‐farm disease diagnosis (30).
lation of infected tissue to susceptible ducklings is con­
sidered more sensitive than the virus isolation in cell
culture. Although a presumptive diagnosis can be made Differential Diagnosis
on the basis of gross and histopathologic lesions, isola­ Differential diagnosis requires consideration of other dis­
tion and identification of DEV confirms the diagnosis eases producing hemorrhagic and necrotic lesions in
even in the absence of typical lesions when coupled with Anseriformes. In domestic ducks, common diseases
serological tests and molecular diagnosis. ­producing such changes are duck virus hepatitis, fowl chol­
era, necrotic enteritis, coccidiosis, and specific intoxications.
Serological Diagnosis Although Newcastle disease, fowl pox, and fowl plague are
reported to produce similar changes in Anseriformes, these
Virus neutralization assay is the most classical method diseases have been infrequently reported.
for identification of DVE. Increase in virus neutralization
(35) titers following convalescence from DEV will dem­
onstrate progress of the disease within a flock. A VN
index of 1.75 or more indicates infection with DEV (12). Intervention Strategies
A VN index of 0–1.5 has been found in sera of domestic
and wild waterfowl not exposed to the disease. The use of Management Procedures
chicken embryo‐adapted virus in chicken eggs for VN Prevention is achieved by maintaining susceptible birds
studies is safer and more convenient than the use of field‐ in environments free from exposure to the virus. These
strain viruses inoculated onto the CAM of duck eggs (12). measures include the addition of stock known to be free
Immunofluorescence tests can be used to detect viral from infection and avoiding direct and indirect contact
antigens in cell cultures or tissue sections (65). Other sero­ with possibly contaminated material. Introduction of the
logic procedures for detecting antibodies include a micro­ disease by free‐flying Anseriformes and contaminated
titer plate isolation and neutralization test using duck aquatic environments must be prevented. All possible
embryo fibroblasts, a reverse passive hemagglutination measures should be taken to prevent dissemination of
test (14), agar gel immunodiffusion test, and ELISA (31, virus by free‐flowing water. After DEV has been intro­
77, 82). A Dot‐ELISA and passive hemagglutination assays duced, control can be achieved by depopulation, removal
have been developed for detection of DEV antibodies; of birds from the contaminated environments, sanitation,
however, the specificity and sensitivity of these two assays disinfection, and vaccination of all susceptible ducklings.
were shown to be moderate (51). An immunochromato­ In countries where the disease is not enzootic and is
graphic strip test has been developed for use as a field test truly exotic, effective quarantine of imported or clinically
for monitoring flock immunity post vaccination and suspected Anseriformes should be done. Accordingly,
detection of exposure to DEV in unvaccinated flocks. This surveillance of ornamental bird collections, zoos, and
test is easy to perform, rapid (15 minutes), specific, and domestic growers of Anseriformes should be performed
with equal sensitivity as the antibody ELISA (64). by using efficient detection assay of DEV in laboratory.

Molecular Diagnosis Vaccination and Types of Vaccines


Duck enteritis virus‐specific DNA segments can be Vaccination has been used as a preventive measure and
amplified by PCR from infected cell culture supernatant also for controlling disease outbreaks. Currently, both
470 Section II  Viral Diseases

live attenuated and inactivated vaccines are being used duration against ­delivering pathogens and DEV infection
in broiler and breeder ducks that are over two weeks of by a ­single dose inoculation. It also suggested DEV has
age (84). Live attenuated vaccines are considered most the potential to be utilized as a promising viral vector
effective against DEV hence it is of foremost importance candidate for developing vaccines for poultry and aquatic
to maintain the vaccine at optimum physical and physi­ birds.
ological conditions of temperature, salt, and pH to pre­
vent any loss in the activity of vaccine candidate Field Vaccination Protocols and Regimes
molecule (50). The vaccinated ducks could excrete the The vaccine can be used in the face of an outbreak,
virus thus demands revaccination of the entire flock because it provides protection immediately after vac­
(26). By comparison, the inactivated DEV vaccine is cination due to an interference phenomenon (67). It
effective in protecting domestic and captive waterfowl should be noted, however, that birds in the period of
from the virulent strain infection (63) without the risk of incubation may not be protected. A naturally apatho­
introducing a live virus. genic and immunogenic strain of DVE was reported to
DNA vaccine is a promising strategy for protection be successful for active and passive immunization of
ducks against DEV infection. Vaccination with plas­ ducks (45, 46).
mids or carrier E. coli for expressing DEV gB/gD/gC/ Attenuated live virus vaccine is administered by sub­
UL24/tgB genes induced potent cellular and humoral cutaneous or intramuscular routes in domestic duck­
immunity against DEV in ducks (3, 44, 49, 87). However, lings more than two weeks of age. Normally, the breeding
their relatively low immunogenicity is an obstacle to flocks are vaccinated. Flocks maintained for more than a
their use. year are revaccinated annually. Apparently, vaccinated
Recent studies supported the use of attenuated DEV ducklings do not excrete inoculated virus to a degree
vaccine strain as an efficient vector for developing poly­ that would be sufficient to bring about contact immuni­
valent live attenuated vaccine against high pathogenic zation (28, 71).
avian influenza virus (AIV) strain H5N1 (47, 75) and
H9N2 (69), Duck Hepatitis A virus type 1 and 3 (88), and
Treatment
duck ­ tembusu virus (DTMUV) (10). This vaccine
provided speedy immunological protection for long
­ There is no specific treatment of DEV infection.

­Hemorrhagic Nephritis Enteritis of Geese


Jean‐Luc Guérin

Summary Introduction
Agent, Infection, and  Disease.  Hemorrhagic nephritis Definition and Synonyms
enteritis of Geese (HNEG) is one of the major viral diseases
Hemorrhagic nephritis enteritis of Geese (HNEG) is
of geese. The causative agent is a polyomavirus, namely
one of the major viral diseases of geese. Due to confu­
Goose hemorrhagic polyomavirus (GHPV). Goslings are
sion with goose parvovirus infection HNEG had long
susceptible up to 14 weeks of age, while ducks are healthy
been named “young geese disease” or “late form of
viral carriers. The main clinical signs are lameness and
Derzsy’s disease”. According to its etiology, a more rele­
prostration, leading to death. Main necropsic signs are
vant denomination should be “goose polyomavirosis.”
edema, ascites, and nephritis. In chronic forms of HNEG,
This systemic, frequently lethal disease is to date the
visceral and/or articular gout is frequent.
only condition associated with a polyomavirus in a
poultry species.
Diagnosis.  Mortality and lesions (edema, ascites, and
nephritis) on goslings are suggestive of HNEG. Confirmation
is routinely based on polymerase chain reaction (PCR).
Public Health Significance
Intervention.  Prevention is based on biosecurity, Polyomaviruses have a very narrow host range. This is
including ­ prevention of contact with ducks which supported by evidence of codivergence of mammalian
may be healthy ­ carriers of goose polyomavirus. and avian polyomaviruses with their respective hosts
Experimental ­vaccines  have been developed but are (7). Hemorrhagic nephritis enteritis of Geese is thought
not commercially available. to have no public health implication.
Chapter 13  Viral Infections of Waterfowl 471

History
Hemorrhagic nephritis enteritis of Geese was first
described in 1969 in Hungary (1) and was described a few
years later in Germany, then in France (16). For many
years, HNEG was suspected to correspond to a late evo­
lution of Derzsy’s disease. Etiology of HNEG was clarified
in France in 2000 (6). More recently, it was shown that
ducks are actually largely healthy carriers of the virus (3).

Etiology
Classification
The agent of HNEG, namely Goose hemorrhagic polyoma-
virus (GHPV), is a member of the Polyomaviridae family.
Figure 13.16  Electron micrograph of a goose hemorrhagic
It has been recently assigned into the Gammapolyomavirus polyomavirus (GHPV)‐infected cell. Notice many naked virions in
genus with all other avian polyomaviruses (9). This virus is the nucleus and peripheral accumulation of chromatin. ×25,000.
significantly divergent from Budgerigar fledgling polyoma-
virus (BFPV), the prototype avian polyomavirus infecting
hours after incubation at 55°C (6). The virus also resists
psittacines, falconiformes, and passerines (9).
freezing–thawing cycles and lipid solvents: treatment
with 1% phenol has no effect on its viability. Avian poly­
Morphology omaviruses are mostly sensitive to chloride‐derived
Virus particles are naked and spherical, and show icosahe­ products (15).
dral symmetry. Their size ranges from 40–50 nm in diame­
ter (6). Buoyant density of virions is of 1.20 g/cm−3 in sucrose Strain Classification
gradient (6), which corresponds to 1.34–1.35 g/cm−3 in CsCl.
Genetic variability among field isolates has been poorly
investigated so far. Nevertheless, polyomavirus genomes
Chemical Composition are generally highly conserved and phylogenic analysis of
The GHPV genome is a circular, double‐stranded DNA of GHPV confirmed that VP1 is remarkably conserved
5,256 base pairs (8). Genome organization of all polyomavi­ among isolates from different countries (14). Moreover,
ruses shares common features, with a set of early genes encod­ duck GHPV strains do not show distinctive genetic
ing polymerases (t and T antigens) and late genes, encoding ­features compared to the goose strains (3). No cross‐
structural proteins: VP1, the main capsid protein, and two neutralization experiment has been performed so far on
other structural proteins, VP2 and VP3 (9). As for avian poly­ GHPV field isolates.
omaviruses, an additional VP4 has been demonstrated, the
precise functions of which still remain to be clarified (8). Laboratory Host System
Hemorrhagic nephritis enteritis of Geese is successfully
Virus Replication
reproduced by parenteral inoculation on 1‐day‐old gos­
Replication of GHPV occurs in the nucleus (6). Infected cells lings. Clinical end points are reached between 6 and 8
show a huge concentration of viral material in the nucleus, days postinoculation (PI), with peracute disease. Goslings
both in cultured cells and tissues of infected goslings Virus is are susceptible to inoculation either by subcutaneous or
easily detected in the nuclei by electron microscopy of intraperitoneal routes. All attempts to adapt the HNEG
immunofluorescence (Figure  13.16). Releasing of virions virus to duck fibroblasts or embryos, as well as goose
implicates disruption of cell membrane. fibroblasts, remained unsuccessful (5, 6). Propagation of
GHPV on goose embryos has been reported: 14‐day‐old
goose embryos inoculated onto the chorioallantoic
Susceptibility to Chemical and membrane (CAM) died from 8–10 days PI, with lesions
Physical Agents similar to those described in goslings (1). Virus propaga­
tion can be accomplished by propagation on epithelial
Goose hemorrhagic polyomavirus shows a great resist­ primary cells derived from 1‐day‐old gosling kidneys.
ance to heating: virus is still fully virulent even two Cytopathic effects appear by day 5 PI; granulations and
472 Section II  Viral Diseases

vesicles are distinguished in the cytoplasm, followed by Clinical Signs, Morbidity, and Mortality
budding of the cell, and finally cell detachment from the
Hemorrhagic nephritis enteritis of Geese has been
monolayer (6). Cell‐based dilution titration procedures
described in goslings from 4–14 weeks of age. In affected
are seldom done, because cytopathic effect appears late
flocks, morbidity ranges from 10–80% and death is the
after infection. Alternatively, detection and quantifica­
most common outcome (6). Clinical signs develop rap­
tion of virus yields from cell cultures or tissues can be
idly only a few hours before death; birds sit alone, away
determined by quantitative real‐time PCR (3, 5, 12).
from the flock, stay in a coma, and die (2, 10). Nervous
signs, such as opisthotonos, are only observed after
experimental or iatrogenic infections of goslings (11).
Pathobiology and Epizootiology Chronic evolution of the disease leads to urate deposits
on viscera and in joints, resulting in lameness. In these
Incidence and Distribution late forms, mortality may be limited to a few birds every
Until now, HNEG has been mostly described in Hungary, day up to the age of 12 weeks.
Germany, France, and Poland (2, 4, 6, 10, 14, 16).
Occurrence of the disease in other countries seems likely, Pathology
although there are no published reports confirming this.
Cases are frequently observed in winter, probably due to Necropsic findings in goose include edema of subcuta­
climatic conditions or weakness of the goslings hatched neous connective tissues, gelatinous ascites, inflamma­
from light‐conditioned breeders. tion of the kidneys (Figure  13.17), and less frequently,
hemorrhagic enteritis. Renal dysfunction leads to an
increase of blood uric acid concentration. Geese that die
Natural and Experimental Hosts after a chronic infection show visceral gout and deposi­
Hemorrhagic nephritis enteritis of Geese has only been tion of urates in the joints (Figures 13.18 and 13.19) (10,
described to date in growing geese. Subclinical infec­ 11, 16). Histopathologically, the most obvious features
tions have been reported in migrating wild geese (6). are: (1) an interstitial nephritis and necrosis of the kid­
Other waterfowl species, such as mule or Muscovy duck­ ney tubular epithelium, and (2) a moderate to severe
lings, are clinically refractory to GHPV inoculation, but lymphocytosis in cortical and medullary regions of
can be infected at high replication levels. Field surveys ­bursal follicles, suggestive of B‐lymphocyte depletion
have suggested that ducks may constitute a significant (6, 11). Gross lesions of enteritis are associated with
subclinical reservoir of GHPV (3). ­necrosis of intestinal ­epithelium. Hemorrhagic foci also
are observed in most tissues, particularly in acute infec­
tions (11, 14). No inclusion could be detected in tissues
Transmission, Carriers, and Vectors of birds diagnosed with HNEG (6, 11). Electron micros­
Infected geese and ducks excrete significant amounts of copy examination of infected tissues shows aggregated
virus in their droppings, resulting in dissemination of virions in nuclei (Figure  13.16) and large vesicles of
contagious material to the environment and easy direct dense material, including optically clear centers, in the
and indirect contamination. Vertical transmission of the cytoplasm of about 20% of the infected cells (6). In
virus through the egg has never been confirmed, but infected ducks, no macroscopic or microscopic lesion
cannot be excluded. The experimental infection of goose has been observed (3).
embryos has been shown, although this does not for­
mally demonstrate the field occurrence of vertical trans­
mission (1). No biologic vector seems to be involved in
GHPV transmission.

Incubation Period
The incubation period is mostly age dependent.
Inoculation of day‐old goslings results in death within
6–8 days (11). In contrast, in 3‐week‐old goslings, the
incubation period could last for up to 15 days (14). After
four weeks, inoculation results mostly in nonclinical
infection. Although clinical signs rarely start before five
or six weeks of age, contamination from infected birds Figure 13.17  Gosling affected by hemorrhagic nephritis enteritis
could possibly occur early in life. of geese (HNEG), showing edema and swelling of kidneys.
Chapter 13  Viral Infections of Waterfowl 473

Pathogenesis of the Infectious
Process
During the course of infection, GHPV seems to repli­
cate first in endothelial cells; nuclear enlargement of
endothelial cells and arteriolitis are the first lesions
noticed (11, 14). These histological findings suggest a
selective tropism for endothelial cells, which might
be  of great relevance in pathogenesis of HNEG.
Endothelial cells are indeed known to play a critical
role in many biological pathways, resulting in vascular
dysfunctions as ascites or edema. Another main target
of GHPV is lymphoid cells; virions are observed in
many bursal lymphoid cells, and cloacal bursa (bursa
of Fabricius) systematically shows significant lympho­
lysis. However, thymic lymphoid cells are less or not
affected. This feature is fairly relevant with the well
documented tropism of polyomaviruses to B‐lympho­
cytes, suggesting immunodepressive effects in sub­
clinical infections (7).

Immunity

Figure 13.18  Chronic form of hemorrhagic nephritis enteritis of Immunological aspects of HNEG have so far received lit­
geese (HNEG), visceral gout with deposition of urates. tle attention. Neutralizing antibodies are detected in pre­
viously infected birds and their transmission to the
progeny seems very efficient, because goslings hatched
from infected breeders are refractory to experimental
infections with huge viral load (5, 17). The duration of
maternal immunity has not been fully determined, but
serological monitoring assays on goslings from vacci­
nated breeders suggest a complete disappearance of
maternal antibodies within three weeks of life (5).

Diagnosis
Isolation and Identification of Causative
Agent
Goose hemorrhagic polyomavirus can be detected in
clinical material from geese showing clinical disease, as
well as from nonclinical carrier birds. Isolation could be
based on either kidney cell culture (6) or goose embryo
inoculation (1), but these methods are time consuming
and can hardly be applied to routine diagnosis of HNEG.
Detection of the GHPV genome is therefore a more reli­
able way to detect the virus; end‐point or real‐time PCR
detection of DNA extracted from infected tissues (liver,
spleen, kidney) with primers designed on VP1 gene is
efficient and reliable (6, 12). In subclinically infected car­
riers, PCR assays can be advantageously performed on
Figure 13.19  Chronic form of hemorrhagic nephritis enteritis of blood samples, spleen, or cloacal swabs (12). A loop‐
geese (HNEG), urates deposition in the tibio‐metatarsal joints. mediated isothermal amplification (LAMP) assay allows
474 Section II  Viral Diseases

a rapid molecular diagnosis (18). Serology appears of ducks can be healthy carriers of GHPV, mixed goose and
limited value to detect infection by a polyomavirus, duck farming systems should be carefully reconsidered and
because serologic response is greatly variable, while virus biosecurity between these species enforced (3).
may persist in infected birds for months, if not years (12,
14). ELISA has been developed using viral antigens (5) or Vaccination
recombinant VP1.
Management procedures are unlikely to be sufficient for
the control of HNEG infection. Vaccination of breeders
Differential Diagnosis could be used to provide maternal immunity to goslings
Lesions of ascites, subcutaneous edema, visceral urate when they are critically sensitive to virus contamination
deposition, and nephritis in 4‐ to 10‐week‐old goslings (5). An inactivated vaccine, based on viral antigen pro­
are all suggestive of HNEG. However, similar lesions also duced by propagation on goose kidney cells, inactivated
may be associated with goose parvovirus infections. with β‐propiolactone, and adjuvanted with carbopol, has
Histopathological, virological, or serological procedures been the subject of a trial on breeding geese and induced
may be helpful in confirming the etiology. Hemorrhagic maternal antibodies, providing protection to goslings
nephritis enteritis of Geese is probably underdiagnosed against a viral challenge (5). This vaccine, when adminis­
because of the existing confusion with Derzsy’s disease. tered to growing goslings, induced a significant serologi­
cal response for several weeks.
A rational vaccination schedule could rely on: (1)
Intervention Strategies administration to breeders before each laying period and
(2) vaccination of growing goslings to induce an active
Management Procedures immunity covering the whole economic life of birds.
A subunit vaccine, based on recombinant VP1 protein
Goose polyomavirus spreads from carriers and clinically produced in E. coli, also induced protection in goslings
affected birds, mostly by the fecal route (6, 11). Disinfection against a viral challenge, confirming that VP1 is the
procedures should be thoroughly observed; that is, com­ major antigenic determinant of polyomaviruses and can
plete removal of organic material, followed by the use of an be expressed in subunit or vectored vaccines (13).
appropriate disinfectant, is required to prevent or inter­
rupt a disease outbreak. Chloride derived products are
considered efficient to inactivate polyomaviruses, but are Treatment
particularly sensitive to the presence of organic debris (15). There is no effective treatment. Technical management
Because infected birds have a viremia, needles used for of an infected flock should be adapted to prevent stress;
administration of vaccines should be sterilized between this may be helpful in preventing nonclinically infected
uses. Though transmission of HNEG virus through the egg birds from developing HNEG.
is not clarified, biosecurity practices should be respected in
the hatchery to limit potential early contamination of gos­
lings before they reach the farm. When goslings are Acknowledgement
infected by the virus, occurrence of clinical signs may
greatly depend on management factors such as cold and/or Grateful acknowledgment is made to Dr. Richard E,
stress. Oil‐adjuvant vaccines should be administered with Gough, the author of previous chapters, for his contribu­
extreme caution to flocks affected by the disease. Because tions included in this text.

­Parvovirus Infections of Waterfowl


Vilmos J. Palya

Summary feathers. The virus is transmitted both vertically and


horizontally. All geographical areas of the world with
Agent, Infection, and Disease.  Anseriform dependoparvovirus intensive waterfowl production are affected with the
1 can cause a highly contagious fatal disease of young disease.
geese and Muscovy ducks, and less severe chronic
disease in mule and Pekin ducks. Severity of the Diagnosis.  The preferred diagnostic test is molecular
disease decreases with age. The chronic form is detection of the agent by polymerase chain reaction
characterized by growth retardation and loss of (PCR) and virus isolation. Antibodies can be detected by
Chapter 13  Viral Infections of Waterfowl 475

virus neutralization or enzyme‐linked immunosorbent of a new waterfowl parvovirus strain, named Muscovy
assay (ELISA). duck parvovirus (MDPV or DPV), was reported in
France and Taiwan (41). Short beak and dwarfism syn­
Intervention.  Vaccination of breeders with live and/or drome was first reported in mule and Muscovy ducks in
inactivated vaccines provides passive immunity to the France and Poland in the early 1970s and later in mule,
progeny to ­ prevent severe disease in young birds. Muscovy, and Pekin ducks in Taiwan (36, 41, 44). In
Vaccination of progeny provides life long immunity. recent years a similar disease affecting Cherry Valley and
mule ducks has been observed frequently in China (67).

Introduction
Etiology
Definition and Synonyms
In early reports describing the disease several etiological
Waterfowl parvoviruses can cause different disease condi­ agents have been proposed, including bacteria, reovi­
tions depending on the species and the age of affected ruses, and adenoviruses as they were frequently isolated
waterfowl. The highly contagious fatal disease affecting or detected from diseased goslings (41). In subsequent
young geese and Muscovy ducks (Cairina moschata), studies, however, it has been confirmed that the etiologi­
caused by goose parvovirus (GPV) was described under cal agent is a parvovirus (27, 41).
different names depending on the major clinical–patho­
logical features that were observed (41). To eliminate the
discrepancies in nomenclature, the World Poultry Classification
Association in 1974 agreed to name the disease in geese as According to the International Committee on Taxonomy
Derzsy’s disease to acknowledge the pioneering work of of Viruses (ICTV), waterfowl parvoviruses are classified
the Hungarian researcher Domokos Derzsy. The disease as autonomous members of the Dependovirus genus in
caused by the duck parvovirus (DPV) in Muscovy duck is the Parvoviridae family. All GPV and DPV belong to a
often referred to as parvoviruses of Muscovy ducks. A syn­ single species, Anseriform dependoparvovirus 1 (9).
drome associated with parvovirus infection causing short
beak and dwarfism syndrome (SBDS), also called as beak
atrophy and dwarfism syndrome (BADS), in Muscovy, Morphology
mule, and Pekin ducks was also described (36, 39, 44). It Waterfowl parvoviruses have small nonenveloped viri­
has been shown that waterfowl parvoviruses can be divided ons with icosahedral symmetry (Figure  13.20) and a
into GPV and DPV species, the ­latter frequently referred to diameter of 20–22 nm. The density of the virus in cesium
as Muscovy duck parvovirus (MDPV) (30, 68). Most recent chloride is approximately 1.38 g/mL (15).
classification assigned both GPV and DPV into a single
species, namely Anseriform dependoparvovirus 1 (9).
Chemical Composition

Economic Significance The single‐stranded DNA genome of GPV comprises of


5,106 nucleotides, and DPV is 5,132 nucleotides in length
In countries where intensive farming of geese and ducks including the inverted terminal repeats with U‐shaped
is practiced, the disease has important economic signifi­ hairpin structures at both ends. Both plus‐ and minus‐
cance. Introduction of vaccination has greatly reduced strand genomes are encapsidated into the virions. The
the impact of the disease. genome contains two large open reading frames (ORF).
The 5’ open reading frame encodes the non‐structural NS1
Public Health Significance (or Rep1) protein as well as several smaller proteins gener­
ated after mRNA splicing (33). The 3’ open reading frame
There is no known public health risk associated with encodes three related proteins referred to as VP1, VP2, and
infection of waterfowl parvoviruses. VP3. These proteins are generated from the use of different
initiation codons and through proteolytic cleavage (41, 46).
These three capsid proteins constitute the icosahedral cap­
History sid in a ratio of approximately 1 : 1 : 8 (17).

In the late 1950s and early 1960s a highly contagious and


Virus Replication
fatal disease of young goslings was reported in China and
several European countries where intensive goose farm­ The replication of GPV and DPV, similar to that of other
ing was present (41). A few decades later the occurrence parvoviruses, occurs in the nucleus of host cells by using
476 Section II  Viral Diseases

(A) (B)

100 nm 100 nm

Figure 13.20  Electron micrograph of purified goose parvovirus. (A) Purified virions. (B) Virions in the feces of a naturally infected
10‐day‐old gosling showing intact and hollow particles (arrow).

the apparatus of the cells without the presence of helper GPV and DPV and cause a certain level of cross‐immu­
virus (1, 10). Parvoviruses in general can only replicate nity between them (34).
productively in actively mitotic host cell populations.
Accordingly, pathogenic or lethal infections typically Genetic Characteristics
occur in fetal or neonatal hosts, which have many divid­ Phylogenetic analysis based on the structural proteins
ing cell populations, or involve adult tissues that remain verified the existence of two monophyletic groups of
actively dividing in later life (10). The expression strategy waterfowl parvoviruses, the genetic linages of GPV and
of goose parvovirus exhibits features of both the DPV (41). Molecular analysis of the genomes of GPV and
Dependovirus and Parvovirus genera (33, 46). DPV identified approximately 80% (2, 7, 68) nucleotide
sequence homology between the two linages. The nucleo­
tide sequences of GPV and DPV viral capsid genes share
Susceptibility to Chemical and Physical
77% similarity at DNA, and 84.6% at amino acids level.
Agents
The most variable region resides in the N‐terminal of VP2
Goose parvovirus is very resistant to chemical and physical before the initiation codon of VP3 with 35% amino acids
inactivation. The virus is stable at 65°C for 30 minutes and divergence between GPV and DPV, causing major and
at pH 3.0 for 1 hour at 37°C. However, treatment with 0.5% minor changes on surface‐exposed residues of VP3 (7).
formaldehyde can destroy the infectivity of the virus (41). Among the GPV strains, separate monophyletic
­subgroups can be differentiated based on the geogra­
phical origin and pathogenic nature of the strain (57)
Strain Classification
(Figure  13.21). A DPV strain belonging to another
Antigenicity branch was isolated in the United States, showing only
Using cross‐neutralization tests, it was demonstrated 85% identity with both GPV and DPV strains (45).
that GPV differs antigenically from DPV (23). However, Phylogenetic analysis of certain duck parvovirus strains
because of the high level of amino acid sequence identity circulating in Muscovy duck flocks suggest that they are
between GPV and DPV, there is at least one NS1 epitope recombinant of DPV and GPV (50, 69). Strains isolated
and three VP1 epitopes that might cross‐react between from SBDS cases belong to a distinct linage of GPV (44).
Chapter 13  Viral Infections of Waterfowl 477

Bócsa/99HU
Bócsa/98HU
Öcsöd/00HU
Balástya/00HU
European group 1 strains
Nagyecsed/97HU
Lajosmizse/99HU
B/HU
Distance
LB/HU
0.02
06-0329/TW
99-0808/TW
86-1015/TW
82-0321/TW Asian GPV strains
01-1001/TW
82-0408/TW
82-0321v/TW
Csongrád/80HU
Pusztaföldvár/79HU Vaccines & low pathogenic
B42/Hu GPV strains
486/GB
Goose parvoviruses D291/3/03PL
D291/2/03PL
D441/1/04SE European group 2 strains
D462/1/04GB
D462/2/04GB
Hoekstra/FR
SHM319/DE
D523/2/05FR
D518/3/05FR
M15/15CN
QH15/15CN
sdlc01/15CN SBDS related GPV strains
D657/3/1/06FR
D146/3/02FR
D697/3/06FR
Duck parvoviruses D479/12/04FR
PSU-31010/US US DPV strain
89384/FR
European DPV strains
FM/FR
97-0104/TW
90-0219v/TW Asian DPV strains
90-0219/TW

Figure 13.21  Phylogenetic relationship of goose and duck parvoviruses. The tree is based on a 493 bp region of the VP1 protein. GenBank
accession numbers of the strains included: Bócsa/99HU (AY496898), Bócsa/98HU, Öcsöd/00HU, Balástya/00HU, Nagyecsed/97HU, and
Lajosmizse/99HU had the same accession number (AY496897), B/HU (U25749), LB/69HU (AY496900), 06‐0329 (EU583391), 99‐0808
(AY382888), 86‐1015 (AY382887), 82‐0321 (AY382884), 01‐1001 (AY382889), 82‐0408 (AY382886), 82‐0321v (AY382885), GPV486/GB
(AY496904), B42 (AY496901), Csongrád/80HU and Pusztaföldvár/79HU had the same accession number (AY496903), D291/2/03PL
(DQ862009), D291/3/03PL (DQ862010), D441/1/04SE (DQ862008), D462/1/04GB (DQ86011), D462/2/04GB (DQ862012), SHM319/DE
(U34761), Hoekstra/FR (AY496907), D146/3/02FR (AY496906), D479/12/04FR (EU938702), D518/3/05FR (EU938703), D523/2/05/FR
(EU938704), D657/3/06FR (EU938705), D697/3/06FR (EU938706), PSU‐31010 (DQ413026), 89384 (Z68272), FM (U22967), 97‐0104
(AY382893), 90‐0219 (AY382892), 90‐0219v (AY382890), sdlc01 (KT343253), QH15 (KT751090) and M15 (KU844283).

Compared with the full‐length genomes of other classi­ related parvovirus, isolated from mule and Cherry Valley
cal goose and duck parvovirus field isolates, the inverted duck, could produce typical symptoms of SBDS, with
terminal repeat of GPV causing the SBDS contains sev­ high morbidity and low mortality rates (6, 32, 44).
eral fragments. These characteristics are similar to those
of certain attenuated GPV vaccine strain (4).
Laboratory Host Systems
Pathogenicity Goose parvovirus and DPV can be propagated in embry­
The two linages of waterfowl parvoviruses differ from onating goose or Muscovy duck eggs or young suscepti­
each other regarding their pathogenicity and host range. ble birds of the same species and primary cell cultures
Goose parvovirus can cause disease both in goslings and prepared from the embryos of these species. After a cer­
ducklings, whereas the antigenically distinct DPV causes tain level of adaptation the virus can be grown in Pekin
disease only in Muscovy ducks (13, 23). Normally, Cherry duck embryos as well (20). Certain strains of GPV and
Valley ducks and mule ducks are resistant to classical MDPV could be propagated in continuous cell lines from
goose parvovirus infection. However, a distinct GPV‐ Muscovy ducks (24).
478 Section II  Viral Diseases

Pathobiology and Epizootiology survive infection or those that are subclinically infected


frequently become carriers and transmit the virus both
Incidence and Distribution horizontally and vertically. Latently infected breeders
play an important role in the transmission of virus
Waterfowl parvoviruses have been reported from all through their eggs to susceptible goslings or ducklings in
major goose and Muscovy duck farming countries of the hatchery (5, 12, 41, 48). Duck parvovirus might cause
Europe and Asia (41). There have been reports of DPV asymptomatic infection in geese and they can shed virus
outbreaks in Muscovy ducks in the United States (45, 62) from cloaca for one to four weeks post‐inoculation,
as well. Short beak and dwarfism syndrome in mule and therefore geese can be a source for infection of DPV (66).
Pekin ducks has been observed in France, Poland, Wild geese visiting the same pasture or water pond as
Taiwan, and similar disease was described in meat type domestic geese and Muscovy ducks could also be a
Pekin ducks in China (3, 6, 36, 41, 67). source of infection and may play a role in the introduc­
tion of the infection to a disease‐free country or flocks
Natural and Experimental Hosts (22). No biological vectors have been identified.

All breeds of domestic geese, Muscovy, mule, and Pekin


ducks, and some other members of the Anatidae family Incubation Period
are the only species in which clinical disease has been Infection of susceptible birds during the first week of life
observed (41). Muscovy, mule, Tsaiya, and Pekin ducks results in the appearance of clinical signs within 3–5
are susceptible to GPV strains causing SBDS (4), and a days. In 2‐ to 3‐week‐old birds, the incubation period
genetically closely related GPV was detected in other may vary between 5 and 10 days (13, 20, 48). In general
duck species in Poland (mandarin, wood, falcated, and the older the bird at infection, the more time passes
silver teal ducks) (49). The embryo‐adapted GPV given between infection and the appearance of clinical signs.
by parenteral route can also infect Pekin ducks. Other In mule and Pekin ducks the incubation period is consid­
breeds of domestic poultry appear refractory to experi­ erably longer. Following infection of one‐day‐old duck­
mental infection with GPV (20). lings, it can take 2–3 weeks before the characteristic
In a serological survey (19), some species of wild geese signs of SBDS can be seen (4, 44).
also tested positive for neutralizing antibody to GPV.
Disease caused by GPV has been reported in Canada geese
(Branta canadensis) and snow geese (Chen hypoborea Clinical Signs
atlantica) as a consequence of accidental infection (47). Depending on the age and the immune status of gos­
lings and Muscovy ducklings when GPV infection
Age of Host Commonly Affected occurs, the disease may be present either in acute, sub­
The diseases caused by waterfowl parvoviruses are acute, or chronic forms, representing multiple patho­
strictly age dependent. Losses decrease with age, reach­ logical features of the disease (41). In the acute form,
ing a negligible level when infection occurs in birds older the course of the disease is usually rapid. The birds
than five to six weeks (41). Although older birds do not develop signs of illness with anorexia, polydipsia, weak­
show clinical signs of infection, they respond immuno­ ness, watery diarrhea, and prostration, followed by
logically (29). death within a few days. Many birds show nasal and
ocular discharge, conjunctivitis, profuse white diar­
rhea, and loss of natal down (Figure  13.22). Some of
Transmission, Carriers, and Vectors
these birds have mucosal necrosis and a fibrinous pseu­
Infection of birds may occur both by vertical and hori­ domembrane covering the tongue and oral cavity. In
zontal routes. The most serious outbreaks occur in sus­ birds infected at an older age, between one and three
ceptible goslings following vertical transmission of the weeks, the course of the disease can be either subacute
virus. In horizontal transmission, ingestion of virus‐con­ or a more prolonged. In birds that survive the acute
taminated feed and water is of prime importance. Goose phase of the disease and in those that develop the
parvovirus and DPV are similar in that the two viruses chronic form of the disease, as a consequence of infec­
re‐excreted in large amounts in the feces of infected tion at an older age, the birds lose their feathers, espe­
waterfowl, subsequently spreading rapidly to susceptible cially on the back, neck, and wing, and have profound
birds by both direct and indirect routes (41). growth retardation (Figure  13.23). Deaths may occur
Due to its high resistance to extreme physical and between the third and tenth weeks of age (11).
chemical effects (heat, cold, dryness, disinfectants), the Duck parvovirus affects only Muscovy ducks and the
virus can remain infectious for long periods in the course of disease is similar the one of Derzsy’s disease in
­contaminated environment of animal houses. Birds that geese (13). In Muscovy ducks, locomotor problems
Chapter 13  Viral Infections of Waterfowl 479

including weakness, lateral recumbency, and inability to and ducklings (41). Infection of susceptible birds during
walk are usually evident. the first 2–3 weeks of life still results in almost 100%
In SBDS parvovirus‐infected duck flocks the disease is morbidity and high mortality (10–60%). Resistance
characterized with high morbidity and low mortality. against the disease increases with age; infection later
The disease mostly affects young mule and Cherry Valley than 3–4 weeks of age usually remains subclinical (44).
ducklings, characterized by a notably shortened beak However, in birds with impaired immune systems, as a
and leg bones, protruded swollen tongue, stunted consequence of concomitant infection with immuno­
growth, fractured feathers (Figure  13.24). The patho­ suppressive viruses (e.g., reovirus, circovirus) or due to
genicity of SBDS parvovirus is lower than that of classical nutritional (mycotoxins) and environmental factors the
waterfowl parvoviruses (4, 6, 44). age of susceptibility may extend leading to the “late
form” of the disease, causing significant economic losses
Morbidity and Mortality
Infection in the hatcheries or at less than one week of
age can result in 100% mortality in susceptible goslings

Figure 13.23  Derzsy’s disease. Gosling that survived the acute


Figure 13.22  Gosling infected with goose parvovirus (GPV) phase of the disease showing growth retardation and loss of
showing weakness, prostration and loss of natal down. feathers on the back and neck (natural case).

(A) (B)

Figure 13.24  Short beak and dwarfism syndrome (SBDS) in Mule duck infected with goose parvovirus (GPV) at one day of age
(experimental infection). (A) Growth retardation, shortening of tibia and fractured feathers on the back. (B) Short beak and protruded
tongue. For comparison, an uninfected duck of the same age is shown next to the diseased one.
480 Section II  Viral Diseases

up to 6–10 weeks of age. The presence of maternally‐ Usually no gross lesions of the internal organs can be
derived antibodies can considerably influence the course seen in SBDS. In a few cases, when the bird dies at a very
of the disease and consequently the rate of morbidity young age following infection, fibrinous perihepatitis,
and mortality (41). hydropericardium, and ascites are observed (44).
In SBDS cases, typical clinical signs usually appeared
in 15‐day‐old ducks, and population of diseased ducks is Microscopic Lesions
continually growing with age until the day of slaughter. The specific lesions observed in the affected geese and
The feed conversion ratio (FCR) of infected ducks is ducks include myopathy of skeletal muscle, hepatitis,
worse than that of healthy ducks (4). myocarditis, sciatic neuritis, and polioencephalomyelitis.
Other commonly observed lesions include atrophy of
lymphoid organs (bursa of Fabricius, spleen, and thymus)
Pathology
(13, 55).
Gross Lesions Both GPV and DPV infections produce extensive
In acute cases, the lesions considered characteristic in degenerative changes of myocardial cells, loss of striation
geese include enlarged and congested liver with fibrin­ of myocardial fibers, and the presence of scattered
ous membrane on the surface (Figure 13.25B), pale myo­ Cowdry type‐A intranuclear inclusions. In liver, the pre­
cardium, and dilatation of the heart. Typically, a dominant lesions are vacuolic degeneration of hepato­
serofibrinous perihepatitis with large volumes of straw‐ cytes (Figure 13.27), multifocal single‐cell necrosis, and
colored fluid in the abdominal cavity and pale dilated occasional eosinophilic intranuclear inclusion bodies. In
heart (Figure  13.25A) are present (41). In the acute the case of the predominantly enteric form of the dis­
enteric form of the disease, severe necrotic enteritis with ease, necrosis of intestinal epithelium, especially the
diphtheritic lesion in the small intestine (Figure 13.26) is crypt‐lining epithelium, can be seen in the duodenum
the predominant postmortem finding (21). Diphtheritic and proximal jejunum. Intranuclear inclusions may be
and ulcerative lesions may be observed in the mouth and found in the damaged epithelium cells. In Muscovy
pharynx as well. ducklings, degeneration and necrosis of muscle fiber
In Muscovy ducks, the gross pathological lesions with lympho‐histiocytic infiltration, together with mild
include pale thigh and heart muscles, increased pericar­ sciatic neuritis and polioencephalomyelitis (Figure 13.28)
dial fluid, serofibrinous perihepatitis, and ascites. In are frequent findings (13, 62). The pathological features,
­prolonged cases, the birds become stunted, have chronic however, vary depending on the clinical course of the
congestion of liver, and ascites (13, 23). disease. The main histological lesions in SBDS include

(A) (B)

Figure 13.25  Derzsy’s disease. Gross pathology of gosling got infected during the first week of life. (A) Enlarged and congested liver with
fibrinous exudate on the surface, ascites, pale myocardium, and dilatation of heart (natural case). (B) Fibrinous membrane on the surface
of liver.
Chapter 13  Viral Infections of Waterfowl 481

hemorrhage of the thymus, calcification of the tip of the liver, spleen, kidney, and lymphoid organs, but not in any
tongue and degeneration of the liver, whereas, no other part of the gastrointestinal and respiratory system. From
obvious lesions could be observed in other organs or 8 hours to 9 days postinfection, the virus was detected in
tissues. all organs tested, with the highest amount in the liver,
spleen, kidney, and lymphoid organs (65). The level of
Pathogenesis of the Infection Process GPV and MDPV replication and distribution plays a sig­
nificant role in the parvoviral infection progress and is
Detailed studies on the pathogenesis of waterfowl parvo­ strictly correlated to clinical symptoms. Analysis of
virus are not available. The most accepted theory is that quantitative real‐time PCR results revealed correlation
following infection, replication occurs in the intestinal
wall. The virus enters the bloodstream and reaches
­secondary target organs, including the liver and heart,
where the most severe pathological changes occur (28).
Another possibility is that the virus enters the blood­
stream through the nasopharyngeal lymphoid tissues
and reaches the secondary target organs more rapidly
(26). This latter theory is supported by recent data
obtained by quantification of GPV in different organs of
experimentally infected goslings. Goose parvovirus was
first detected at 4 hours postinfection in blood, heart,

Figure 13.26  Enteric form of Derzsy’s disease. Haemorrhagic‐ Figure 13.27  Liver section from a 10‐day‐old gosling infected
necrotic enteritis with pseudomembrane formation in the small with goose parvovirus (GPV) showing widespread vacuolation
intestine. and degeneration of hepatocytes.

(A) (B)

Figure 13.28  Parvovirosis of Muscovy duck. (A) Zenker’s necrosis of muscle fiber with lympho‐histiocytic infiltration. (B) Focal lymphocytic
infiltration and gliosis in the cerebrum.
482 Section II  Viral Diseases

between the age of the infected birds, the observed Molecular Identification
­clinical symptoms, and DNA copy number of GPV and The classical detection of GPV and MDPV by virus isolation
MDPV in the different organs (63). in gosling or duckling embryos, cell cultures, and serological
assays like seroneutralization test (SN) is time consuming
and dependent on the availability of SPF gosling embryos
Immunity
and tissue cultures. These limitations resulted in the applica­
Active Immunity tion of PCR based techniques which allowed fast detection
Following infection or vaccination, humoral immune and identification of both GPV and MDPV. Primers have
response develops, which is characterized by the initial been designed to amplify conserved regions of the capsid
production of IgM and then IgY‐type immunoglobulin protein genes (2, 57) or the nonstructural gene (59).
(29). Using virus neutralization (VN) and agar gel pre­ Differentiation of GPV and DPV can be done by restriction
cipitin (AGP) tests, high and persistent antibody levels fragment length polymorphism (RFLP) or nucleic acid
could be detected for several months after infection (16). sequencing, the latter being the most distinctive (2, 35, 45,
Although much less is known about the role of cell‐­ 52, 57). Nucleotide sequence analysis of capsid protein genes
mediated immunity, it is not thought to play a significant of GPV allows the differentiation of vaccine and wild strains
role in immunity to waterfowl parvovirus. (51, 57, 58). Another fast molecular method for the detection
of GPV and DPV is the loop‐mediated isothermal amplifica­
Passive Immunity tion assay (LAMP) that could remarkably simplify the detec­
From adult breeding geese and ducks that have been vac­ tion of both viruses (64). Quantification of GPV and DPV by
cinated or naturally infected with parvovirus, transfer of real‐time PCR has also been described (63, 65). The Real‐
maternal antibody occur via the egg yolk to their progeny time PCR method was developed for the rapid detection of
(16, 20, 28). This passively acquired antibody may persist novel duck‐origin goose parvovirus as well (40, 61).
until about 2–3 weeks of age based on the starting level
at hatch (14, 29).
Serology
Serology is useful for evaluating the immune status of
breeding flocks and their progeny. It is widely used to
Diagnosis confirm recent infection, efficiency of vaccination, and
determine the level of maternally‐derived antibodies
Isolation and Identification
(MDA) in newly hatched birds.
of the Causative Agent
A number of serological methods have been developed
Waterfowl parvovirus can be isolated from a variety of for detection of antibodies against GPV and DPV, including
organs of affected animals using embryonated goose or AGP, VN, and plaque neutralization assay, ELISA using
Muscovy duck eggs or tissue cultures prepared from whole virus or recombinant antigens, and indirect fluores­
them (41). Ten‐ to 15‐day‐old embryonated goose or cent antibody test (41). Viral proteins expressed by
Muscovy duck eggs should be inoculated via the allan­ Escherichia coli proved to be suitable antigens for western
toic cavity. Embryo mortality occur 5–10 days postin­ blotting or multiscreen western blotting assays (60). Non‐
oculation with liver lesions and hemorrhages on the structural (NS) protein‐based serological tests may be used
skin. Inoculation of goose or Muscovy duck embryo for differentiation of birds that have been vaccinated with
fibroblast cultures should be done before they reach subunit vaccines from those that are naturally infected (60).
confluency. The virus produces a well‐defined cyto­ The VN test performed in primary goose or Muscovy
pathic effect at 3–5 days postinfection, although several duck embryo fibroblast cell cultures is the most widely used
blind passages may be required before a detectable CPE method to detect the presence of antibodies (14, 29). Cross‐
is observed. In infected cultures Cowdry type‐A intra­ neutralization tests can be used to differentiate between
nuclear inclusions are often present. The presence of GPV and DPV antibodies (23). Duck‐origin GPV shows
the virus can be confirmed by immune‐­ staining or stronger cross‐reaction with GVP than with DPV (67). The
molecular methods (41). AGP test is less sensitive than the VN test and does not dif­
ferentiate between antibodies against GPV and DPV, but is
Direct Detection of Viral Antigens still useful method for testing large numbers of sera (14).
Immunofluorescence has been used to detect the pres­
ence of viral antigen in goslings, embryonated goose
Differential Diagnosis
eggs, and infected cell cultures. Flow cytometry assay
has been used to detect virus‐infected cells in the spleen. Goose parvovirus can be differentiated from DPV by using
Other methods, including immunoperoxidase tech­ serological and molecular methods. Very few pathogens of
niques, also have been developed (41). geese and ducks exist that show the strict age‐relatedness
Chapter 13  Viral Infections of Waterfowl 483

of waterfowl parvoviruses. The diseases to be considered vertically infected birds during hatching, the practice of
for differential diagnosis include duck virus enteritis, duck incubating and hatching eggs that have originated from
hepatitis, HNEG, reovirus, adenovirus, circovirus, and different breeding flocks should be avoided. Only eggs
Riemerella anatipestifer infections. from flocks with the same parvovirus status should be
The herpesvirus of duck viral enteritis produces dis­ incubated together and good hatchery hygiene should be
ease with high mortality in geese and ducks of all ages. maintained. The practice of breeding from parent stocks
Isolation and identification of the causal virus clearly dif­ that have survived the disease when young also should
ferentiate it from parvoviruses. Duck hepatitis viruses be discouraged, since these birds are potential carriers of
also cause fatal diseases in ducks under the age of six the virus.
weeks, but these viruses are not pathogenic for geese.
Hemorrhagic nephritis and enteritis of geese usually
Vaccination
affects geese from three weeks of age; however, the virus
generally causes only subclinical infection of ducks (8). Types of Vaccines
Mortality ranges from a few percentage points to Both live and killed oil emulsion vaccines, containing
50–70%. On postmortem, apart from enteritis and either whole inactivated virus or baculovirus expressed
nephritis, edema and hemorrhages of the subcutaneous VP2 capsid protein, are available and are widely used in
connective tissue and hydropericardium and ascites can countries where the disease is endemic. Vaccination
be seen (43). Diagnosis can be confirmed by the detec­ against Derzsy’s disease and SBDS relies on the use of
tion of GHPV with specific PCR (18, 43). attenuated live and inactivated GPV‐based vaccines,
Reovirus diseases have been described in young, 2‐ to while bivalent inactivated vaccines containing both GPV
6‐week‐old Muscovy duck and geese. The disease is and DPV antigens are used to ensure protection against
characterized by splenitis with miliary necrotic foci dur­ the two waterfowl parvoviruses causing disease in
ing the acute phase and epicarditis, arthritis, and teno­ Muscovy ducks (41). Recently a live attenuated parvovi­
synovitis during the subacute/chronic phase (37, 42). rosis vaccine, consisting of a live DPV strain, attenuated
Circovirus infection of geese and ducks may result in by passages on duck embryo cells has been developed.
growth retardation and feathering disorders without This vaccine induces a protective immunity against both
causing significant mortality. Histopathological exami­ GPV (Derzsy’s disease) and DPV (38).
nation shows lymphocyte depletion, necrosis, and histi­ Live vaccines containing attenuated goose and duck
ocytosis in the bursa of Fabricius. Globular or coarse parvovirus can stimulate rapid immune response and
granular inclusion bodies can be detected in the cells of protection in MDA‐free birds (41). Maternally‐derived
bursa follicles (53, 54). antibodies, even at a very low level, can neutralize the
Adenovirus infection causing hepatitis and hydroperi­ live vaccine, thus preventing the virus from stimulating
cardium in young (2–3 weeks old) geese is characterized immune responses. Inactivated vaccines containing the
by the accumulation of clear fluid in the dilated pericardial whole parvovirus antigens, either in the monovalent
sac and enlarged liver with multiple necrotic foci. (GPV) or bivalent (GPV and DPV) forms have the disad­
Histologically, basophilic intranuclear inclusion bodies vantage of being relatively slow to induce immune
can be found in the liver cells around the necrotic foci (21). response, but they are less sensitive to the interference
Riemerella anatipestifer may also cause high mortality with maternal antibodies than live vaccines (56).
in goslings and Muscovy ducklings. Treatment of birds Recombinant subunit vaccines have also been devel­
with appropriate antibiotics and isolation of the etiologic oped both from GPV and DPV using the baculovirus
agent in suitable media will enable differentiation from expression system (41). The structural protein(s)
waterfowl parvovirus. expressed were able to self‐assemble into virus‐like
In most cases the course of the disease, clinical signs and particles (VLP), and vaccines formulated from these
­
gross and histopathological lesions help to differentiate VLPs in oil emulsion proved to be comparable to inacti­
these diseases from those caused by waterfowl parvovi­ vated whole virus containing vaccines in Muscovy ducks
ruses; however, in questionable cases molecular methods (31) or geese (25).
should be used to detect and identify the causal agents (15).
Field Vaccination Protocols and Regimes
Historically, hyperimmune or convalescence serum
injected subcutaneously in day‐old goslings was used to
Intervention Strategies avoid heavy losses in flocks exposed to an early parvovi­
rus infection (12, 20). This technique was effective but
Management Procedures
presented the risk of carrying over undetected infectious
Because outbreaks of waterfowl parvovirus are fre­ agents by the contaminated serum. Currently, prophy­
quently attributed to transmission of the infection by laxis based on vaccination is the preferred choice.
484 Section II  Viral Diseases

An optimal vaccination strategy must take into account of virus transmission to the progenies, and (2) supplying
the presence or absence of MDA, their levels and hetero­ the progenies with passive immunity. An optimal vacci­
geneity within a flock, and the susceptible period of gos­ nation strategy must protect goslings and ducklings
lings and ducklings to the disease. Breeder geese and against both the early and the late forms of the disease.
ducks that have been naturally infected or vaccinated Therefore, knowing the antibody titers against GPV and
transfer MDA to their progeny, which may persist until DPV in parent flocks or the level of MDA in the day‐old
two to four weeks of age depending on the antibody lev­ birds is fundamental to establishing an adequate vacci­
els of individual birds at hatch. Because the disease is nation strategy. The development of an early immune
confined to young age, control measures have been response is crucial when the birds are reared at a farm
aimed at providing adequate immunity during the first contaminated with a parvovirus. Any gap between the
six to eight weeks of life. To achieve this, different meth­ waning of MDA and vaccination in a contaminated envi­
ods have been applied. These include: passive immuniza­ ronment is likely to lead to disease. In order to extend the
tion of newly hatched birds with convalescence or protection after MDA declines to nonprotective levels,
hyperimmune serum and the use of attenuated vaccine the vaccination of goslings and ducklings around one to
alone or in combination with an inactivated one for the two weeks of age is essential to stimulate active immu­
active immunization of both adult and young animals. nity in face of still persisting residual maternal antibod­
Immunization of breeders has two objectives: (1) pro­ ies. This can be best achieved by the use of inactivated
tection of breeders from infection, and thus prevention vaccine with high antigen content.

­References
Introduction 8 Chen, C.‐L., P.‐X. Wang, M.‐S. Lee, J.‐H. Shien, H.K.
Shieh, S.‐J. Ou, C.‐H. Chen, and P.‐C. Chang. 2006.
1 Austin, R.J., T.L. Whiting, R.A. Anderson, and M.A.
Development of a polymerase chain reaction procedure
Drebot. 2004. An outbreak of West Nile virus‐associated
for detection and differentiation of duck and goose
disease in domestic geese (Anser domesticus) upon
circovirus. Avian Dis. 50:92–95.
initial introduction to a geographic region, with
9 Chen, H., G.J. Smith, S.Y. Zhang, K. Qin, J. Wang, K.S.
evidence of bird to bird transmission. Can Vet J.
Li, R.G. Webster, J.S. Peiris, and Y. Guan. 2005. Avian
45:117–123.
flu: H5N1 virus outbreak in migratory waterfowl.
2 Banda, A., R. Galloway‐Haskins, T. Sandhu, and K.A.
Nature. 436:191–192.
Schat. 2006. Detection by PCR and nucleotide sequence
10 Cheng, A.C., M.S. Wang, X.Y. Chen, Y.F. Guo, Z.Y. Liu,
analysis of duck circovirus detected in Pekin ducks in
and P.F. Fang. 2001. Pathogenic and pathological
the United States. In: Convention Notes, AAAP/AVMA.
characteristic of new type gosling viral enteritis first
Honolulu, HI, AAAP.
observed in China. World J Gastroenterol. 7:678–684.
3 Banet‐Noach, C., L. Simanov, and M. Malkinson. 2003.
11 Delnatte P., C. Berkvens, M. Kummrow, D.A. Smith,
Direct (non‐vector) transmission of West Nile virus in
D. Campbell, G. Crawshaw, D. Ojkic, and J. DeLay.
geese. Avian Pathol. 32:489–494.
2011. New genotype of avian bornavirus in wild
4 Banyai, K., V. Palya, M. Benko, J. Bene, V. Havasi, B.
geese and trumpeter swans in Canada. Vet Rec.
Melegh, and G. Szucs. 2005. The goose reovirus genome
169:108.
segment encoding the minor outer capsid protein,
12 Dharmayanti, N.L., R. Hartawan R., Pudjiatmoko, H.
sigma1/sigmaC, is bicistronic and shares structural
Wibawa, A. Balish, R. Donis, C.T. Davis, and G.
similarities with its counterpart in Muscovy duck
Samaan. 2014. Genetic characterization of clade 2.3.2.1
reovirus. Virus Genes. 31:285–291.
avian influenza A(H5N1) viruses, Indonesia, 2012.
5 Bennett, R.S., R. LaRue, D. Shaw, Q. Yu, K.V. Nagaraja,
Emerg Infect Dis. 20:671–674.
D.A. Halvorson, and M.K. Njenga. 2005. A wild goose
13 Ellis, T.M., R.B. Bousfield, L.A. Bissett, K.C. Dyrting,
metapneumovirus containing a large attachment
G.S. Luk, S.T. Tsim, K. Sturm‐Ramirez, R.G. Webster, Y.
glycoprotein is avirulent but immunoprotective in
Guan, and J.S. Malik Peiris. 2004. Investigation of
domestic turkeys. J Virol. 79:14834–14842.
outbreaks of highly pathogenic H5N1 avian influenza
6 Bidin, M., I. Lojkic, Z. Bidin, M. Tisljar, D. Majnaric, and
in waterfowl and wild birds in Hong Kong in late 2002.
M. Mikec. 2011. Detection and characterization of avian
Avian Pathol. 33:492–505.
nephritis virus in ducklings. Avian Pathol. 40:173–177.
14 Glavits, R., E. Ferenczi, E. Ivanics, T. Bakonyi, T. Mato,
7 Bouquet, J.F., Y. Moreau, J.B. McFerran, and T.J. Connor.
P. Zarka, and V. Palya. 2005. Co‐occurrence of West
1982. Isolation and characterisation of an adenovirus
Nile Fever and circovirus infection in a goose flock in
isolated from Muscovy ducks. Avian Pathol. 11:301–307.
Hungary. Avian Pathol. 34:408–414.
Chapter 13  Viral Infections of Waterfowl 485

15 Gough, R.E., and W.R. Hansen. 2000. Characterization 26 Lanciotti, R.S., J.T. Roehrig, V. Deubel, J. Smith, M.
of a herpesvirus isolated from domestic geese in Parker, K. Steele, B. Crise, K.E. Volpe, M.B. Crabtree,
Australia. Avian Pathol. 29:417–422. J.H. Scherret, R.A. Hall, J.S. MacKenzie, C.B. Cropp, B.
16 Haider, N., K. Sturm‐Ramirez, S.U. Khan, M.Z. Panigrahy, E. Ostlund, B. Schmitt, M. Malkinson, C.
Rahman, S. Sarkar, M.K. Poh, H.L. Shivaprasad, M.A. Banet, J. Weissman, N. Komar, H.M. Savage, W. Stone,
Kalam, S.K. Paul, P.C. Karmakar, A. Balish, A. T. McNamara, and D.J. Gubler. 1999. Origin of the
Chakraborty, A.A. Mamun, A.B. Mikolon, C.T. Davis, West Nile virus responsible for an outbreak of
M. Rahman, R.O. Donis, J.D. Heffelfinger, S.P. Luby, encephalitis in the northeastern United States. Science.
and N. Zeidner. 2017. Unusually high mortality in 286:2333–2337.
waterfowl caused by highly pathogenic avian influenza 27 Lee, D.H., M.K. Torchetti, K. Winker, H.S. Ip, C.S.
A (H5N1) in Bangladesh. Transbound Emerg Dis. 64(1): Song, and D.E. Swayne. 2015. Intercontinental spread
144–156. of Asian‐origin H5N8 to North America through
17 Hollmen, T., J.C. Franson, M. Kilpi, D.E. Docherty, W.R. Beringia by migratory birds. J Virol. 89:6521–6524.
Hansen, and M. Hario. 2002. Isolation and 28 Li, C., C. Shen, A. Cheng, M. Wang, N. Zhang, Y. Zhou,
characterization of a reovirus from common eiders D. Zhu, R. Jia, Q. Luo, and X. Chen. 2010. Development
(Somateria mollissima) from Finland. Avian Dis. and application of an indirect immunoperoxidase assay
46:478–484. for the detection of duck swollen head hemorrhagic
18 Hollmen, T.E., J.C. Franson, P.L. Flint, J.B. Grand, R.B. disease virus antigen in Pekin ducks (Anas
Lanctot, D.E. Docherty, and H.M. Wilson. 2003. An platyrhynchos). J Vet Diagn Invest. 22:10–19.
adenovirus linked to mortality and disease in long‐ 29 Liu, Q., G. Zhang, Y. Huang, G. Ren, L. Chen, J. Gao, D.
tailed ducks (Clangula hyemalis) in Alaska. Avian Dis. Zhang, B. Han, W. Su, J. Zhao, X. Hu, and J. Su. 2011.
47:1434–1440. Isolation and characterization of a reovirus causing
19 Hollmen, T.E., J.C. Franson, M. Kilpi, D.E. Docherty, spleen necrosis in Pekin ducklings. Vet Microbiol.
and V. Myllys. 2003. An adenovirus associated with 148:200–206.
intestinal impaction and mortality of male common 30 Liu, S.N., X.X. Zhang, J.F. Zou, Z.J. Xie, Y.L. Zhu, Q.
eiders (Somateria mollissima) in the Baltic Sea. J Wildl Zhao, E.M. Zhou, and S.J. Jiang. 2010. Development of
Dis. 39:114–120. an indirect ELISA for the detection of duck circovirus
2 0 Hulse‐Post, D.J., K.M. Sturm‐Ramirez, J. Humberd, infection in duck flocks. Vet Microbiol. 145:41–46.
P. Seiler, E.A. Govorkova, S. Krauss, C. Scholtissek, 31 Lu, Y., Y. Dou, J. Ti, A. Wang, B. Cheng, X. Zhang, and
P. Puthavathana, C. Buranathai, T.D. Nguyen, H.T. Y. Diao. The effect of Tembusu virus infection in
Long, T.S. Naipospos, H. Chen, T.M. Ellis, Y. Guan, different week‐old Cherry Valley breeding ducks. 2016.
J.S. Peiris, and R.G. Webster. 2005. Role of Vet Microbiol. 192:167–174.
domestic ducks in the propagation and biological 32 McComb, B., R. Bennett, H.J. Shin, K.V. Nagaraja, F.J.
evolution of highly pathogenic H5N1 influenza Jirjis, and D.A. Halvorson. 2001. Wild waterfowl as a
viruses in Asia. Proc Natl Acad Sci USA. source of avian pneumovirus (APV) infection in
102:10682–10687. domestic poultry. In: 50th Western Poultry Disease
21 Ivanics, E., V. Palya, R. Glavits, A. Dan, V. Palfi, T. Conference. Davis, CA. 76–77.
Revesz, and M. Benko. 2001. The role of egg drop 33 Mishra N., S. Kalaiyarasu, S. Nagarajan, M.V. Rao, A.
syndrome virus in acute respiratory disease of goslings. George, R. Sridevi, S.P. Behera, S.C. Dubey, T.
Avian Pathol. 30:201–208. McCracken, S.H. Newman. 2012. Serological evidence
22 Jinding, C., L. Ming, R. Tao, and X. Chaoan. 2005. A of West Nile virus infection in wild migratory and
goose‐sourced paramyxovirus isolated from southern resident water birds in Eastern and Northern India.
China. Avian Dis. 49:170–173. Comp Immunol Microbiol Infect Dis. 35:591–598.
23 Kalaiyarasu S., N. Mishra, R.K. Khetan, and V.P. Singh. 34 Nagarajan, S., C. Tosh, D.K. Smith, J.S. Peiris, H.V.
2016. Serological evidence of widespread West Nile Murugkar, R. Sridevi, M. Kumar, M. Katare, R. Jain, Z.
virus and Japanese encephalitis virus infection in native Syed, P. Behera, C.L. Cheung, R. Khandia, S. Tripathi,
domestic ducks (Anas platyrhynchos var domesticus) Y. Guan, and S.C. Dubey. 2012. Avian influenza H5N1
in Kuttanad region, Kerala, India. Comp Immunol virus of clade 2.3.2 in domestic poultry in India. PLoS
Microbiol Infect Dis. 48:61–8. One. 7:e31844.
24 Ketterer, P.J., B.J. Rodwell, H.A. Westbury, P.T. Hooper, 35 Office International des Epizooties. 1999. West Nile
A.R. Mackenzie, J.G. Dingle, and H.C. Prior. 1990. fever in Israel: in geese. Dis Inform. 12:166–167.
Disease of geese caused by a new herpesvirus. Austral 36 Office International des Epizooties. 2000. West Nile
Vet J. 67:446–448. fever in the United States of America. Dis Inform. 13.
25 Komar, N. 2000. West Nile viral encephalitis. Rev Sci 37 Palya, V., R. Glavits, M. Dobos‐Kovacs, E. Ivanics, E.
Tech OIE. 19:166–176. Nagy, K. Banyai, G. Reuter, G. Szucs, A. Dan, and M.
486 Section II  Viral Diseases

Benko. 2003. Reovirus identified as cause of disease in West Nile virus infections in native and exotic birds
young geese. Avian Pathol. 32:129–138. during the 1999 outbreak in New York City, New York.
38 Payne S., L. Covaleda, G. Jianhua, S. Swafford, J. Vet Pathol. 37:208–224.
Baroch, P.J. Ferro, B. Lupiani, J. Heatley, and I. Tizard. 49 Sun, S., F. Chen, C. Sheng, L. Jiajia, L. Wen, L.
2011. Detection and characterization of a distinct Guangwei, S. Yongfeng, L. Junpeng, L. Chuang, Q.
bornavirus lineage from healthy Canada geese (Branta Jianping, and L. Haiyan. 2014. Isolation and
canadensis). J Virol. 85:12053–12056. characterization of a subtype C avian
39 Saito, M., Y. Osa, and M. Asakawa. 2009. Antibodies to metapneumovirus circulating in Muscovy ducks in
flaviviruses in wild ducks captured in Hokkaido, Japan: China. Vet Res. 45:74.
risk assessment of invasive flaviviruses. Vector Borne 50 Swayne, D.E., and D.L. Suarez. 2000. Highly
Zoonotic Dis. 9:253–258. pathogenic avian influenza. Rev Sci Tech OIE.
40 Savage, H.M., C. Ceianu, G. Nicolescu, N. Karabatsos, 19:463–482.
R. Lanciotti, A. Vladimirescu, L. Laiv, A. Ungureanu, C. 51 Toquin, D., M.H. Bayon‐Auboyer, N. Eterradossi and V.
Romanca, and T.F. Tsai. 1999. Entomologic and avian Jestin. 1999. Isolation of a pneumovirus from a
investigations of an epidemic of West Nile fever in Muscovy duck. Vet Rec. 145:680.
Romania in 1996, with serologic and molecular 52 Tsai, H.‐J., M.‐J. Pan, and Y.‐K. Liao. 1999. A serological
characterization of a virus isolate from mosquitoes. survey of ducks, geese, pigeons and cows for antibodies
Amer J Trop Med Hyg. 61:600–611. to Japanese B encephalitis virus in Taiwan. J Chin Soc
41 Seal, B.S., M.G. Wise, J.C. Pedersen, D.A. Senne, R. Vet Sci. 25:104–112.
Alvarez, M.S. Scott, D.J. King, Q. Yu, and D.R. 53 Turpin, E.A., D.E. Stallknecht, R.D. Slemons, L. Zsak,
Kapczynski. 2005. Genomic sequences of low‐virulence and D.E. Swayne. 2008. Evidence of avian
avian paramyxovirus‐1 (Newcastle disease virus) metapneumovirus subtype C infection of wild birds in
isolates obtained from live‐bird markets in North Georgia, South Carolina, Arkansas and Ohio, USA.
America not related to commonly utilized commercial Avian Pathol. 37:343–351.
vaccine strains. Vet Microbiol. 106:7–16. 54 Wen, G., M. Wang, H. Wang, L. Li, Q. Luo, T. Zhang,
42 Shi, S.H., Y. Huang, S.J. Cui, L.F. Cheng, G.H. Fu, X. Li, G. Cheng, and H. Shao. 2016. Genome sequence of a
Z. Chen, C.X. Peng, F. Lin, J.S. Lin, and J.L. Su. 2011. virulent genotype III Newcastle disease virus isolated
Genomic sequence of an avian paramyxovirus type 1 from laying ducks in China. Genome Announc.
strain isolated from Muscovy duck (Cairina moschata) 4(6):e01436‐16.
in China. Arch Virol. 156:405–412. 55 Yeh, J.Y., J.Y. Park, and E.N. Ostlund. 2011. Serologic
43 Shin, H.J., K.V. Nagaraja, B. McComb, D.A. Halvorson, evidence of West Nile virus in wild ducks captured in
F.F. Jirjis, D.P. Shaw, B.S. Seal, and M.K. Njenga. 2002. major inland resting sites for migratory waterfowl in
Isolation of avian pneumovirus from mallard ducks that South Korea. Vet Microbiol. 154:96–103.
is genetically similar to viruses isolated from 56 Zou, J., S. Shan, N. Yao, and Z. Gong. 2005. Complete
neighboring commercial turkeys. Virus Res. genome sequence and biological characterizations of a
83:207–212. novel goose paramyxovirus‐SF02 isolated in China.
44 Shin, H.J., M.K. Njenga, B. McComb, D.A. Halvorson, Virus Genes. 30:13–21.
and K.V. Nagaraja. 2000. Avian pneumovirus (APV)
RNA from wild and sentinel birds in the United States
Duck Hepatitis
has genetic homology with RNA from APV isolates
from domestic turkeys. J Clin Microbiol. 38:4282–4284. 1 Adamiker, D. 1970. Die Virushepatitis der EntenkUken
45 Smyth, J., D. Soike, D. Moffett, J.H. Weston, and D. im elektronenmikroskopischen Bild. Teil II: Befunde an
Todd. 2005. Circovirus‐infected geese studied by in situ der Milz und am Muskel. Zentralbl Veterinaermed (B).
hybridization. Avian Pathol. 34:227–232. 17:880–889.
46 Soike, D., K. Albrecht, K. Hattermann, C. Schmitt, and Adamiker, D. 1969. Elektronenmikroskopische
2
A. Mankertz. 2004. Novel circovirus in mulard ducks Untersuchungen zur Virushepatitis der EntenUken.
with developmental and feathering disorders. Vet Rec. Zentralbl Veterinaermed (B). 16:620–636.
154:792–793. Ahmed, A.A., Y.Z. El‐Abdin, S. Hamza, and F.E. Saad.
3
47 Soike, D., B. Koehler, and K. Albrecht. 1999. A 1975. Effect of experimental duck virus hepatitis
circovirus‐like infection in geese related to a runting infection on some biochemical constituents and
syndrome. Avian Pathol. 28:199–202. enzymes in the serum of white Pekin ducklings. Avian
48 Steele, K.E., M.J. Linn, R.J. Schoepp, N. Komar, T.W. Dis. 19:305–310.
Geisbert, R.M. Manduca, P.P. Calle, B.L. Raphael, T.L. Akulov, A.V., L.M. Kontrimavichus, and A.D.
4
Clippinger, T. Larsen, J. Smith, R.S. Lanciotti, N.A. Maiboroda. 1972. Sensitivity of geese to duck hepatitis
Panella, and T.S. McNamara. 2000. Pathology of fatal virus. Veterinariia. 48:47.
Chapter 13  Viral Infections of Waterfowl 487

5 Asplin, F.D. 1958. An attenuated strain of duck hepatitis 20 Chen, Y., W. Xiong, L. Zeng, Y. Wang, S. Zhang, M.
virus. Vet Rec. 70:1226–1230. Xu, M. Song, Y. Wang, H. Du, J. Liu, D. Wang, Y. Wu,
6 Asplin, F.D. 1965. Duck hepatitis: vaccination against and Y. Hu. 2014. Bush Sophora Root polysaccharide
two serological types. Vet Rec. 77:1529–1530. and its sulfate can scavenge free radicals resulted
7 Asplin, F.D. 1970. Examination of sera from wildfowl from duck virus hepatitis. Int J Biol Macromol.
for antibodies against the viruses of duck plague, duck 66:186–193.
hepatitis and duck influenza. Vet Rec. 87:182–183. 21 Chen, Y., L. Zeng, W. Xiong, M. Song, H. Du, Y. Wang,
8 Asplin, F.D. 1961. Notes on epidemiology and K. Ming, Y. Wu, D. Wang, Y. Hu, and J. Liu. 2015. Anti‐
vaccination for virus hepatitis of ducks. Off Int Epizoot duck virus hepatitis mechanisms of Bush Sophora Root
Bull. 56:793–800. polysaccharide and its sulfate verified by intervention
9 Balla, L., and T. Veress. 1984. Immunization experiments. Virus Res. 204:58–67.
experiments with a duck virus hepatitis vaccine. I 22 Cheng, A., M. Wang, H. Xin, D. Zhu, X. Li, H. Chen, R.
Antibody response of ducklings of different immune Jia, and M. Yang. 2009. Development and application of
status after subcutaneous, oral and aerosol vaccination. a reverse transcriptase polymerase chain reaction to
Magy Allatorv Lapja. 39:395–400. detect Chinese isolates of duck hepatitis virus type 1.
10 Bezrukavaya, I.J. 1978. Vaccine against duck virus J. Microbiol. Methods 77:1–5.
hepatitis from strain ZM. Abstr Landwirtsch Zentralbl 23 Crighton, G.W., and P.R. Woolcock. 1978. Active
Abt IV 1981: 1061.90–95. immunisation of ducklings against duck virus hepatitis.
11 Bosch, A., S. Guix, N.K. Krishna, E. Mendez, S.S. Vet Rec. 102:358–361.
Monroe, M. Pantin‐Jackwood, and S. Schultz‐Cherry. 24 Davis, D. 1987. Temperature and pH stability of duck
2011. Astroviridae. In: Virus Taxonomy. Classification hepatitis virus. Avian Pathol. 16:21–30.
and Nomenclature of Viruses: Ninth Report of the 25 Davis, D. 1987. Triple plaque purified strains of duck
International Committee on the Taxonomy of Viruses. hepatitis virus and their potential as vaccines. Res Vet
Elsevier Academic Press, London, United Kingdom. Sci. 43:44–48.
12 Calnek, B.W. 1993. Duck virus hepatitis. In: Virus 26 Davis, D., and D. Hannant. 1987. Fractionation of
Infections of Birds, Vol. 4, J.B. McFerran and M.S. neutralizing antibodies in serum of ducklings
McNulty, eds. Elsevier Science Publishers B.V., vaccinated with live duck hepatitis virus vaccine. Res
Amsterdam. 485–495. Vet Sci. 43:276–277.
13 Calnek, B.W. 1988. Personal communication. 27 Demakov, G.P., S.N. Ostashev, V.N. Ogordnikova, and
14 Cao, J., X. Ou, D. Zhu, G. Ma, A. Cheng, M. Wang, S. M.A. Shilov. 1975. Infection of brown rats with the
Chen, R. Jia, M. Liu, K. Sun, Q. Yang, Y. Wu, and X. duck hepatitus virus. Veterinariia. 57–58.
Chen. 2016. The 2A2 protein of duck hepatitis A virus 28 Ding, C., and D. Zhang. 2007. Molecular analysis of
type 1 induces apoptosis in primary cell culture. Virus duck hepatitis virus type 1. Virology., 361:9–17.
Genes. 52:780–788. 29 Doan, H.T., X.T. Le, R.T. Do, C.T. Hoang, K.T. Nguyen,
15 Chalmers, W.S., H. Farmer, and P.R. Woolcock. 1985. and T.H. Le. 2016. Molecular genotyping of duck
Duck hepatitis virus and Chlamydia psittaci outbreak hepatitis A viruses (DHAV) in Vietnam. J Infect Dev
[letter]. Vet Rec. 116:223. Ctries. 10:988–995.
16 Chen, L., M. Ma, R. Zhang, Q. Xu, X. Si, Y. Wang, Z. 30 Doroshko, I.N., and, I.Y. Bezrukavaya. 1975. Field trials
Xie, and S. Jiang. 2014. Simultaneous detection of duck of duck viral hepatitis vaccine. Veterinariya. 1:52–53.
hepatitis A virus types 1 and 3, and of duck astrovirus 31 Du, H., S. Zhang, M. Song, Y. Wang, L. Zeng, Y. Chen,
type 1, by multiplex RT‐PCR. Virol Sin. 29:196–198. W. Xiong, J. Yang, F. Yao, Y. Wu, D. Wang, Y. Hu and J.
17 Chen, L., Q. Xu, R. Zhang, J. Li, Z. Xie, Y. Wang, Y. Liu. 2016. Assessment of a flavone‐polysaccharide
Zhu, and S. Jiang. 2012. Complete genome sequence of based prescription for treating duck virus hepatitis.
a duck astrovirus discovered in eastern China. J Virol. PLoS One. 11:e0146046.
86:13833–13834. 32 Dvorakova, D., and Z. Kozusnik. 1970. The influence of
18 Chen, Y., M. Song, Y. Wang, W. Xiong, L. Zeng, S. temperature and some disinfectants on duck hepatitis
Zhang, M. Xu, H. Du, J. Liu, D. Wang, Y. Wu, and Y. virus. Acta Brno. 39:151–156.
Hu. 2015. The anti‐DHAV activities of Astragalus 33 Fabricant, J., C.G. Rickard, and P.P. Levine. 1957. The
polysaccharide and its sulfate compared with those of pathology of duck virus hepatitis. Avian Dis.
BSRPS and its sulfate. Carbohydr Polym. 117:339–345. 1:256–275.
19 Chen, Y., W. Xiong, L. Zeng, D. Wang, J. Liu, Y. Wu, 34 Fitzgerald, J.E., and L.E. Hanson. 1966. Certain
and Y. Hu. 2014. Comparison of Bush Sophora Root properties of a cell‐culture‐modified duck hepatitis
polysaccharide and its sulfate’s anti‐duck hepatitis A virus. Avian Dis. 10:157–161.
virus activity and mechanism. Carbohydr Polym. 35 Fitzgerald, J.E., L.E. Hanson, and J. Simon. 1969.
102:333–340. Histopathologic changes induced with duck hepatitis
488 Section II  Viral Diseases

virus in the developing chicken embryo. Avian Dis. 50 Guerin, J.‐L., V. Noutary, C. Boissieu, O. Albaric, and
13:147–157. M. Wyers. 2005. Viral pancreatitis and encephalitis of
36 Friend, M., and D.O. Trainer. 1972. Experimental Muscovy ducklings. Vet Rec. 157:328.
duck virus hepatitis in the mallard. Avian Dis. 51 Haider, S.A. 1980. Duck virus hepatitis. In: Isolation
16:692–699. and Identification of Avian Pathogens, S.B. Hitchner,
37 Fu, Y., M. Pan, W. Wang, Y. Xu, X. Xie, K.N.J., H. Yang, C.H. Domermuth, H.G. Purchase, and J.E. Williams,
and D. Zhang. 2009. Complete sequence of a duck eds. American Association of Avian Pathologists,
astrovirus associated with fatal hepatitis in ducklings., Kennett Square, PA. 75–76.
J Gen Virol. 90:1104–1108. 52 Haider, S.A., and B.W. Calnek. 1979. In vitro isolation,
38 Fu, Y., M. Pan, X. Wang, Y. Xu, H. Yang and D. Zhang. propagation, and characterization of duck hepatitis
2008. Molecular detection and typing of duck hepatitis virus type III. Avian Dis. 23:715–729.
A virus directly from clinical specimens. Vet Microbiol. 53 Hanson, L.E., H.E. Rhoades, and R.L. Schricker. 1964.
131:247–257. Properties of duck hepatitis virus. Avian Dis.
39 Gazdzinski, P. 1979. Attenuation of duck hepatitis virus 8:196–202.
and evaluation of its usefulness for duckling 54 Hanson, L.E., and D.N. Tripathy. 1976. Oral
immunization. I Studies on attenuation of the virus. immunization of ducklings with attenuated duck
Bull Vet Inst Pulawy. 23:80–89. hepatitis virus. Dev Biol Stand. 33:357–363.
40 Gazdzinski, P. 1979. Attenuation of duck hepatitis virus 55 Hu, Q., D. Zhu, G. Ma, A. Cheng, M. Wang, S. Chen, R.
and evaluation of its usefulness for duckling Jia, M. Liu, K. Sun, Q. Yang, Y. Wu, and X. Chen. 2016.
immunization. II. Studies on application of the A one‐step duplex rRT‐PCR assay for the simultaneous
attenuated strain of DVH for vaccination of ducklings. detection of duck hepatitis A virus genotypes 1 and 3.
Bull Vet Inst Pulawy. 23:89–98. J Virol Methods. 236:207–214.
41 Golubnichi, V.P., G.P. Tishchenko, and V.I. Korolkov. 56 Hwang, J. 1965. A chicken‐embryo‐lethel strain of duck
1976. Preparation of tissue culture antigens of duck hepatitis virus. Avian Dis. 9:417–422.
hepatitis virus. Abstr Landwirtsch Zentralbl Abt IV 57 Hwang, J. 1969. Duck hepatitis virus‐neutralization test
1977: 2251. 14:88–90. in chicken embryos. Am J Vet Res. 30:861–864.
42 Gough, R.E. 1986. Duck hepatitis type 2 associated 58 Hwang, J. 1965. Duck hepatitis virus in duck embryo
with an astrovirus. In: Acute Virus Infections of Poultry, fibroblast cultures. Avian Dis. 9:285–290.
J.B. McFerran and M.S. McNulty, eds. Martinus 59 Hwang, J. 1970. Immunizing breeder ducks with
Nijhoff, Dordrecht. 223–230. chicken embryo‐propagated duck hepatitis virus for
43 Gough, R.E. 2001. personal communication. production of parental immunity in their progenies.
44 Gough, R.E., E.D. Borland, I.F. Keymer, and J.C. Am J Vet Res. 31:805–807.
Stuart. 1985. An outbreak of duck hepatitis type 60 Hwang, J. 1974. Susceptibility of poultry to duck
II in commercial ducks. Avian Pathol. hepatitis viral infection. Am J Vet Res. 35:477–479.
14:227–236. 61 Hwang, J., and I.E. Dougherty. 1964. Distribution and
45 Gough, R.E., M.S. Collins, E. Borland, and L.F. Keymer. concentration of duck hepatitis virus in inoculated
1984. Astrovirus‐like particles associated with hepatitis ducklings and chicken embryos. Avian Dis. 8:264–268.
in ducklings. Vet Rec. 114:279. 62 Kaleta, E.F. 1988. Duck viral hepatitis type 1
46 Gough, R.E., and D. Spackman. 1981. Studies with vaccination: monitoring of the immune response with a
inactivated duck virus hepatitis vaccines in breeder microneutralization test in Pekin duck embryo kidney
ducks. Avian Pathol. 10:471–479. cell cultures. Avian Pathol. 17:325–332.
47 Gough, R.E., and J.C. Stuart. 1993. Astroviruses in 63 Kaszanyitzky, E.J., and J. Tanyi. 1980. Studies on the
ducks (duck virus hepatitis type II). In: Virus Infections laboratory diagnosis and epizootiology of duck virus
of Birds, Vol. 4, J.B. McFerran and M.S. McNulty, eds. hepatitis. Magy Allatorv Lapja. 35:808–814.
Elsevier Science Publishers, B.V., Amsterdam. 64 Kim, M.C., M.J. Kim, Y.K. Kwon, A.M. Lindberg, S.J.
505–508. Joh, H.M. Kwon, Y.J. Lee, and J.H. Kwon. 2009.
48 Gough, R.E., and A.S. Wallis. 1986. Duck hepatitis type Development of duck hepatitis A virus type 3 vaccine
I and influenza in mallard ducks (Anas platyrhynchos). and its use to protect ducklings against infections.
Vet Rec. 119:602. Vaccine. 27:6688–6694.
49 Guérin, J.‐L., O. Albaric, V. Noutary, and C. Boissieu. 65 Kim, M.C., Y.K. Kwon, S.J. Joh, S.J. Kim, C. Tolf, J.H.
2007. A duck hepatitis virus type 1 is agent of Kim, H.W. Sung, A.M. Lindberg, and J.H. Kwon. 2007.
pancreatitis and encephalitis in Muscovy duckling. In: Recent Korean isolates of duck hepatitis virus reveal
Proceedings of the 147th AVMA/ 50th American the presence of a new geno‐ and serotype when
Association of Avian Pathologists Conference. compared to duck hepatitis virus type 1 type strains.
Washington, DC. 4585. Arch Virol. 152:2059 ‐2072.
Chapter 13  Viral Infections of Waterfowl 489

66 Kim, M.C., Y.K. Kwon, S.J. Joh, J.H. Kwon, J.H. Kim, 80 Ma J., C.T. and H. Yue. 2014. Diagnosis of
and S.J. Kim. 2007. Development of one‐step reverse simultaneously infection of duck hepatitis A virus
transcriptase‐polymerase chain reaction to detect duck genotype A and duck hepatitis A virus genotype C in
hepatitis virus type 1. Avian Dis. 51:540–545. duckling Journal of Southwest University for
67 Kim, M.C., Y.K. Kwon, S.J. Joh, J.H. Kwon, and Nationalities, Natural Science Edition. 40:192–195.
A.M. Lindberg. 2008. Differential diagnosis between 81 Maiboroda, A.D. 1972. Formation of duck hepatitis
type‐specific duck hepatitis virus type 1 (DHV‐1) virus in culture cells. Veterinariya. (8):50–52.
and Korean DHV‐1‐like isolates using a multiplex 82 Mennella, G.R, and G. Mandelli. 1977. Glutamic‐
polymerase chain reaction. Avian Pathol. oxaloacetic (GOT) and glutamic‐pyruvic (GPT)
37:171–177. transaminases in the blood serum in experimental viral
68 Kim, M.C., Y.K. Kwon, S.J. Joh, A.M. Lindberg, J.H. hepatitis of ducklings. Arch Vet Ital. 28:187–190.
Kwon, J.H. Kim, and S.J. Kim. 2006. Molecular analysis 83 Ming, K., Y. Chen, F. Yao, J. Shi, J. Yang, H. Du, X.
of duck hepatitis virus type 1 reveals a novel lineage Wang, Y. Wang, and J. Liu. 2017. Phosphorylated
close. J Gen Virol. 87:3307–3316. Codonopsis pilosula polysaccharide could inhibit the
69 Korolkov, V.I., V.P. Golubnichi, P.S. Khandogin, and virulence of duck hepatitis A virus compared with
M.A. Karvus. 1979. Aerosol vaccination method for Codonopsis pilosula polysaccharide. Int J Biol
virus hepatitis in ducklings. Abstr Landwirtsch Macromol. 94:28–35.
Zentralbl Abt IV 1977: 2251. 17:82–83. 84 Monroe, S.S., M.J. Carter, J. Herrmann, D.K. Mitchel,
70 Levine, P.P., and J. Fabricant. 1950. A hitherto‐ and A. Sanchez‐Fauquier. 2005. Family Astroviridae.
undescribed virus disease of ducks in North America. In: Virus Taxonomy Eighth Report of the
Cornell Veterinarian. 40:71–86. International Committee on Taxonomy of Viruses.
71 Levine, P.P., and M.S. Hofstad. 1945. Duck disease C.M. Faquet, M.A. Mayo, J. Maniloff, U. Desselberger,
investigation. Annual Report of the New York State and L.A. Ball, eds. Elsevier Academic Press, London.
Veterinary College, Ithaca. 55–56. 859–864.
72 Li, C., Z. Chen, C. Meng, and G. Liu. 2014. Rapid 85 Nikitin, M.G., I.I. Panikar, and V.V. Garkavaya. 1976.
detection of duck hepatitis A virus genotype C using Aerosol immunization against duck virus hepatitis.
reverse transcription loop‐mediated isothermal Vestn Skh Nauki. (1):124–126.
amplification. J Virol Methods. 196:193–198. 86 Panikar, I., and V. Gostrik. 1981. Aerosol vaccination of
73 Li, X., R. Zhao, W. Lin, C. Li, T. Zhang, F. Meng, M. ducklings against duck virus hepatitis. Abstr
Liu, and Y. Zhang. 2017. Evidence of VP1 of duck Landwirtsch Zentralbl Abt IV 1982. 1475.35.
hepatitis A type 1 virus as a target of neutralizing 87 Peng, N.X. 1998. Dynamic pathological study on
antibodies and involving receptor‐binding activity. viral hepatitis in ducklings. Chinese J Vet Sci Tech.
Virus Res. 227:240–244. 28:22–23.
74 Liao, Q., L. Zheng, Y. Yuan, J. Shi, and D. Zhang. 2014. 88 Pollard, M., and T.J. Starr. 1959. Propagation of duck
Genomic characterization of a novel picornavirus in hepatitis virus in tissue culture. Proc Soc Exp Biol Med.
Pekin ducks. Vet Microbiol. 172:78–91. 101:521–524.
75 Liu, G., F. Wang, Z. Ni, T. Yun, B. Yu, J. Huang, and J. 89 Polyakov, A.A., and G.D. Volkovskii. 1969. Survival of
Chen. 2008. Genetic diversity of the VP1 gene of duck duckling hepatitis virus outside the host and methods
hepatitis virus type I (DHV‐I) isolates from southeast of disinfection. Abstr Vet Bull. 34:278–290.
China is related to isolate attenuation. Virus Res. 90 Priz, N.N. 1973. Comparative study of virus hepatitis in
137:137–141. animals (dogs and ducks) using different routes of
76 Liu, M., Fanyi‐Meng, X.J. Li, Z. Zhang, S. Liu, and Y. influence. Abstr Vet Bull. 44(2746):696–700.
Zhang. 2011. Goose haemorrhagic hepatitis caused by 91 Rahn, D.P. 1962. Susceptibility of Turkeys to Duck
a new subtype duck hepatitis type 1 virus. Vet Hepatitis Virus and Turkey Hepatitis Virus, University
Microbiol. 152(2–3):280–283. of Illinois.
77 Liu, M., T. Zhang, Y. Zhang, F. Meng, X. Li, Z. Hou, X. 92 Rao, S.B., and B.R. Gupta. 1967. Studies on a filterable
Feng, and X. Kong. 2010. Development and evaluation agent causing hepatitis in ducklings, and biliary
of a VP1‐ELISA for detection of antibodies to duck cirrhosis and blood dyscrasia in adults. IndJ Poult Sci.
hepatitis type 1 virus. J Virol Methods. 169:66–69. 2:18–30.
78 Liu, N., F. Wang, and D. Zhang. 2014. Complete 93 Reuss, U. 1959. Versuche zur aktiven und passiven
sequence of a novel duck astrovirus. Arch Virol. Immunisierung bei der Virushepatitis der Entenkuken.
159:2823–2827. Zentralbl Veterinaermed. 6:808–815.
79 Luff, P.R., and I.G. Hopkins. 1986. live duck virus 94 Reuss, U. 1959. Virusbiologische untersuchungen bei
hepatitis vaccination of maternally immune ducklings. der Entenhepatitis. Zentralbl Veterinaermed.
Vet Rec. 119:502–503. 6:209–248.
490 Section II  Viral Diseases

95 Richter, W.R., E.J. Rozok, and S.M. Moize. 1964. 109 Todd, D., V.J. Smyth, N.W. Ball, B.M. Donnelly, M.
Electron microscopy of virus‐like particles associated Wylie, N.J. Knowles, and B.M. Adair. 2009.
wit duck viral hepatitis. Virology. 24:114–116. Identification of chicken enterovirus‐like viruses,
96 Rispens, B.H. 1969. Some aspects of control of duck hepatitis virus type 2 and duck hepatitis virus
infectious hepatitis in ducklings. Avian Dis. type 3 as astroviruses. Avian Pathol. 38:21–30.
13:417–426. 110 Toth, T.E. 1975. Duck virus hepatitis. In: Isolation and
97 Roszkowski, J., W. Kozaczynski, and P. Gazdzinski. Identification of Avian Pathogens, S.B. Hitchner, C.H.
1980. Effect of attenuation on the pathogenicity of Domermuth, H.G. Purchase and J.E. Williams, eds.
duck hepatitis virus, histopathological study. Bull Vet American Association of Avian Pathologists, College
Inst Pulawy. 24:41–48. Station, TX. 192–196.
98 Sandhu, T.S., B.W. Calnek, and L. Zeman. 1992. 111 Toth, T.E. 1969. Studies of an agent causing mortality
Pathologic and serologic characterization of a variant among ducklings immune to duck virus hepatitis.
of duck hepatitis type I virus. Avian Dis. 36:932–936. Avian Dis. 13:834–846.
99 Schoop, G., H. Staub, and K. Erguney. 1959. Virus 112 Toth, T.E., and N.L. Norcross. 1981. Humoral immune
hepatitis of ducks. V. Attempted adaptation of the virus response of the duck to duck hepatitis virus: virus‐
to chicken embryos. Monatsh Tierheilkd. 11:99–106. neutralizing vs. virus‐precipitating antibodies. Avian
100 Shalaby, M.A., M.N. K. Ayoub, and I.M. Reda. 1978. A Dis. 25:17–28.
study on a new isolate of duck hepatitis virus and its 113 Tseng, C.H., N.J. Knowles, and H.J. Tsai. 2007.
relationship to other duck hepatitis virus strains. Vet Molecular analysis of duck hepatitis virus type 1
Med J Cairo Univ. 26:215–221. indicates that it should be assigned to a new genus.
101 Shen, Y., A. Cheng, M. Wang, S. Chen, R. Jia, D. Zhu, Virus Res. 123:190–203.
M. Liu, K. Sun, Q. Yang, and X. Chen. 2015. 114 Tseng, C.H., and H.J. Tsai. 2007. Molecular
Development of an indirect ELISA method based on characterization of a new serotype of duck hepatitis
the VP3 protein of duck hepatitis A virus type 1 virus. Virus Res. 126:19–31.
(DHAV‐1) for dual detection of DHAV‐1 and 115 Tseng, C.H., and H.J. Tsai. 2007. Sequence analysis of
DHAV‐3 antibodies. J Virol Methods. 225:30–34. a duck picornavirus isolate indicates that it together
102 Sheng, X.D., W.P. Zhang, Q.R. Zhang, C.Q. Gu, X.Y. with porcine enterovirus type 8 and simian
Hu, and G.F. Cheng. 2014. Apoptosis induction in picornavirus type 2 should be assigned to a new
duck tissues during duck hepatitis A virus type 1 picornavirus genus. Virus Res. 129:104–114.
infection. Poult Sci. 93:527–534. 116 Ulbrich, F. 1971. Significance of wild ducks in the
103 Soliman, M., M.M. Alfajaro, M.H. Lee, Y.J. Jeong, D.S. transmission of duck viral hepatitis. Monatsh
Kim, K.Y. Son, J. Kwon, J.S. Choi, J.S. Lim, J.S. Choi, Veterinaermed. 26:629–631.
T.U. Lee, K.O. Cho, and M.I. Kang. 2015. The 117 Vertinskii, K.I., B.F. Bessarabov, A.N. Kurilenko, A.P.
prevalence of duck hepatitis A virus types 1 and 3 on Strelnikov, and P.M. Makhno. 1968. Pathogensis and
Korean duck farms. Arch Virol. 160:493–498. diagnosis of duck viral hepatitis. Veterinariya. 7:27–30.
104 Song, C., Y. Liao, W. Gao, S. Yu, Y. Sun, X. Qiu, L. Tan, 118 Wang, L., M. Pan, Y. Fu, and D. Zhang. 2008.
A. Cheng, M. Wang, Z. Ma, and C. Ding. 2014. Classification of duck hepatitis virus into three
Virulent and attenuated strains of duck hepatitis A genotypes based on molecular evolutionary analysis.
virus elicit discordant innate immune responses in Virus Genes. 37:52–59.
vivo. J Gen Virol. 95:2716–2726. 119 Wang, W., A. Said, Y. Wang, Q. Fu, Y. Xiao, S. Lv, and
105 Song, C., H. Wan, S. Yu, X. Han, X. Qiu, Q. Hu, L. Tan, Z. Shen. 2016. Establishment and characterization of
and C. Ding. 2012. Rapid detection of duck hepatitis duck embryo epithelial (DEE) cell line and its use as a
virus type‐1 by reverse transcription loop‐mediated new approach toward DHAV‐1 propagation and
isothermal amplification. J Virol Methods. 182:76–81. vaccine development. Virus Res. 213:260–268.
106 Sung H.W., Kim, J.H., Song, C.S., Han, M.K., Lee, Y.J., 120 Wang, X., N. Liu, F. Wang, K. Ning, Y. Li, and D.
and Mo, I.P. 2000. Development of a live vaccine strain Zhang. 2014. Genetic characterization of a novel
of duck viral hepatitis using a Korean isolate. Kor J Vet duck‐origin picornavirus with six 2A proteins. J Gen
Res. 40:106–110. Virol. 95:1289–1296.
107 Syurin, V.N., I.I. Panikar, and I.M. Shchetinskii. 1977. 121 Wang, X., Y. Wang, X. Xie, B. Zhang, and D. Zhang.
Immunogenesis and pathogenesis of viral hepatitis of 2011. Expression of the C‐terminal ORF2 protein of
ducks. Veterinariya. 53–55. duck astrovirus for application in a serological test.
108 Tauraso, N.M., G.E. Coghill, and M.J. Klutch. 1969. J Virol Methods. 171:8–12.
Properties of the attenuated vaccine strain of duck 122 Wei, C.Y., S. Su, Z. Huang, W.J. Zhu, J.D. Chen, F.R.
hepatitis virus. Avian Dis. 13:321–329. Zhao, Y.J. Wang, J.X. Xie, H. Wang, and G. Zhang.
Chapter 13  Viral Infections of Waterfowl 491

2012. Complete genome sequence of a novel duck 137 Xiong, W., X. Ma, Y. Wu, Y. Chen, L. Zeng, J. Liu, W.
hepatitis A virus discovered in southern China. Sun, D. Wang, and Y. Hu. 2015. Determine the
J Virol. 86:10247. structure of phosphorylated modification of icariin
123 Wen, X.J., A.C. Cheng, M.S. Wang, R.Y. Jia, D.K. Zhu, and its antiviral activity against duck hepatitis virus A.
S. Chen, M.F. Liu, F. Liu, and X.Y. Chen. 2014. BMC Vet Res. 11:205.
Detection, differentiation, and VP1 sequencing of 138 Xu, Q., R. Zhang, L. Chen, L. Yang, J. Li, P. Dou, H.
duck hepatitis A virus type 1 and type 3 by a 1‐step Wang, Z. Xie, Y. Wang, and S. Jiang. 2012. Complete
duplex reverse‐transcription PCR assay. Poult Sci. genome sequence of a duck hepatitis a virus type 3
93:2184–2192. identified in eastern China. J Virol. 86:13848.
124 Wildy, P. 1971. Classification and nomenclature of 139 Yang, M., A. Cheng, M. Wang, and H. Xing. 2008.
viruses. Monographs in Virology. 5:1–118. Development and application of a one‐step real‐time
125 Woolcock, P. 2010. Duck virus hepatitis. In: Manual Taqman RT‐PCR assay for. J Virol Methods.
of Diagnostic Tests and Vaccines for Terrestrial 153:55–60.
Animals. Office International des Epizooties, Paris, 140 Yao, F., Y. Chen, J. Shi, K. Ming, J. Liu, W. Xiong, M.
France. Song, H. Du, Y. Wang, S. Zhang, Y. Wu, D. Wang, and
126 Woolcock, P. 2008. Duck virus hepatitis. In: Diseases Y. Hu. 2016. Replication cycle of duck hepatitis A
of Poultry, 12th, Y.M. Saif, A.M. Fadly, J.R. Glisson, virus type 1 in duck embryonic hepatocytes. Virology.
L.R. McDougald, L.K. Nolan, and D.E. Swayne, eds. 491:73–78.
Blackwell, Ames, IA. 373. 141 Zell, R., Delwart, E., Gorbalenya, A.E., Hovi, T., King,
127 Woolcock, P.R. 1986. An assay for duck hepatitis virus A.M.Q., Knowles, N.J., Lindberg, A.M., Pallansch,
type I in duck embryo liver cells and a comparison M.A., Palmenberg, A.C., Reuter, G., Simmonds, P.,
with other assays. Avian Pathol. 15:75–82. Skern, T., Stanway, G., Yamashita, T., and ICTV
128 Woolcock, P.R. 2008. Duck hepatitis. In: A Laboratory Report Consortium. 2017. ICTV virus taxonomy
Manual for the Isolation, Identification and profile: Picornaviridae. J Gen Virol. 98:2421–2422.
Characterization of Avian Pathogens, 5th, L. Dufour‐ 142 Zhao, X.L., R.M. Phillips, G.D. Li, and A.Q. Zhong.
Zavala, D.E. Swayne, J.R. Glisson, J.E. Pearson, W.M. 1991. Studies on the detection of antibody to duck
Reed, M.W. Jackwood, and P.R. Woolcock, eds. hepatitis virus by enzyme‐linked immunosorbent
American Association of Avian Pathologists, assay. Avian Dis. 35:778–782.
Jacksonville, FL. 175–178. 143 Zou, Z., J. Ma, K. Huang, H. Chen, Z. Liu, and M. Jin.
129 Woolcock, P.R. 1991. Duck hepatitis virus type I: 2016. Live attenuated vaccine based on duck enteritis
Studies with inactivated vaccines in breeder ducks. virus against duck hepatitis A virus types 1 and 3.
Avian Pathol. 20:509–522. Front Microbiol. 7:1613.
130 Woolcock, P.R. 2016. Overview of duck viral hepatitis. 144 Zubtsova, R.A. 1971. Laboratory trial of live vaccine
http://www.cdc.gov/flu/avian/gen‐info/avian‐flu‐ against duck hepatitis containig GNKI attenuated
humans.htm. strains. Abstr Vet Bull 42: 1870. 17:127–132.
131 Woolcock, P.R. 1995. unpublished data.
132 Woolcock, P.R., W.S. K. Chalmers, and D. Davis. 1982.
Duck Virus Enteritis (Duck Plague)
A plaque assay for duck hepatitis virus. Avian Pathol.
11:607–610. 1 Ahamed, M.M., and M.T. Hossain. 2015. Molecular
133 Woolcock, P.R., and G.W. Crighton. 1979. Duck virus characterization of duck plague virus isolated
hepatitis: serial passage of attenuated virus in from Bangladesh. J Adv Vet Anim Res. 1.
ducklings. Vet Rec. 105:30–32. 2 Aravind, S., and N.M. Kamble. 2015. Adaptation and
134 Woolcock, P.R., and G.W. Crighton. 1981. Duck virus growth kinetics study of an Indian isolate of virulent
hepatitis: the effect of attenuation on virus stability in duck enteritis virus in Vero cells. Microb Pathog.
ducklings. Avian Pathol. 10:113–119. 78:14–19.
135 Woolcock, P.R., and J. Fabricant. 1991. Duck virus 3 Aravind, S., and N.M. Kamble. 2015. Protective effects of
hepatitis. In: Diseases of Poultry, 9th, B.W. Calnek, recombinant glycoprotein D based prime boost
H.J. Barnes, C.W. Beard, W.M. Reid, and H.W. approach against duck enteritis virus in mice model.
Yoder, eds. Iowa State University Press, Ames, IA. Microb Pathog. 88:78–86.
597–608. 4 Baudet, A.E. 1923. Mortality in ducks in the Netherlands
136 Woolcock, P.R., and H.J. Tsai. 2013. Duck hepatitis. In: caused by a filtrable virus; fowl plague. Tijdschr
Diseases of Poultry, D.E. Swayne, J.R. Glisson, L.R. Diergeneeskd. 50:455–459.
McDougald, L.K. Nolan, D.L. Suarez, and V.L. Nair, 5 Bos, A. 1942. Some new cases of duck plague. Tijdschr
eds. John Wiley & Sons. 422–431. Diergeneeskd. 69:372–381.
492 Section II  Viral Diseases

6 Burgess, E.C., and J. Ossa. 1979. Duck plague: a carrier virus in duck embryo fibroblasts. Bing Du Xue Bao.
state in waterfowl. Avian Dis. 23:940. 24:352–357.
7 Burgess, E.C., and T.M. Yuill. 1981. Increased cell 23 Guo, Y.F., and A.C. Cheng. 2005. Morphological
culture incubation temperatures for duck plague virus observation of virulent duck enteritis virus strain CH‐
isolation. Avian Dis. 25:222. Infected duck embryo fibroblasts by transmission
8 Burgess, E.C., and T.M. Yuill. 1982. Superinfection in electron microscopy. Chinese J Vet Sci. 25(6):632–635.
ducks persistently infected with duck plague virus. 24 Hess, W.R., and A.H. Dardiri. 1968. Some properties of
Avian Dis. 26:40–46. the virus of duck plague. Arch Virol. 24:148.
9 Burgess, E.C., and T.M. Yuill. 1983. The influence of 25 Hu, Y., and X. Liu. 2013. Glycoprotein C plays a role in
seven environmental and physiological factors on duck the adsorption of duck enteritis virus to chicken
plague virus shedding by carrier mallards. Journal of embryo fibroblasts cells and in infectivity. Virus Res.
Wildlife DiseasesJ Wildl Dis. 19:77. 174:1–7.
10 Chen, P., and J. Liu. 2014. The vaccine efficacy of 26 Huang, J., and R. Jia. 2014. An attenuated duck plague
recombinant duck enteritis virus expressing secreted E virus (DPV) vaccine induces both systemic and
with or without PrM proteins of duck tembusu virus. mucosal immune responses to protect ducks against
Vaccine. 32:5271–5277. virulent DPV infection. Clin Vaccine Immunol. 21:457.
11 Cheng, A.C., and Y.H. Liao. 2008. Replication of duck 27 Jansen, J. 1964. Duck plague (a concise survey). Ind Vet
plague virus in artificially infected ducks detected by in J. 41:309–316.
situ hybridization. Chin J Virol. 24:72–75. 28 Jansen, J. 1968. Duck plague. J Am Vet Med Assoc.
12 Dardiri, A.H., and W.R. Hess. 1967. The incidence of 152:1009.
neutralizing antibodies to duck plague virus in 29 Jansen, J. 1949. Is duck plague related to Newcastle
serums from domestic ducks and wild waterfowl in disease or to fowl plague. Int Vet Congr Rep.
the United States of America. Proceedings Annual 14:363–365.
Meeting of the United States Animal Health 30 Ji, J., and L.Q. Du. 2009. Rapid diagnosis of duck
Association. 71:225. plagues virus infection by loop‐mediated isothermal
13 Dardiri, A.H., and W.R. Hess. 1968. A plaque assay for amplification. Res Vet Sci. 87:53–58.
duck plague virus. Can J Vet Rev. 32:505–510. 31 Jia, R., and A. Cheng. 2009. Development and
14 Deng, M.Y., and E.C. Burgess. 1984. Detection of duck evaluation of an antigen‐capture ELISA for detection of
plague virus by reverse passive hemagglutination test. the UL24 antigen of the duck enteritis virus, based on a
Avian Dis. 28:616. polyclonal antibody against the UL24 expression
15 Dhama, K., and N. Kumar. 2017. Duck virus enteritis protein. J Virol Methods. 161:38–43.
(duck plague)–a comprehensive update. Vet Quart. 32 Jing, Y., W.U. Ying, K. Sun, et al. 2017. Role of duck
37:57–80. plague virus glycoprotein C in viral adsorption: absence
16 El‐Samadony, H.A., and L.A. Tantawy. Molecular of specific interactions with cell surface heparan
characterization of circulating duck viral enteritis in sulfate. J Integr Agric. 16:1145–1152.
Egypt during 2012–2013. Brit J Poult Sci. 2(3):38–44. 33 Jr, B.S., and A.H. Dardiri. 1968. Electron microscopic
17 Friend, M., and G.L. Pearson. 1973. Duck plague (duck characterization of duck plague virus. Virology.
virus enteritis) in wild waterfowl. Center for Integrated 34:160–169.
Data Analytics Wisconsin Science Center. 34 Kaleta, E.F., and A. Kuczka. 2007. Outbreak of duck
18 Goldberg, D.R., and T.M. Yuill. 1990. Mortality from plague (duck herpesvirus enteritis) in numerous species
duck plague virus in immunosuppressed adult mallard of captive ducks and geese in temporal conjunction with
ducks. J Wildl Dis. 26:299. enforced biosecurity (in‐house keeping) due to the threat
19 Gough, R.E., and W.R. Hansen. 2000. Characterization of avian influenza A virus of the subtype Asia H5N1.
of a herpesvirus isolated from domestic geese in Dtw Deutsche Tierrztliche Wochenschrift. 114:3–11.
Australia. Avian Pathol. 29:417–422. 35 Kennedy, P.G., and J. Rovnak. 2015. A comparison of
20 Guo, Y.‐f., and A.‐c. Cheng. 2006. Studies on herpes simplex virus type 1 and varicella‐zoster virus
morphogenesis of duck enteritis virus CH virulent latency and reactivation. J Gen Virol. 96:1581–1602.
isolate in infected duck embryo fibroblasts. Acta 36 Ketterer, P.J., and B.J. Rodwell. 1990. Disease of geese
Veterinaria et Zootechnica Sinica. 37:274. caused by a new herpesvirus. Aust Vet J. 67:446.
21 Guo, Y.F., and A.C. Cheng. 2006. Establishment and 37 Keymer, I.F., and R.E. Gough. 1986. Duck virus enteritis
application of quantitative real time PCR assay for (anatid herpesvirus infection) in mute swans (Cygnus
detecting duck viral enteritis virus. Veterinary Science olor). Avian Pathol 15:161–170.
in China. 36:444–448. 38 Lam, K.M. 1984. Antibody‐ and complement‐mediated
22 Guo, Y.F., and A.C. Cheng. 2008. Studies on the cytolysis against duck‐enteritis‐virus‐infected cells.
propagation characteristics of duck plague virulent Avian Dis. 28:1125.
Chapter 13  Viral Infections of Waterfowl 493

39 Leibovitz, L. 1969. The comparative pathology of duck 56 Proctor, S.J. 1975. Pathogenesis of digestive tract
plague in wild Anseriformes. J Wildl Manage. 33:294. lesions in duck plague. Vet Pathol. 12:349.
40 Leibovitz, L. 1971. Gross and histopathologic changes of 57 Proctor, S.J., G.L. Pearson, and L. Leibovitz. 1975. A
duck plague (duck virus enteritis). Am J Vet Res 32:275. color atlas of wildlife pathology. 2. Duck plague in
41 Leibovitz, L. 2015. Progress report: duck plague free‐flying water‐fowl. Wildl Dis Color Fiche. 67.
surveillance of American Anseriformes. Wildl Dis 58 Qi, X., and X. Yang. 2008. Quantitative analysis of
4:87–91. virulent duck enteritis virus loads in experimentally
42 Leibovitz, L., and J. Hwang. 1968. Duck Plague on the infected ducklings. Avian Dis. 52:338–344.
American Continent. Avian Dis. 12:361–378. 59 Roizman, B., and L.E. Carmichael. 1981. Herpesviridae.
43 Leibovitz, L., and J. Hwang. 2001. DUCK PLAGUE IN Definition, provisional nomenclature, and taxonomy. The
AMERICAN ANSERIFORMES. J Wildl Dis. 4:13–14. Herpesvirus Study Group, the International Committee
44 Lian, B., and A. Cheng. 2011. Induction of immune on Taxonomy of Viruses. Intervirology. 16:201–217.
responses in ducks with a DNA vaccine encoding duck 60 Shawky, S. 2000. Target cells for duck enteritis virus in
plague virus glycoprotein C. Virology Journal 8:1–8. lymphoid organs. Avian Pathol. 29:609–616.
45 Lin, W., and K.M. Lam. 1984. Isolation of an 61 Shawky, S., and T. Sandhu. 2000. Pathogenicity of a
Apathogenic Immunogenic Strain of Duck Enteritis low‐virulence duck virus enteritis isolate with apparent
Virus from Waterfowl in California. Avian Dis. 28:641. immunosuppressive ability. Avian Dis. 590–599.
46 Lin, W.Q., and K.M. Lam. 1984. Active and passive 62 Shawky, S., and K.A. Schat. 2015. Latency sites and
immunization of ducks against duck viral enteritis. reactivation of duck enteritis virus. Avian Dis.
Avian Dis. 28:968–973. 46:308.
47 Liu, J., and P. Chen. 2013. Recombinant duck enteritis 63 Shawky, S.A., and T.S. Sandhu. 1997. Inactivated
virus works as a single‐dose vaccine in broilers vaccine for protection against duck virus enteritis.
providing rapid protection against H5N1 influenza Avian Dis. 41:461–468.
infection. Antiviral Res. 97:329–333. 64 Shen, C., and A. Cheng. 2010. Development and
48 Liu, J., and P. Chen. 2011. A duck enteritis virus‐ evaluation of an immunochromatographic strip test
vectored bivalent live vaccine provides fast and based on the recombinant UL51 protein for detecting
complete protection against H5N1 avian influenza antibody against duck enteritis virus. Virology J. 7:268.
virus infection in ducks. J Virology. 85:10989. 65 Shen, F., and G. Ma. 2010. Development and
49 Liu, X., and Q. Liu. 2016. Attenuated Salmonella application of an indirect immunohistochemical
Typhimurium delivery of a novel DNA vaccine induces method for the detection of duck plague virus vaccine
immune responses and provides protection against antigens in paraffin sections and localization in the
duck enteritis virus. Vet Microbiol. 186:189. vaccinated duckling tissues. Poult Sci. 89:1915–1923.
50 Makhija, A., and S. Kumar. 2017. Characterization of 66 Spieker, J.O., and T.M. Yuill. 1996. Virulence of six
duck plague virus stability at extreme conditions of strains of duck plague virus in eight waterfowl species. J
temperature, pH and salt concentration. Biologicals. Wildl Dis. 32:453–460.
45:102–105. 67 Sr, J.J. 1964. The interference phenomenon in the
51 Malmarugan, S., and S. Sulochana. 2002. Comparison development of resistance against duck plague. J Comp
of dot‐elisa passive haemagglutination test for the Pathol Ther. 74:3–7.
detection of antibodies to duckplague. Ind Vet J. 68 States, U. 1969. Duck virus enteritis.
79:648–651. 69 Sun, Y., and C. Yang. 2017. Construction of a
52 Mo, C.L., and E.C. Burgess. 1987. Infection of duck recombinant duck enteritis virus vaccine expressing
plague carriers with Pasteurella multocida and P. hemagglutinin of H9N2 avian influenza virus and
anatipestifer. Avian Dis. 31:197. evaluation of its efficacy in ducks. Arch Virol. 162:1–9.
53 Mondal, B., and T.J. Rasool. 2010. Propagation of 70 Tantaswasdi, U., and W. Wattanavijarn. 1988. Light,
vaccine strain of duck enteritis virus in a cell line of immunofluorescent and electron microscopy of duck
duck origin as an alternative production system to virus enteritis (duck plague). Nihon Juigaku Zasshi the
propagation in embryonated egg. Biologicals. Japanese Journal of Veterinary Science. 50:1150.
38:401–406. 71 Toth, T.E. 1971. Active immunization of White Pekin
54 Ning, L., and T. Hong. 2016. Pathogenicity of duck ducks against duck virus enteritis (duck plague)
plague and innate immune responses of the Cherry with modified‐live‐virus vaccine: serologic and
Valley ducks to duck plague virus. Sci Reports. 6:32183. immunologic response of breeder ducks. Am J Vet Res.
55 Owens, and R.A. 2012. Virus Taxonomy: Ninth Report 32:75–81.
of the International Committee on Taxonomy of Viruses. 72 Toth, T.E. 1971. Two aspects of duck virus enteritis:
A.M.Q. King, M.J. Adams, E.B. Carstons, and E.H. parental immunity, and persistence/excretion of
Leftowitc, eds. virulent virus. Us Livest Sanit Assoc Proc.
494 Section II  Viral Diseases

73 Umamaheswararao, S. 1993. Assay of cell mediated Hemorrhagic Nephritis Enteritis of Geese


immune responses of ducks vaccinated against duck
1 Bernath, S., A., Farsang, A., Kovacs, E., Nagy, and M.
plague. Ind J Poult Sci. 28:256–258.
Dobos‐Kovacs. 2006. Pathology of Goose hemorrhagic
74 Van Dorssen, C., and H. Kunst. 1955. Susceptibility of
polyomavirus infection in goose embryos. Avian
ducks and various other water fowl to duck plague
Pathol. 35(1): 49–52.
virus. Tijdschr Diergeneesk. 80:1286–1295.
2 Bernath, S., and F. Szalai. 1970. Investigations for
75 Wang, J., and A. Ge. 2015. Construction of a
clearing the etiology of the disease appeared among
recombinant duck enteritis virus (DEV) expressing
goslings in 1969. Magyar Alla Lap. 25:531–536.
hemagglutinin of H5N1 avian influenza virus based on
3 Corrand, L., J. Gelfi, O. Albaric M. Etievant, J‐L.,
an infectious clone of DEV vaccine strain and
Pingret, and J‐L. Guérin. 2011. Pathological and
evaluation of its efficacy in ducks and chickens.
epidemiological significance of Goose haemorrhagic
Virology J. 12:126.
polyomavirus (GHPV) infection in ducks. Avian Pathol.
76 Wei, S., and X. Liu. 2014. The US2 protein is involved
40(4):355–60.
in the penetration and cell‐to‐cell spreading of DEV in
4 Gaweł A., G. Woźniakowski, E. Samorek‐
vitro. J Basic Microbiol. 54:1005–1011.
Salamonowicz, W. Kozdruń, K. Bobrek, K. Bobusia,
77 Wen, Y., and A. Cheng. 2010. A thymidine kinase
and M. Nowak. 2014. Hemorrhagic nephritis and
recombinant protein‐based ELISA for detecting
enteritis in a goose flock in Poland‐‐disease course
antibodies to duck plague virus. Virology J. 7:77.
analysis and characterization of etiologic agent. Avian
78 Wobeser, G. 2015. Experimental duck plague in
Dis. 58(4):518–22.
Blue‐Winged Teal and Canada geese. J Wildl Dis.
5 Gelfi, J., M., Pappalardo, C., Claverys, B., Peralta, and J‐L.
23:368–375.
Guérin. 2010. Safety and efficacy of an inactivated
79 Wobeser, G., and D.E. Docherty. 1987. A solitary case
Carbopol‐adjuvanted Goose haemorrhagic polyomavirus
of duck plague in a wild mallard. J Wildl Dis.
vaccine for domestic geese. Avian Pathol. 39(2):111–116.
23:479–482.
6 Guérin, J‐L., J., Gelfi, L., Dubois, A., Vuillaume, C.,
80 Woźniakowski, G., and E. Samorek‐Salamonowicz.
Boucraut‐Baralon, and J‐L. Pingret. 2000. A novel
2014. First survey of the occurrence of duck enteritis
polyomavirus Goose hemorrhagic polyomavirus is the
virus (DEV) in free‐ranging Polish water birds. Arch
agent of hemorrhagic nephritis enteritis of geese.
Virol. 159:1439–1444.
J Virol. 74:4523–4529.
81 Wu, Y., and A. Cheng. 2012. Complete genomic
7 Imperiale, M. and E.O. Major. 2007. Polyomaviruses.
sequence of Chinese virulent duck enteritis virus.
In: Fields Virology, 5th. Knipe, D.M. and P.M. Howley
J Virology. 86:5965.
Eds. Lippincott Williams & Wilkins Publishers,
82 Wu, Y., and A. Cheng. 2011. Serologic detection of
Philadelphia. 2263–2298.
duck enteritis virus using an indirect ELISA based on
8 Johne, R., and H. Muller. 2003. The genome of Goose
recombinant UL55 protein. Avian Dis. 55:626–632.
hemorrhagic polyomavirus, a new member of the
83 Wu, Y., and A. Cheng. 2012. Comparative genomic
proposed subgenus Avipolyomavirus. Virology.
analysis of duck enteritis virus strains. J Virology.
10(2):291–302.
86:13841–13842.
9 King A.M., J.A. Adams, E.B. Cartens and E.L.J.
84 Yang, C., and J. Li. 2015. Biological properties of a duck
Lefkowitz (Ed). 2012. Virus Taxonomy: 9th Report of
enteritis virus attenuated via serial passaging in chick
the International Committee, on Taxonomy of Viruses.
embryo fibroblasts. Arch Virol. 160:267–274.
Academic Press.
85 Ying, S., and C. Yang. 2017. Construction of a
10 Kisary, J. 1993. Haemorragic nephritis and enteritis of
recombinant duck enteritis virus vaccine expressing
geese. In: Virus Infections of Birds. J‐B. Mc Ferran, and
hemagglutinin of H9N2 avian influenza virus and
M.S. McNulty, Elsevier, London. 513–514.
evaluation of its efficacy in ducks. Arch Virol.
11 Lacroux, C., O., Andreoletti, B, Payre, J‐L., Pingret, A.,
162:171.
Dissais, and J‐L. Guérin. 2004. Pathology of
86 Yuan, G.P., and A.C. Cheng. 2005. Electron microscopic
spontaneous and experimental infections by Goose
studies of the morphogenesis of duck enteritis virus.
haemorrhagic polyomavirus. Avian Pathol.
Avian Dis. 49:50–55.
33(3):351–358.
87 Zhao, Y., and Y. Cao. 2014. Duck enteritis virus
12 Leon O., L. Corrand, T.N. Bich, O. Le Minor, M.
glycoprotein D and B DNA vaccines induce immune
Lemaire, and J‐L. Guérin. 2013. Goose hemorrhagic
responses and immunoprotection in Pekin ducks. Plos
polyomavirus detection in geese using real‐time PCR
One. 9:e95093.
assay. Avian Dis. 57(4):797–799.
88 Zou, Z., and J. Ma. 2016. Live attenuated vaccine based
13 Mató T., Z., Pénzes, P., Rueda, C., Vela, V., Kardi, A.,
on duck enteritis virus against duck hepatitis A virus
Zolnai, F., Misák, and V. Palya. 2009. Recombinant
types 1 and 3. Front Microbiol. 7.
Chapter 13  Viral Infections of Waterfowl 495

subunit vaccine elicits protection against goose 8 Corrand, L., J. Gelfi, O. Albaric, M. Etievant, J.L. Pingret,
haemorrhagic nephritis and enteritis. Avian Pathol. and J.L. Guerin. 2011. Pathological and epidemiological
38(3):233–237. significance of goose haemorrhagic polyomavirus
14 Palya, V., E., Ivanics, R., Glavits, A., Dan, T., Mato, and infection in ducks. Avian Pathol. 40:355–360.
P. Zarka. 2004. Epizootic occurrence of haemorrhagic 9 Cotmore, S.F., M. Agbandje‐McKenna, J.A. Chiorini,
nephritis enteritis infections of geese. Avian Pathol. D.V. Mukha, D.J. Pintel, J. Qiu, M. Soderlund‐Venermo,
33(2): 244–250. P. Tattersall, P. Tijssen, D. Gatherer, and A.J. Davison.
15 Ritchie, B.W, N., Pritchard, D., Pest, FD., Niagro, KS., 2014. The family Parvoviridae. Arch Virol.
Latimer, and PD. Lukert. 1993. Susceptibility of avian 159:1239–1247.
polyomavirus to inactivation. J Assoc Avian Vet 10 Cotmore, S.F., and P. Tattersall. 2007. Parvoviral host
7(4):193–195. range and cell entry mechanisms. Adv Virus Res.
16 Schettler, C.H. 1980. Clinical picture and pathology of 70:183–232.
haemorragic and enteritis in geese. Tier Prax. 8: 11 Coudert, M., Fedida, M., Dannacher, G., and Peillon, M.
313–320. 1974. Parvovirus disease of goslings. Late form. Recl
17 Szalai, F., and S. Bernath.1971. Investigations for Med Vet. 150:899–906.
clearing the etiology of the disease appeared among 12 Derzsy, D. 1967. A viral disease of goslings. I.
goslings in 1969: II. The elaboration of passive Epidemiological, clinical, pathological and aetiological
immunization, the production of hyperimmune serum. studies. Acta Vet Acad Sci Hung. 17:443–448.
III. Pathohistological investigations. Magyar Alla Lap. 13 Glávits, R., A. Zolnai, É. Szabó, É. Ivanics, P. Zarka, T.
26:420–423. Mató, and V. Palya. 2005. Comparative pathological
18 Woźniakowski, G., and K. Tarasiuk. 2015. Visual studies on domestic geese (Anser anser domestica) and
detection of goose haemorrhagic polyomavirus in geese Muscovy ducks (Cairina moschata) experimentally
and ducks by loop‐mediated isothermal amplification. infected with parvovirus strains of goose and Muscovy
Avian Pathol. 44(4):311–318. duck origin. Acta Vet Hung. 53:73–89.
14 Gough, R.E. 1984. Application of the agar gel precipitin
and virus neutralisation tests to the serological study of
Parvovirus Infections of Waterfowl
goose parvovirus. Avian Pathol. 13:501–509.
1 Brown, K.E., S.W. Green, and N.S. Young. 1995. Goose 15 Gough, R.E. 2008. Parvovirus infections. In: Diseases of
parvovirus—an autonomous member of the Poultry, 12th, Y.M. Saif, ed. Blackwell Publishing,
dependovirus genus? Virology. 210:283–291. Ames, Iowa. 397–404.
2 Chang, P.C., J.H. Shien, M.S. Wang, and H.K. Shieh. 16 Gough, R.E. 1987. Persistence of parvovirus antibody in
2000. Phylogenetic analysis of parvoviruses isolated in geese that have survived Derzsy’s disease. Avian Pathol.
Taiwan from ducks and geese. Avian Pathol. 29:45–49. 16:327–330.
3 Chen, H., Y. Dou, Y. Tang, Z. Zhang, X. Zheng, X. Niu, J. 17 Grieger, J.C., J.S. Johnson, B. Gurda‐Whitaker, M.
Yang, X. Yu, and Y. Diao. 2015. Isolation and genomic Agbandje‐McKenna, and R.J. Samulski. 2007.
characterization of a duck‐origin gpv‐related parvovirus from Surface‐exposed adeno‐associated virus Vp1‐NLS
Cherry Valley ducklings in China. PLoS One. 10:e0140284. capsid fusion protein rescues infectivity of
4 Chen, H., Y. Dou, Y. Tang, X. Zheng, X. Niu, J. Yang, X. noninfectious wild‐type Vp2/Vp3 and Vp3‐only
Yu, and Y. Diao. 2016. Experimental reproduction of capsids but not that of fivefold pore mutant virions. J
beak atrophy and dwarfism syndrome by infection in Virol. 81:7833–7843.
cherry valley ducklings with a novel goose parvovirus‐ 18 Guerin, J.L., J. Gelfi, L. Dubois, A. Vuillaume, C.
related parvovirus. Vet Microbiol. 183:16–20. Boucraut‐Baralon, and J.L. Pingret. 2000. A novel
5 Chen, H., Y. Tang, Y. Dou, X. Zheng, and Y. Diao. 2016. polyomavirus (goose hemorrhagic polyomavirus) is the
Evidence for vertical transmission of novel duck‐origin agent of hemorrhagic nephritis enteritis of geese.
goose parvovirus‐related parvovirus. Transbound Emerg J Virol. 74:4523–4529.
Dis. 63:243–247. 19 Hlinak, A., T. Muller, M. Kramer, R.U. Muhle, H.
6 Chen, S., S. Wang, X. Cheng, S. Xiao, X. Zhu, F. Lin, N. Liebherr, and K. Ziedler. 1998. Serological survey of
Wu, J. Wang, M. Huang, M. Zheng, and F. Yu. 2016. viral pathogens in bean and white‐fronted geese from
Isolation and characterization of a distinct duck‐origin Germany. J Wildl Dis. 34:479–486.
goose parvovirus causing an outbreak of duckling short 20 Hoekstra, J., T. Smit, and C. Van Brakel. 1973.
beak and dwarfism syndrome in China. Arch Virol. Observations on host range and control of goose virus
161:2407–2416. hepatitis. Avian Pathol. 2:169–178.
7 Chu, C.Y., M.J. Pan, and J.T. Cheng. 2001. Genetic 21 Ivanics, É., Glávits, R., Nagy, E‐ne, Édes, I‐ne, Révész,
variation of the nucleocapsid genes of waterfowl T., and Pálfy, V. 1998. Predominantly enteral form of
parvovirus. J Vet Med Sci. 63:1165–1170. Derzsy’s disease. Magy Állatorv Lap. 120:744–752.
496 Section II  Viral Diseases

22 Jansson, D.S., R. Feinstein, V. Kardi, T. Mato, and V. Palya. genome by polymerase chain reaction: distribution of
2007. Epidemiologic investigation of an outbreak of goose Goose parvovirus in Muscovy ducklings. Virus Res.
parvovirus infection in Sweden. Avian Dis. 51:609–613. 42:167–172.
23 Jestin, V., L.B.M. Cherbonnel, M. Le Gall, and G. 36 Lu, Y.S., D.F. Lin, Y.L. Lee, Y.K. Liao, and H.J. Tsai. 1993.
Bennejean. 1991. Demonstration of very pathogenic Infectious bill atrophy syndrome caused by parvovirus
parvovirus (Derzsy’s disease virus) in Muscovy duck in a co‐outbreak with duck viral hepatitis in ducklings
farms. Rec Med Vet. 167:849–857. in Taiwan. Avian Dis. 37:591–596.
24 Jordan, I., K. John, K. Howing, V. Lohr, Z. Penzes, E. 37 Malkinson, M., K. Perk, and Y. Weisman. 1981.
Gubucz‐Sombor, Y. Fu, P. Gao, T. Harder, Z. Zadori, Reovirus infection of young Muscovy ducks (Cairina
and V. Sandig. 2016. Continuous cell lines from the moschata). Avian Pathol. 10:433–440.
Muscovy duck as potential replacement for primary 38 Maurin‐Bernaud, L., S. Goutebroze, O. Merdy, A.
cells in the production of avian vaccines. Avian Pathol. Chanay, V. Cozette, and F.X. Le Gros. 2014. Efficacy of
45:137–155. a new attenuated duck parvovirosis vaccine in Muscovy
25 Ju, H., N. Wei, Q. Wang, C. Wang, Z. Jing, L. Guo, D. ducks. Vet Rec. 175:281.
Liu, M. Gao, B. Ma, and J. Wang. 2011. Goose 39 Ning, K., T. Liang, M. Wang, Y. Dong, S. Qu, and D.
parvovirus structural proteins expressed by Zhang. 2017. Genetic detection and characterization of
recombinant baculoviruses self‐assemble into virus‐like goose parvovirus: implications for epidemiology and
particles with strong immunogenicity in goose. pathogenicity in Cherry Valley Pekin ducks. Infect
Biochem Biophys Res Commun. 409:131–136. Genet Evol. 51:101–103.
26 Kisary, J. 1989. Clinical and pathological features: 40 Niu, X., H. Chen, J. Yang, X. Yu, J. Ti, A. Wang, and Y.
Diagnosis of avian parvovirus infections. In: CRC Diao. 2016. Development of a TaqMan‐based real‐time
Handbook of Parvoviruses, Vol. II., P. Tijssen, ed. CRC PCR assay for the detection of Novel GPV. J Virol
Press Inc., Boca Raton, FL. 193–203. Methods. 237:32–37.
27 Kisary, J. 1974. Cross‐neutralization tests on parvoviruses 41 Palya, V. 2013. Parvovirus infections of waterfowl. In:
isolated from goslings. Avian Pathol. 3:293–296. Diseases of Poultry, 13th, D.E. Swayne, ed. Wiley
28 Kisary, J. 1986. Diagnosis and control of parvovirus Blackwell, Ames, Iowa. 444–451.
infection of geese (Derzsy’s disease). In: Acute Virus 42 Palya, V., R. Glavits, M. Dobos‐Kovacs, E. Ivanics, E.
Infections of Poultry, J.B. McFerran, and M.S. McNulty, Nagy, K. Banyai, G. Reuter, G. Szucs, A. Dan, and M.
eds. Martinus Nijhoff, Dordrecht, Netherlands. 239–242. Benko. 2003. Reovirus identified as cause of disease in
29 Kisary, J. 1977. Immunological aspects of Derzsy’s young geese. Avian Pathol. 32:129–138.
disease in goslings. Avian Pathol. 6:327–334. 43 Palya, V., E. Ivanics, R. Glavits, A. Dan, T. Mato, and P.
30 Le Gall‐Recule, G., and V. Jestin. 1994. Biochemical and Zarka. 2004. Epizootic occurrence of haemorrhagic
genomic characterization of muscovy duck parvovirus. nephritis enteritis virus infection of geese. Avian
Arch Virol. 139:121–131. Pathol. 33:244–250.
31 Le Gall‐Recule, G., V. Jestin, P. Chagnaud, P. Blanchard, 44 Palya, V., A. Zolnai, Z. Benyeda, E. Kovacs, V. Kardi,
and A. Jestin. 1996. Expression of muscovy duck and T. Mato. 2009. Short beak and dwarfism syndrome
parvovirus capsid proteins (VP2 and VP3) in a of mule duck is caused by a distinct lineage of goose
baculovirus expression system and demonstration of parvovirus. Avian Pathol. 38:175–180.
immunity induced by the recombinant proteins. J Gen 45 Poonia, B., P.A. Dunn, H. Lu, K.W. Jarosinski, and K.A.
Virol. 77(9):2159–2163. Schat. 2006. Isolation and molecular characterization
32 Li, C., Q. Li, Z. Chen, and G. Liu. 2016. Novel duck of a new Muscovy duck parvovirus from Muscovy
parvovirus identified in Cherry Valley ducks (Anas ducks in the USA. Avian Pathol. 35:435–441.
platyrhynchos domesticus), China. Infect Genet Evol. 46 Qiu, J., F. Cheng, Y. Yoto, Z. Zadori, and D. Pintel. 2005.
44:278–280. The expression strategy of goose parvovirus exhibits
33 Li, L., J. Qiu, and D.J. Pintel. 2009. The choice of features of both the Dependovirus and Parvovirus
translation initiation site of the rep proteins from goose genera. J Virol. 79:11035–11044.
parvovirus P9‐generated mRNA is governed by splicing 47 Schettler, C.H. 1971. Goose virus hepatitis in the
and the nature of the excised intron. J Virol. Canada Goose and Snow Goose. J Wildl Dis.
83:10264–10268. 7:147–148.
34 Li, M., and T.F. Yu. 2013. Immunologic cross‐reactivity 48 Schettler, C.H. 1971. Virus hepatitis of geese. II. Host
between Muscovy duck parvovirus and goose range of goose hepatitis virus. Avian Dis. 15:809–823.
parvovirus on the basis of epitope prediction. Braz J 49 Shehata, A.A., D.M. Gerry, K. Heenemann, M.Y. Halami,
Microbiol. 44:519–521. S. Tokarzewski, P. Wencel, and T.W. Vahlenkamp. 2016.
35 Limn, C.K., T. Yamada, M. Nakamura, and K. Goose parvovirus and circovirus coinfections in
Takehara. 1996. Detection of Goose parvovirus ornamental ducks. Avian Dis. 60:516–522.
Chapter 13  Viral Infections of Waterfowl 497

50 Shen, H., W. Zhang, H. Wang, Y. Zhou, and S. Shao. Escherichia coli and their use in serological assays.
2015. Identification of recombination between Avian Pathol. 34:376–382.
Muscovy duck parvovirus and goose parvovirus 61 Wang, J., Y. Cui, H. Nan, and W. Yuan. 2017.
structural protein genes. Arch Virol. 160:2617–2621. Development of a taqman‐based real‐time PCR assay
51 Shien, J.H., Y.S. Wang, C.H. Chen, H.K. Shieh, C.C. Hu, for the rapid and specific detection of novel duck‐
and P.C. Chang. 2008. Identification of sequence origin goose parvovirus. Mol Cell Probes. 34:56–58.
changes in live attenuated goose parvovirus vaccine 62 Woolcock, P.R., V. Jestin, H.L. Shivaprasad, F.
strains developed in Asia and Europe. Avian Pathol. Zwingelstein, C. Arnauld, M.D. McFarland, J.C.
37:499–505. Pedersen, and D.A. Senne. 2000. Evidence of Muscovy
52 Sirivan, P., M. Obayashi, M. Nakamura, U. Tantaswasdi, duck parvovirus in Muscovy ducklings in California.
and K. Takehara. 1998. Detection of goose and Vet Rec. 146:68–72.
Muscovy duck parvoviruses using polymerase chain 63 Wozniakowski, G., E. Samorek‐Salamonowicz, and W.
reaction‐restriction enzyme fragment length Kozdrun. 2012. Quantitative analysis of waterfowl
polymorphism analysis. Avian Dis. 42:133–139. parvoviruses in geese and Muscovy ducks by real‐time
53 Soike, D., K. Albrecht, K. Hattermann, C. Schmitt, and polymerase chain reaction: correlation between age,
A. Mankertz. 2004. Novel circovirus in mulard ducks clinical symptoms and DNA copy number of waterfowl
with developmental and feathering disorders. Vet Rec. parvoviruses. BMC Vet Res. 8:29.
154:792–793. 64 Yang, J., R. Yang, A. Cheng, M. Wang, L. Fu, S. Yang,
54 Soike, D., B. Kohler, and K. Albrecht. 1999. A S. Zhang, L. Yang, and Z. Xu. 2010. A simple and
circovirus‐like infection in geese related to a runting rapid method for detection of Goose Parvovirus in
syndrome. Avian Pathol. 28:199–202. the field by loop‐mediated isothermal amplification.
55 Takehara, K., T. Nishio, Y. Hayashi, J. Kanda, M. Sasaki, Virol J. 7:14.
N. Abe, M. Hiraizumi, S. Saito, T. Yamada, M. Haritani, 65 Yang, J.L., A.C. Cheng, M.S. Wang, K.C. Pan, M. Li, Y.F.
et al. 1995. An outbreak of goose parvovirus infection Guo, C.F. Li, D.K. Zhu, and X.Y. Chen. 2009.
in Japan. J Vet Med Sci. 57:777–779. Development of a fluorescent quantitative real‐time
56 Takehara, K., T. Ohshiro, E. Matsuda, T. Nishio, T. polymerase chain reaction assay for the detection of
Yamada and M. Yoshimura. 1995. Effectiveness of an Goose parvovirus in vivo. Virol J. 6:142.
inactivated goose parvovirus vaccine, in Muscovy 66 Yen, T.‐Y., K.‐P. Li, S.‐C. Ou, J.‐H. Shien, and P.‐C.
ducks. J Vet Med Sci. 57:1093–1095. Chang. 2015. The White Roman Goose as a host for
57 Tatár‐Kis, T., T. Mató, B. Markos, and V. Palya. 2004. infection and viral shedding of Muscovy duck
Phylogenetic analysis of Hungarian goose parvovirus parvovirus. Taiwan Veterinary Journal. 41:85–89.
isolates and vaccine strains. Avian Pathol. 33:438–444. 67 Yu, K., X. Ma, Z. Sheng, L. Qi, C. Liu, D. Wang, B.
58 Tsai, H.J., C.H. Tseng, P.C. Chang, K. Mei, and S.C. Huang, F. Li, and M. Song. 2016. Identification of
Wang. 2004. Genetic variation of viral protein 1 genes goose‐origin parvovirus as a cause of newly emerging
of field strains of waterfowl parvoviruses and their beak atrophy and dwarfism syndrome in ducklings.
attenuated derivatives. Avian Dis. 48:512–521. J Clin Microbiol. 54:1999–2007.
59 Wan, C.H., H.M. Chen, Q.L. Fu, S.H. Shi, G.H. Fu, L.F. 68 Zadori, Z., R. Stefancsik, T. Rauch, and J. Kisary. 1995.
Cheng, C.T. Chen, Y. Huang, and K.H. Hu. 2016. Analysis of the complete nucleotide sequences of goose
Development of a restriction length polymorphism and muscovy duck parvoviruses indicates common
combined with direct PCR technique to differentiate ancestral origin with adeno‐associated virus 2. Virology.
goose and Muscovy duck parvoviruses. J Vet Med Sci. 212:562–573.
78:855–858. 69 Zhu, Y., Z. Zhou, Y. Huang, R. Yu, S. Dong, Z. Li, and Y.
60 Wang, C.Y., H.K. Shieh, J.H. Shien, C.Y. Ko, and P.C. Zhang. 2014. Identification of a recombinant Muscovy
Chang. 2005. Expression of capsid proteins and non‐ duck parvovirus (MDPV) in Shanghai, China. Vet
structural proteins of waterfowl parvoviruses in Microbiol. 174:560–564.

You might also like