Sei sulla pagina 1di 20

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/256488773

Avian influenza: Virology, diagnosis and


surveillance

Article in Future Microbiology · September 2013


DOI: 10.2217/fmb.13.81 · Source: PubMed

CITATIONS READS

10 251

4 authors, including:

Mohamed El Zowalaty Stephen Andrew Bustin


University of KwaZulu-Natal Anglia Ruskin University
54 PUBLICATIONS 440 CITATIONS 221 PUBLICATIONS 22,111 CITATIONS

SEE PROFILE SEE PROFILE

Mohamed Husseiny
Zagazig University
26 PUBLICATIONS 1,055 CITATIONS

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Graphene oxide Biomedical applications View project

Prevalence, Isolation and molecular characterization of shiga toxin producing Escherichia coli from
duck and chicken View project

All content following this page was uploaded by Stephen Andrew Bustin on 27 February 2015.

The user has requested enhancement of the downloaded file.


Avian influenza: virology, diagnosis

Review
Future Microbiology
and surveillance
Mohamed E El Zowalaty*1,2,3, Stephen A Bustin1, Mohamed I Husseiny3,4
& Hossam M Ashour*5,6
1
Postgraduate Medical Institute, Faculty of Health, Social Care & Education, Anglia Ruskin University,
Chelmsford, Essex, UK
2
Faculty of Public Health & Tropical Medicine, Jazan University, Jazan, Saudi Arabia
3
Department of Microbiology & Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
4
Beckman Research Institute at City of Hope, Duarte, CA, USA
5
Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy & Health Sciences, Wayne
State University, Detroit, MI, USA
6
Department of Microbiology & Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt„
*Authors for correspondence: hossamking@mailcity.com n elzow001@gmail.com

Avian influenza virus (AIV) is the causative agent of a zoonotic disease that affects
populations worldwide with often devastating economic and health consequences.
Most AIV subtypes cause little or no disease in waterfowl, but outbreaks in poultry
can be associated with high mortality. Although transmission of AIV to humans

of
occurs rarely and is strain dependent, the virus has the ability to mutate or reassort
into a form that triggers a life-threatening infection. The constant emergence of
new influenza strains makes it particularly challenging to predict the behavior,
spread, virulence or potential for human-to-human transmission. Because it is

ro
difficult to anticipate which viral strain or what location will initiate the next
pandemic, it is difficult to prepare for that event. However, rigorous implementation
of biosecurity, vaccination and education programs can minimize the threat of
AIV. Global surveillance programs help record and identify newly evolving and
potentially pandemic strains harbored by the reservoir host.
rP
Avian influenza (AI) is an acute avian respira- has been transmitted to humans (e.g., H1N1 in
tory disease caused by AI viruses (AIVs) that 1976, 1986 and 1988; H3N2 in 1993; H7N7
affect many species of domestic and wild birds in 1996; and H9N2 in 1998, 1999 and 2003)
and can also infect other animal species. In inducing mild, severe or fatal infections [3,4] .
ho

poultry, AI is characterized by marked varia- While it has been suggested that aerosolized
tion in morbidity and mortality. The notifiable droplets or direct contact with virus in poultry
form of AI (NAI) describes an outbreak of AI feces can lead to infections in humans [5] , poul-
in poultry due to any H5 or H7 subtype of try-to-human transmission of IAVs is unlikely
influenza A virus (IAV) or an infection by any to cause pandemic influenza. This requires the
AIV resulting in an intravenous pathogenicity emergence of viral subtypes with human-to-
ut

index >1.2 or causing at least 75% mortality [1] . human transmissibility arising from mutations,
HPNAI is a ‘list A’ notifiable disease (Box 1) of the genetic rearrangement of viral RNA segments or
World Organization for Animal Health (OIE), genetic reassortment between different strains of
which describes serious transmissible diseases virus [6,7] in a permissive host [8] , with pigs being
A

that can potentially spread very rapidly between the probable ‘mixing vessel’ [9] .
countries and are of major significance for the Several examples of human-adapted IAVs exist
international trade of animals and animal prod- that have initiated a number of pandemics in the
ucts, and so have severe socioeconomic or public last century [10] . The emergence of the Asian-
health costs [1] . LPNAI includes all IAVs of H5 based HPAI H5N1 virus, in particular, high-
and H7 subtype that are not HPNAI viruses. lights the continuing public health concerns over
Low pathogenicity avian influenza (LPAI) refers human fatalities and its potential emergence as a
to all infections caused by AIVs that are not NAI future pandemic virus. This implicates AI as one
viruses and include H1-H4, H6, and H8-H16 of the most threatening trans-boundary diseases Keywords
LPAI [2, 201] . facing humans [11] , highlights the potential risk n avian influenza n diagnosis
The HPAI viruses are restricted to subtypes IAV poses to human health and economy [12–14] n real-time PCR n surveillance
H5 and H7, although not all H5 and H7 viruses and strongly emphasizes the urgent requirement n vaccines n virus isolation
n waterfowl
cause HPAI. The risk from AI is generally low for continued high levels of awareness and sur-
in most people, as AIVs do not usually infect veillance programs to monitor for emerging AIVs
humans. However, there have been several cases [15] . Accordingly, the WHO has complemented
of conjunctivitis in humans related to LPAI. AIV its human surveillance network with a program part of

10.2217/FMB.13.81 © 2013 Future Medicine Ltd Future Microbiol. (2013) 8(9), 1–19 ISSN 1746-0913 1
Review El Zowalaty, Bustin, Husseiny & Ashour

China, spreading to Hong Kong and the rest


Box 1. Notifiable avian influenza.
of the world, and causing the deaths of a few
„„List A notifiable diseases of the World Organization for Animal Health are defined million individuals [22] .
as transmissible diseases that have the potential for very serious and rapid spread, In February 2009, a new swine-origin influ-
irrespective of national borders, are of serious socioeconomic or public health
enza A (H1N1) virus emerged in Mexico from
consequence and are of major importance in the international trade of animals
and animal products. multiple reassortment events involving a unique
„„Notifiable avian influenza (AI) is defined as an infection of poultry caused by any
combination of gene segments from human,
influenza A virus of the H5 or H7 subtypes or by any AI virus with an intravenous swine and avian type A viruses [23–27] , result-
pathogenicity index in chickens of greater than 1.2 (or, as an alternative, at least ing in the first influenza pandemic of the 21st
75% mortality). AI can be categorized into highly pathogenic notifiable avian century [28,29] . By mid-April, it had reached the
influenza, including high-pathogenicity H5 and H7 subtypes, low-pathogenicity USA via human-to-human transmission, leading
avian influenza (LPAI), which includes all H5 and H7 LPAI subtypes, and all other the WHO to raise its pandemic alert successively
LPAIs, which are not notifiable to the World Organization for Animal Health,
to phases 4 (27 April), 5 (29 April) and 6 (11
including H1–H4, H6 and H8–H16 LPAIs.
June) [24] . Although now in the post-pandemic
focusing on the ecology and surveillance of phase, the H1N1 (2009) virus is one of the
influenza viruses in wild animals [16,17] . seasonal influenza viruses in global circulation

of
and continues to be a major challenge to public
Human influenza pandemics health.
The three worldwide influenza outbreaks (pan-
demics) in the 20th century occurred in 1918, History of HPAI

ro
1957 and 1968 [18] . Each pandemic caused In 1878, a contagious disease of poultry asso-
significant social and economic upheaval. The ciated with high mortality was first described
rapid spread, combined with high morbidity and in Italy and named ‘fowl plague’. In 1955, the
mortality, gave rise to intense levels of anxiety causative agent was identified as an IAV, based
and fear among the public. The ‘Spanish flu’ on the presence of A type-specific ribonucleo-
rP
(1918–1919) was caused by direct transmission protein. This resulted in the term fowl plague
of the H1N1 virus from birds [19] and is consid- being replaced by the more suitable term HPAI
ered to be the ‘mother of all pandemics’ [20] . It [17, 30–32] . In 1961, a high proportion of deaths in
spread across the entire world within 3 months, common terns (Sterna hirundo) in South Africa
infecting 50% of the world’s population and was attributed to the HP A/H5N3 virus, which
ho

resulting in an estimated 30–50 million deaths was the first demonstration of a HPAI virus in
[21] . The ‘Asian flu’ (1957–1958) was caused by wild birds before the HP A/H5N1 [33] . The 1997
genetic reassortment between human H1N1 outbreak of respiratory disease in humans in
and avian H2N2 influenza viruses. This pan- Hong Kong caused by H5N1, which was previ-
demic spread across the world in approximately ously known to infect only avian species, raised
6 months and, together with a second wave of serious concerns about the potential for another
ut

infections, affected approximately 40–50% of pandemic in humans [34,35] . The H5N1 virus
the world’s population, killing approximately seems to have emerged in southeast China dur-
1 million people [22] . The ‘Hong-Kong flu’ ing 1996 [36] , where it reassorted with viruses
(1968–1969) pandemic was caused by a H3N2 circulating in other avian species and emerged
A

virus, which resulted from reassortment of cir- as a new H5N1 virus in Hong Kong. After an
culating human H2N2 and avian H3 virus in initial major outbreak of HPAI in chickens and
other birds, it was unexpectedly transmitted
to humans [37] , and in May 1997 a 3-year-old
Box 2. Antigenic drift versus antigenic shift.
boy became the first human to die of a respira-
„„Antigenic drift is defined as the process whereby small changes or mutations tory illness related to an H5N1 virus infection
accumulate in the virus, mainly in the surface glycoproteins (predominantly [38] . An additional 17 cases were diagnosed in
hemagglutinin [HA]), which happen continually over time. The new virus strains
November and December of the same year [17] . A
thus produced have different antigenic profiles and may not be recognized by
antibodies against the older strains, thereby making the individual susceptible to
few years later, the virus spread to several Asian
infection with the new strains. countries, infecting humans for the second time
„„Antigenic shift is an abrupt, major change in the HA and/or neuraminidase (NA) in February 2003 [39] .
proteins of the virus that occurs when surface HA and NA segments reassort Since late 2003 until the present day, the
between different viruses, resulting in the emergence of a new subtype. world has witnessed the deadliest outbreak in
Alternatively, the new HA and NA combination may have emerged from an the history of HPAI in birds. In addition, the
animal population that is so different from the same subtype in humans that transmission of A/H5N1 virus to humans is on
most people do not have immunity to the new virus. the increase in Asia, southeast Asia, Africa and

2 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

the Middle East. Between 1997 and April 2013, evident and has attracted the attention of influ-
HPAI A/H5N1 infection has been identified in enza researchers. Influenza A/H3N2 viruses dis-
628 human cases, with 374 deaths confirmed covered in horses were also found in pigs, cattle,
in 15 countries, resulting in a fatality rate of chickens, dogs and other species throughout the
59.55%; the highest numbers of cases/deaths world [46] . An outbreak of severe respiratory dis-
were reported in Indonesia, Egypt, Vietnam ease in a pack of English foxhounds in the UK
and China, respectively [202] . Although A/H5N1 in September 2002 was caused by an equine A/
originated in China, it is still unclear why the H3N8 virus [47] . H3N8 and H3N2 canine influ-
number of cases is not the highest in its originat- enza viruses have been reported in racing canine
ing location. So far, there has been no evidence populations in the USA [48] .
of sustained human-to-human transmission of Aquatic birds have been revealed as the source
AIV. However, if the virus mutates or rearranges of influenza viruses in sea mammals, such as seals
deliberately or spontaneously into a form that is (H4N5, H7N7) and whales (H13N2, H13N9,
more easily transmissible between humans, it will H1N3) [5] . In this context, it is worth noting
have the potential to kill millions in a pandemic. the concerns expressed about the use of ferrets
The concurrent A/H5N1 virus possesses as a model. Ferrets are considered to be the best

of
three of the four properties necessary to cause model for IAV because of their high suscepti-
a pandemic: it infects people, most people are bility and the fact that IAV infection in ferrets
immunologically naive and it is highly lethal. closely resembles that in humans with respect
However, it lacks the capacity for sustained to clinical signs, pathogenesis and immunity.

ro
human-to-human transmission [18,40] . Although Indeed, recent studies in ferrets have shown that
that capacity may require only a single genetic IAV can acquire the capacity for airborne trans-
reassortment or mutation, up until now, most mission between mammals without recombina-
infected people have been infected through tion in an intermediate host, and therefore con-
direct contact with infected poultry [41] . While stitute a high risk for human pandemic influenza
rP
there have been extensive efforts by the WHO to [49,50] . The ability to produce such an infection
report all cases, the numbers might not reflect the raises both biosecurity and biosafety concerns
real picture, especially in developing countries, and could even lead to a human pandemic [51] .
since not all cases are being admitted to hospitals
or reported by governments. In developed coun- Genome & virology of AIV
ho

tries, the strict implementation of biosecurity Influenza viruses are members of the family
and preventive procedures in poultry farms helps Orthomyxoviridae (orthos, Greek for straight;
guard against the spread of AIV. Annual surveil- myxa, Greek for mucus) which belongs to
lance programs among waterfowl have also been group V (negative-polarity ssRNA) of the Bal-
implemented in the USA and Europe. timore system of virus classification [52] . There
In the spring of 2013, another ongoing out- are five genera: influenza virus A, B and C,
ut

break of novel avian A/H7N9 virus that origi- Isavirus and Thogotovirus [53,54] . Each genus
nated from multiple reassortment events has includes only one species of IAV, influenza B
again emerged in China, where H7N9 has so virus and influenza C virus. Influenza A and C
far caused infection in 131 laboratory-confirmed viruses infect multiple species, while influenza B
A

human cases, including 36 deaths [42,43] . The viruses almost exclusively infect humans [55] .
recent A/H7N9 could potentially produce IAVs include avian, swine, equine and canine
a human pandemic since the virus is more influenza viruses, as well as human IAVs.
humanized in traits and could increase human- The structure of IAVs has been extensively
to-human transmission, if transmitted among studied and reported [56] . The genome of IAVs
poultry or pigs, raising public health concerns of comprises eight negative-sense, viral ssRNA seg-
a looming pandemic influenza in human [44,45] . ments that are numbered in order of decreasing
In addition to AIV, other influenza viruses length, as summarized in Table 1. The two main
exist and can cause influenza epizootics among surface glycoproteins of influenza viruses as
various animal species [5] . In some species, shown in Figure 1 are the hemagglutinin (HA) and
the disease mimics human influenza, while in neuraminidase (NA) proteins, which function in
others, there are no signs of disease. Since the the attachment and release of the virion to and
demonstration of swine influenza (Hsw1N1) in from cells. They are major antigenic determi-
1930 and later in 1955, and with the identifica- nants and so are major targets for the immune
tion of influenza viruses in horses and ducks, response to which neutralizing antibodies are
transmission to other animals has become more made [57] . Theoretically, different combinations

future science group www.futuremedicine.com 3


4
Table 1. The genomic segments of influenza A virus and their encoded proteins and functions.
RNA RNA segment Gene Encoded Localization and features Nascent Approximate Proposed protein function(s)
segment (mRNA†) production protein(s)‡ protein number of
Review

number length in description length§ molecules


nucleotides in amino per virion
acids
1 2341 (2320) PB1 Polymerase Virion interior, infected cell 757 30–60 Heterotrimeric P complex associated with NP and virion
basic 1, 87 kDa nuclei RNA, RNA transcription and the replication component of
RNA polymerase (polymerase subunit); initiation of
transcription, RNA transcriptase, RNA elongation and
endonuclease activity
PB1-F2
A 87 Proapoptotic activity
2 2341 (2320) PB2 Polymerase Virion interior, infected cell 759 30–60 Polymerase subunit; RNA transcriptase, host cell mRNA cap
basic 2, 96 kDa nuclei recognition and virulence
3 2233 (2211) PA Polymerase Virion interior, infected cell 716 30–60 Polymerase subunit; RNA transcriptase and protease
acidic, nuclei
ut
85.5 kDa
El Zowalaty, Bustin, Husseiny & Ashour

4 1778 (1757) HA Hemagglutinin, Globular head bears 550 500 Surface glycoprotein, virus binding to sialic acid-containing
220 kDa antigenic sites and receptor receptors on host cell; penetration of virus genome into
homotrimer binding site; stem; host cell cytoplasm by fusion of virus and host cell
transmembrane span; membranes (attachment to cell membranes and receptor
ho
cytoplasmic tail binding) and major antigenic determinant
5 1565 (1540) NP Nucleoprotein, Virion interior, associated 498 1000 Encasidates RNA; RNA synthesis and RNA nuclear import
55 kDa with P complex and viral RNA regulation; interacts with RNAs and polymerase proteins as
a major component of the nucleocapsid; role in vRNA
replication; role in virus maturation and packaging

Future Microbiol. (2013) 8(9)


rP
6 1413 (1392) NA Neuraminidase, Virion envelope, infected cell 454 100 Surface glycoprotein; antigenic determinant, sialidase
240 kDa surface, cytoplasmic tail; (neuraminidase)-catalyzing cleavage of terminal sialic acid
homotetramer transmembrane span; residues from glycoconjugates, thereby digesting mucin to
extracellular stalk; globular enable the virus to reach its target epithelium and
head bears antigenic sites facilitating release of infectious progeny virus
ro
and enzyme active site

Deduced from RNA sequence, excluding poly-A tract. Total RNA nucleotide sequence length is 13,588.

Determined by biochemical and genetic approaches.
§
Determined by nucleotide sequence analysis and protein sequencing.
P complex: Polymerase protein complex; RNP: Ribonucleoprotein; vRNP: Viral ribonucleoprotein.
Data taken from [194].
of

future science group


Table 1. The genomic segments of influenza A virus and their encoded proteins and functions (cont.).
RNA RNA segment Gene Encoded Localization and features Nascent Approximate Proposed protein function(s)
segment (mRNA†) production protein(s)‡ protein number of
number length in description length§ molecules
nucleotides in amino per virion

future science group


acids
7 1027 (1005) M1 M1, 28 kDa Beneath lipid bilayer of virion 252 3000 Central role in replication and virus assembly, modulating
envelope; associates with nuclear transport of vRNP; early infection: bound to vRNP
vRNPs in mature virion to prior to RNP transport to nucleus; late infection: binds RNP,
A form nucleocapsid signaling RNP transport from nucleus to cell surface,
mediates association of RNP with hemagglutinin and
neuraminidase at the cell membrane, promoting virion
formation and budding
M2 M2, 15 kDa Virion envelope, infected cell 97 3000 Membrane cation channel activity; virus uncoating and
homotetramer surface (abundant); assembly; infected cell: specifically raises pH of Golgi to
ut
extracellular region; protect pH-sensitive hemagglutinin; virion: may permit
transmembrane span; acidification of virus interior during passage through
cytoplasmic tail; tetramers endosomal pathway in order to dissociate vRNP from M1
form cation-selective channel
ho
8 890 (868) NS1 NS1, 25 kDa Infected cell nuclei 230 Multifunctional protein and viral interferon antagonist
dimer protein; regulation of host gene expression; binds RNA,
thereby inhibiting host mRNA translation, regulating viral
pre-mRNA splicing and translation and viral polymerase
activity, and downregulating dsRNA-induced antiviral

www.futuremedicine.com
responses
rP
NEP/NS2 NS2, 14 kDa Associated with core 121 Mediates nuclear export of vRNPs
components of virion;
cytoplasm of infected cells

Deduced from RNA sequence, excluding poly-A tract. Total RNA nucleotide sequence length is 13,588.

Determined by biochemical and genetic approaches.
§
Determined by nucleotide sequence analysis and protein sequencing.
ro
P complex: Polymerase protein complex; RNP: Ribonucleoprotein; vRNP: Viral ribonucleoprotein.
Data taken from [194].
of
Avian influenza viruses: virology, diagnosis & surveillance
Review

5
Review El Zowalaty, Bustin, Husseiny & Ashour

Hemagglutinin

Neuraminidase

of
M2 ion channel

ro
rP
RNP

Figure 1. Molecular structure representation of influenza A virus showing two surface


glycoproteins (hemagglutinin and neuraminidase), the major antigenic determinants of the
virus. The hemagglutinin protein is responsible for binding to sialic acid receptors on host cells.
Human influenza A viruses preferentially bind to sialic acids in an a2,6 conformation, while those
ho

from avian species bind mostly to sialic acids in an a2,3 conformation.


RNP: Ribonucleoprotein.
Image was reproduced from the Centers for Diseases Control and Prevention (CDC), Atlanta,
Georgia, USA [206] with permission.

from the 17 HA and ten NA subtypes [58,59] can distinct H17N10 virus is the only subtype that is
ut

be found, and each subtype may contain several not found in an avian species [58,59] .
subtypes. Mutations can introduce additional
variability, and so the number of AIV strains Epidemiology of AI
is indefinite. The evolutionary success of IAV is a prototypic
A

In 1972, the WHO recommended a new sys- example of the ability of microbes to adapt to
tem for influenza nomenclature and classifica- their many hosts. Influenza is fundamentally a
tion of influenza viruses into genetically distinct recurring background disease that re-emerges
subtypes based on their NA and HA antigens slightly differently each year due to the con-
[60,61] . The nomenclature consists of two parts: tinuously evolving nature of their surface gly-
a strain designation and a description of the NA coproteins, referred to as antigenic drift [63] . At
and HA antigens [60,62] . To date, only three HA unpredictable time periods, influenza presents a
(H1, H2 and H3) and two NA (N1 and N2) sub- newly emerging disease caused by viruses with
types have caused human epidemics, as defined completely different surface antigens, termed
by sustained and widespread person-to-person antigenic shift (Box 2) , infecting humans with
transmission [8] . Nevertheless, the recent discov- little or no immunity against these strains [57] .
ery of the highly divergent IAV subtype H17N10 This antigenic variability of AIVs is due to their
in bats from Guatemala in South America rein- highly error-prone replication process [64] that,
forces the importance of surveillance for moni- together with viral genome assortment, allows
toring the evolution of influenza viruses among influenza viruses to adapt to their host, thus
different animal populations. Thus far, this new acquiring a pandemic potential [65] .

6 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

The ecology and epidemiology of AI have LPAI viruses have an HA cleavage site with one
changed substantially in the last two decades basic arginine or lysine residue that is cleaved
[66] . AI infections due to LP A/H9N2 subtype only by proteases with monobasic specificity. As
have become widespread in Asia, whereas the HP a result, LPAI infection in birds is restricted to
A/H5N1 subtype has been the causative agent the digestive or respiratory tracts and results in
of widespread infections in poultry across other milder illness or no disease at all. By contrast,
areas of the world, resulting in a modified eco- HPAI viruses are invariably HA subtypes H5
epidemiology and zoonotic potential [67] . or H7 with a polybasic cleavage site, introduced
As shown in Figure 2 , the primary difference either as a result of insertion or substitution
between LPAI and HPAI virus is local versus [69–71] . This polybasic cleavage site is susceptible
systemic replication, respectively. One of the key to the protease furin, which is ubiquitous in cells
determinants of virulence is the ability of the [72] . The factors that cause mutation from LPAI
host to proteolytically cleave the HA precursor to HPAI, which occurs only in birds, are not
HA0 into HA1 and HA2 subunits, an essen- known, but it seems that the wider the circula-
tial requirement for binding to the cell surface tion of LPAI in poultry, the higher the chance
receptors and fusion of the viral envelope with that mutation into HPAI will occur [73] . It is

of
the endosomal membrane. An analysis of HA important to remember, however, that acquisi-
subtypes that circulate in aquatic birds, as well tion of a polybasic HA cleavage site is the only
as HAs representative of the subtypes that have necessary step for the evolution of LPAI strains
infected the human population, indicates that into HPAI viruses, since not all H5 or H7 sub-

ro
the cleavage efficiency can vary significantly for types are hypervirulent. There are additional
different HA subtypes and some display strin- virulence determinants within the HA itself
gent selectivity for specific proteases [68] . The [74,75] and in other viral proteins, suggesting that
HA of mammalian viruses contains only a sin- virulence is under polygenic control [73,76,77] .
gle arginine and, rarely, a single lysine, at the
rP
cleavage site and is cleaved extracellularly, limit- Persistence of AIV in the environment
ing their spread in mammalian hosts to tissues LPAI viruses are ubiquitous [16] and approxi-
that express the appropriate proteases. Similarly, mately 90 species from some 12 of the 50 orders

N– HA0 TM –C
ho

Cleavage site

Avirulent avian isolate H5 RETR G LPAI: proteases localized in


Avirulent avian isolate H7 KXR G respiratory and intestinal organs
ut

Virulent avian isolate H5 RKRKKR G HPAI: ubiquitous proteases


Virulent avian isolate H7 KKRKKR G

Avian strains isolated from human H5N1 (1997 HK) RERRRKKR G


Avian strains isolated from human H9N2 (1999 HK) RSSR G
A

Avian strains isolated from human H7N7 (2003 NL) KRRRR G


Avian strains isolated from human H5N1 (2004 Asia) RRKKR G

N– HA1 –C N– HA2 TM –C

S–S

Figure 2. Schematic representation of the protease cleavage site in influenza A viruses


hemagglutinin. HA0 is cleaved into disulfide linked subunits HA1 and HA2 at a specific cleavage site
shown in red (single letter amino acid code is used to identify the amino acid sequence at the
hemagglutinin cleavage site). The hemagglutinins of LPAI viruses are cleaved by proteases that are
localized in respiratory and intestinal organs, resulting in mild localized infections, whereas the
hemagglutinins of HAI viruses have multiple basic amino acids, which are cleaved by ubiquitous
proteases in a wide range of organs, resulting in lethal systemic infection. Most human isolates of
avian strains also possess polybasic cleavage sites. The TM domain is shown in orange.
HK: Hong Kong; HPAI: High-pathogenicity avian influenza; LPAI: Low-pathogenicity avian influenza;
NL: The Netherlands; TM: Transmembrane.

future science group www.futuremedicine.com 7


Review El Zowalaty, Bustin, Husseiny & Ashour

of birds carry all strains of influenza, with birds The mechanisms by which influenza viruses
inhabiting wetland and aquatic environments pass from one bird to another and cause infec-
showing the highest rates of influenza infection tion are not fully understood, but may be due
[78] . Mixed infections with different influenza to combinations of factors that include strain of
subtypes among waterfowl are also common virus, species of bird and environmental factors.
[12,13,79] . Carriers are mainly of the orders Anseri- In wild ducks, for example, influenza viruses rep-
formes (especially the families Anatidae [ducks, licate preferentially in the cells lining the intesti-
geese, and swans], Charadriiformes [terns and nal tract and are excreted in high concentrations
waders] and Procellariiformes [shorebirds, gulls in the feces. Contamination of water supplies by
and seabirds]) and thus act as a reservoir for infective feces and fomites that contain concen-
the virus [80] . The migratory nature of many of trations as high as 107 infectious particles/g [5]
these species and the persistence of influenza in is one obvious route of infection [5,90] . This has
these populations facilitates the dissemination led to the assessment of ducks being the ‘Trojan
of influenza viruses worldwide [81] . Influenza horses’ of H5N1 in their surreptitious spread of
viruses infect different species and have become viruses because they do not show symptoms after
very successful parasites in their avian hosts, HP H5N1 infection [91] . Influenza viruses have

of
causing mostly silent or asymptomatic infections coevolved with ducks over a very long period of
while creating the opportunity to infect other time, allowing the establishment of equilibria
immunologically naive hosts [5] . between hosts and viruses so that neither suffers
Interspecies barriers and the host species speci- a significant loss of biological fitness. Hence, it is

ro
ficity mechanisms, such as receptor preference important to determine whether antigenic diver-
and host factors interacting with HA, NA, viral sity is driven naturally in ducks or whether it is it
polymerase and other internal genes, are impor- the consequence of vaccine usage. Furthermore,
tant molecular constraints and determinants for it will be necessary to determine the genomic
their transmissibility of AIVs among different characteristics of ducks that are associated with
rP
species [82,83] . IAV particles bind their target cells natural resistance in some species and ascertain
through interaction between their HA molecules what dose of vaccine antigen is required to pre-
and the sialic acid-containing cell-surface recep- vent transmissible levels of virus excretion by
tors on the host cells. Human influenza viruses ducks of different species [91] .
bind preferentially to N-acetylneuraminic acid Clearly, the natural reservoir of IAVs could
ho

(sialic acid), which is attached to a galactose be the source of the next human influenza pan-
molecule by an a2,6 linkage (SAa2,6Gal), demic [92] and so the routine surveillance and
while AIVs mostly bind to silalic acid with an early detection of these viruses [93,94] , their hosts
a2,3 linkage [84] . In human tracheal epithelial and their migratory patterns [95] , as well as the
cells, a2,6 linkages predominate, while a2,3- study of the impact of environmental and social
linkages are more common in duck gut epi- factors on their interactions [97] , are key to the
ut

thelial cells [83] . Recently, it was indicated that control, management and eradication of the
epithelial cells of the human lower respiratory disease [97] . One consequence of this has been
tract contain both SAa2,3Gal and SAa2,6Gal the formulation in the USA of an interagency
but with different distributions [85] . In humans, strategic plan by a group of federal and state
A

a2,3 linkages are also present in respiratory epi- resource, as well as science agencies, to conduct
thelial cells, but their presence is less abundant surveillance in wild birds in order to determine
than a2,6 linkages [86] . This explains the low the possible pathways of entry [98] .
infectivity but high pathogenicity of some AI In addition to carrying mixed populations
strains in mammalian species [56] . AIVs do not of AIVs, waterfowl can host additional viruses,
generally replicate well in humans and vice versa especially the avian paramyxoviruses (APMVs)
[87,88] , so it is possible that reassortment took [99,100] . Several studies have demonstrated the
place in an intermediate host. Pig populations circulation of influenza virus with APMV-1
have been proposed as a potential mixing vessel (also known as Newcastle disease virus [NDV]),
where reassortment takes place, since both avian APMV-2, APMV-4 and APMV-6 [101–105] . The
and human strains can infect and replicate in coexistence of NDV with AIV in field samples
pigs [89] . This is attributed to the fact that tra- might present a diagnostic problem when it is
cheal cells of pigs, where influenza replication desired to isolate and characterize only AIVs for
occurs, contain a2,3-linked sialic acid receptors surveillance and epidemiological purposes. The
preferred by avian viruses, as well as those with overwhelming growth of NDV may inhibit AIV
the a2,6-linkage favored by human strains [89] . growth, which in turn will decrease the chance

8 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

of detection of AI subtypes. In several AIV this results in significantly decreased virus titers
surveillance studies, if a sample is found to be [79] .
In addition, technical details such as the
positive for NDV, it is not processed for AIV use of dry or wet swabs, the pH of viral transfer
detection; only if a sample is found to be negative medium and the time taken to transport sam-
for NDV it is screened for AIV [106] . In a recent ples to the testing laboratory play a crucial role
report, it was found that treatment of the sample in the successful isolation of AIVs from field
with NDV polyclonal antiserum facilitates the samples [114] .
isolation of AI from samples containing both
AIVs and NDV [102] . Serological methods
Serological methods for the diagnosis of AI
Diagnosis of AI in birds play an important role in monitoring
An effective strategy for understanding the the disease in populations and can provide an
ecology and epidemiology of the virus, and accurate method for the detection of influenza
thus controlling the spread of influenza viruses, infection [118] . Serological diagnosis is important
depends on the availability of reliable laboratory in case of LPAI to ensure freedom of infection
techniques permitting accurate and sensitive for commercial purposes. Conversely, in cases

of
detection of AIVs in waterfowl [107,108] . Clinical of HPAI, serological methods are of little value
diagnosis of AIV in birds is performed by the since birds die before producing antibodies [111] .
isolation and characterization of the virus, which Serological methods are thus useful epidemio-
varies with species and type of infection [109] and logically for strain surveillance, but cannot be

ro
can be either presumptive or definitive. Clinical used for rapid diagnosis, which would allow
symptoms can be a valuable tool for presump- therapeutic intervention [119] . Common serologic
tive diagnosis of HPAI, whereas LPAI infection tests for AIV include hemagglutination inhibi-
is asymptomatic [110] . Definitive diagnosis of tion (HI), NA inhibition, agar gel precipitation
AIV depends on specific laboratory methods, (also known as agar gel immunodiffusion),
rP
including the indirect evidence of infection by virus neutralization, complement fixation (CF),
serological methods, which detect anti-influenza enzyme immunoassay and indirect immunofluo-
antibodies, and direct detection methods for live rescence [120,121] . These tests are based on the
virus, viral antigen or viral nucleic acid [111] . presence of influenza-specific antibodies that
first appear approximately 2 weeks after initial
ho

Specimen collection infection and peak at 4–7 weeks after initial


Clinical specimens for AIV diagnosis in birds infection [121] . HI and NA inhibition assays are
can be obtained from either live or dead birds. labor-intensive and time-consuming and require
Wild birds in surveillance programs of migrating several controls for standardization. However,
waterfowl are usually trapped using night light- they are inexpensive, use readily available rea-
ing, rocket netting and hunter harvesting tech- gents and display high specificity in identifying
ut

niques [203,204] . Samples from live birds should strains [122] . HI is more specific in differentiating
include both tracheal and cloacal swabs, since between HA subtypes [123] . The agar gel immu-
it has been shown that the analysis of both oro- nodiffusion test detects IgM and some IgG and
pharyngeal and cloacal swabs provides an accu- is type-specific [124] . Although CF tests have
A

rate snapshot of AIV status in birds [79,112–114] . If been used for the identification of influenza iso-
this is not possible, the collection of fresh feces lates [119] , the Canadian Public Health Labora-
may also serve as an alternative [115] . tories stopped offering this test in 2009 because
Samples should be placed in an isotonic phos- influenza CF serology does not provide a reliable
phate-buffered saline or viral transfer medium indicator of immunity or response to vaccination
consisting of brain–heart infusion medium con- and is not recommended for the diagnosis of
taining several antibiotics to eliminate any bacte- acute influenza infection [205] .
ria that may interfere with subsequent diagnostic
procedures [116] . Although diagnostic samples Virus isolation
can be stored temporarily at 40°C, it is recom- Influenza virus was first isolated in 1933, by
mended that all samples be kept at -800°C until inoculation of specimens into the amniotic cav-
tested, especially if prolonged storage is antici- ity of 10–12-day-old embryonating chicken eggs
pated [117] . In addition, field samples should be (ECEs) [125] , where permissive infection occurs
maintained under strict cold chain conditions in the cells lining the amniotic and allantoic
from the moment of acquisition and should not cavities [119] . This method, using either spe-
be subjected to frequent freeze–thaw cycles, as cific pathogen-free chicken eggs or specific

future science group www.futuremedicine.com 9


Review El Zowalaty, Bustin, Husseiny & Ashour

antibody-negative eggs [126] , continues to be the embryonating cells (MRC‑5), CACO-2 cell
gold standard technique for AIV diagnosis and lines, CCL-141 (duck embryo) cells, CCL-169
remains the most sensitive method for generat- (goose embryonic kidney) cells, duck embryo
ing very high titers of all but one AIV [120] . The fibroblast cells and chicken embryo fibroblast
exception is the recently discovered bat virus A/ cells [130–135] .
H17N10; to date, all attempts to propagate this Alternatively, since viruses are released slowly
virus in cell cultures or chicken embryos have from the cell surface of virus-infected cells,
failed [59] . Although commercial non-specific hemadsorption results in erythrocytes adher-
pathogen-free eggs have been used for AIV ing directly to these infected cells, which can
propagation and isolation, their vaccination be observed microscopically [119] . In addition to
history should be carefully checked to confirm ECEs, Madin–Darby canine kidney cells are
the absence of other pathogens that might affect now considered a valuable system for the isola-
AIV isolation [13] . tion of influenza viruses. However, since AIVs
Allantoic fluids negative for virus can be have different host and in vitro growth proper-
passaged into ECEs for up to three passages ties depending on the strain [136,137] and not all
and retested for HA before being recorded host cells are universally permissive to all AIV

of
as negative. However, AIV isolation in ECEs subtypes, virus isolation is effective only when
requires a readily available continuous supply the cell culture is sensitive to the inoculated virus
of fertilized chicken eggs and special incuba- [138] . In addition, this method requires the rapid
tors [127] . It can take days to weeks to achieve transport of specimens to the laboratory, since

ro
high titers, the method is labor- and resource- delays may lead to inactivation of the virus and
intensive and there is a mandatory requirement hence failure to isolate any infectious agent [139] .
of a high level of biosecurity facilities (generally Although conventional cell culture takes up
biosafety level 3) if HPAI is suspected. Hence, to 2 weeks to generate results, it is very sensi-
it is not widely used for the routine diagnosis tive. Cytopathic effects such as intracytoplasmic
rP
of influenza infection. On the other hand, egg basophilic inclusion bodies are observed. The
isolation provides high quantities of live infec- presence of influenza virus can be ascertained
tious virus and reference laboratories therefore using either hemadsorption with guinea pig red
utilize this culture system to ensure high sensi- blood cells [108] or immunofluorescence on cul-
tivity and for biological characterization, full- tured cells. Immunofluorescence has the twin
ho

length sequence analysis and antiviral resistance advantages of higher sensitivity in the detection
studies [111,128] . This method also enables the of positive cultures and can also be used to type
production of virus stocks for epidemiological the isolated virus.
monitoring and vaccine updating, which is a Another useful technique for the culture of
critical requirement for the diagnosis of AI in AIVs is the rapid shell vial culture, which uses
the index cases. single or mixed cell lines using monolayers of
ut

The technique of conventional culture of two different cell types in a single vial [121] .
influenza viruses in a cell culture was introduced This technique has the advantage of enhancing
in 1940s [121] and provides a useful method for sensitivity and shortening the time to detection
the primary isolation of some human or swine through enhancing the viral infectivity of the
A

influenza isolates that do not grow well in ECEs cells by centrifugation (time to diagnosis: 24 h
[118,129] . Since viral culture amplifies the inocu- vs 13 days postinoculation) [140,141] . Sensitivity
lum, it is more sensitive than direct methods of is equivalent to that of conventional tube cul-
detection [13] . Cell cultures can be monitored tures and greater than that of direct fluorescent-
for the development of cytopathic effects, the antibody tests [142] .
manifestation of hemadsorption after the addi-
tion of erythrocytes or for the presence of influ- Molecular methods
enza antigens [121] . Various mammalian and Molecular methods, also known as nucleic acid
bird cell lines have been used to isolate influ- tests, detect viral nucleic acid (i.e., RNA) and
enza viruses. The most commonly used cells promise to shorten the turnaround time for the
are: primary rhesus monkey kidney and rhe- laboratory diagnosis of AIV. There is a range
sus monkey kidney (LLC MK2) cells, African of different types of molecular diagnostic tests.
green monkey kidney cells, mink lung epithelial Until recently, the most commonly used was
cell line (Mv1Lu), buffalo green monkey kid- reverse transcription PCR (RT-PCR), where
ney, Madin–Darby canine kidney, green mon- purified RNA of the virus, isolated from chicken
key continuous cell line (Vero), human lung eggs, cell cultures or from clinical specimens, is

10 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

reverse transcribed into cDNA, which is then become a liability if the sequence change or
exponentially amplified using AIV-specific oli- reassortment includes primer binding sites and
gonucleotide primers [143–148] . Conventional the assay no longer amplifies the altered target
RT-PCR is an end point assay, where the PCR sequences. Consequently, nucleic acid-based
product is analyzed on agarose gel after eth- testing requires routine updating of the primers
idium bromide staining to separate the PCR [13] . Another is the potential for obtaining false-
product according to molecular weight, which negative results that may occur due to the pres-
allows presumptive identification [128,146,149] . ence of inhibitors (which are concentrated along
Its main disadvantage is the ease with which with pathogens during sample processing), low
the assay can be contaminated and the tedi- virus titers, degradation of target RNA before
ous amount of work involved in running and amplification and errors in setting up a reaction
visualizing gels. [158,159] . The problem of RNA quality, in par-
Other nucleic acid test methods include the ticular, is a critical issue, as these tests require
use of RT-PCR with detection by ELISA [150] , RNA samples to be of good quality and the
nucleic acid sequence-based amplification [151] use of degraded RNA or inefficient PCR reac-
and H5 RT-loop-mediated isothermal amplifica- tions can make these methods unreliable and

of
tion [152] . However, none of these can compete may yield inaccurate results [13,107] . Hence, it is
with quantitative real-time PCR (RT-qPCR), important to follow a rigid quality assessment
which is a homogeneous assay requiring minimal program and, taking the example of RT-qPCR,
hands-on time and no post-PCR processing [153] . there is a requirement to report the protocols

ro
RT-qPCR typically uses a fluorescently labeled accurately and comprehensively using the mini-
probe to detect the increase in PCR product mum information for publication of quantitative
while the test is being performed and the results real-time PCR experiments (MIQE) guidelines
are reported in real-time. All these methods have [160] . Even then, the conversion of mRNA to
the potential to provide rapid and sensitive diag- cDNA is highly variable, and RT-qPCR results
rP
nostic results. can vary considerably with the choice of reverse
RNA is most commonly extracted from AIVs transcriptase.
using phenol-guanidinium thiocyanate [154] or mRNA molecules are not linear molecules,
one of the recently developed commercially avail- but instead form secondary structures through
able kits that depend on the adsorption of RNA extensive intrastrand base pairing. This results in
ho

to silica gel-based columns. These methods for numerous stem-loop structures consisting of sin-
the detection of influenza are highly sensitive, gle-stranded loops and double-stranded hairpin
specific and versatile [155] , with the column-based structures of varying length. The efficiency of
methods having the advantage in terms of the the RT step depends heavily on the primers used
reliable generation of intact RNA [156] . Once the for cDNA synthesis not targeting the hairpin
viral RNA is extracted, it can be used not only structures, since the intermolecular (i.e., primer
ut

to identify the isolate as AIV, but also to fur- to mRNA) hybridization kinetics are infavour-
ther determine the subtype and even the strain able compared with intramolecular (i.e., mRNA
by sequence ana­lysis utilizing an array of several to itself) hybridization. Hence, it is important to
influenza-specific primers. The viral genotype choose suitable RT primers that target the loops,
A

can be determined by sequencing some or all rather than the stems. Similarly, it is important
of the AIV genes, although some level of virus to ensure that the amplicon itself is free from
amplification in ECEs or cell culture is required secondary structures at the PCR primer binding
for full-length sequence ana­lysis [121] . Molecular sites, as extensive secondary structures there can
methods are rapid, of acceptable sensitivity, and give rise to significantly different results. It must
provide results within a relatively short time for be noted that assays only determine total patho-
targeted treatment and limitation of the spread gen number and do not provide information as
of infection. This is crucial in situations in to whether a pathogen has the ability to establish
which rapid diagnosis is necessary, such as in an infection or not. Hence, a positive result may
epidemics and in patients with specific medical not necessarily pose a public health threat, and
conditions [120,157] . so there is a requirement for additional assays
There are several concerns with molecular that can determine viability.
diagnostic methods. Foremost is the techni- None of the diagnostic tests discussed so far
cal problem associated with the viral sequence are easily carried out in the field, and none can
that gives rise to both genetic drift and shift. provide virtually instant results. Rapid point-
The exquisite specificity of PCR can suddenly of-care diagnosis is critical for the control and

future science group www.futuremedicine.com 11


Review El Zowalaty, Bustin, Husseiny & Ashour

management of influenza infection in humans, Salmonella-containing vacuole and deliver the


as it enables the fast administration of appro- recombinant antigen into the host cell cytosol
priate antiviral therapy within the first 2 days [177,178] . DNA vaccines delivered by attenuated
of illness [161] , reduces the length of in-patient S. typhimurium SL7207 and boosted with a con-
hospital stay and minimizes the unnecessary use ventional killed vaccine confer protection on
of antibiotics [162] . Rapid antigen capture immu- chickens against infection with the H9 subtype
noassay tests can provide results within 30 min of AIV [179] . Another approach is the utiliza-
or less and are easy to perform [163] . They are tion of SPI2-encoded type III secretion system
based on an immunochromatographic reaction [180] for antigen expression and delivery into the
that uses a monoclonal antibody targeted to a cytoplasm of APCs, which increases safety and
specific antigen of influenza, usually the nucleo- efficacy. Such vaccines have been shown to be
protein. They can therefore detect any AIV [164– very effective in eliciting both CD8 + and CD4 +
167] and include tests such as the Directigen™ T‑cell-mediated immune responses in models of
Flu A and Directigen Flu A and B tests (Becton infectious diseases and cancer [178,181–184] .
Dickinson Diagnostic Systems, MD, USA) and Interestingly, the development of an immune
the BinaxNow ® influenza A and B test (Binax, response to HA glycoprotein is important for the

of
Inc., ME, USA) [163] . protection of chickens against challenge with the
AIV infection [185] . Accordingly, a wide variety
Vaccines of vaccines against AIV has been developed and
Vaccination in poultry can minimize the threat tested in experimental conditions, but only inac-

ro
of AI. Vaccines against AI are important tools tivated whole AIV vaccines and live recombinant
for protecting the poultry industry and humans, fowlpox virus vectors expressing HA vaccines
since vaccines help increase resistance to infec- have been licensed and widely used in various
tion, prevent illness and death, reduce virus countries [186] .
replication and reduce viral transmission to
rP
birds and mammals, including humans. DNA Future perspective
vaccination could provide protective immunity The recent outbreak in humans of a novel
in animal models against different infectious influenza A/H7N9 in China in March 2013
diseases. The basic principle of DNA vaccina- shed light on the importance of surveillance
tion is the induction of an immune response by efforts to alleviate public health concerns of
ho

intramuscular injection or the use of a ‘gene gun’ AI. Extensive global surveillance programs are
of naked DNA (plasmid) encoding the targeted urgently required to help predict the circulating
gene into the host cells [168] . The restrictions of AI strains that may produce potential pandem-
DNA vaccination are vaccine cost, problems ics in humans. Vaccines and molecular immu-
with delivery into birds, low efficiency in birds nology approaches are currently the subject of
and its requirement of large amounts of puri- extensive research efforts aimed at minimiz-
ut

fied plasmids [169] . The reason DNA vaccines ing the threat of AI. These are complemented
are not suitable in the poultry industry include by surveillance programs that are essential for
their time-consuming application, expenses and monitoring the AI dynamics in their recognized
undue stress to the chickens. reservoirs. The outcome of active surveillance for
A

DNA delivery technologies are effective and AIVs in different parts of the world, including
essential for inducing a strong and long-lasting Malaysia, will fill the gap in knowledge regard-
immune response that is required to induce high ing the true disease status in Malaysia, Egypt
and continued levels of antigen production at and several countries in southeast Asia. Similar
proper target sites. A variety of intracellular bac- to many countries in southeast Asia, Malaysia
teria have been utilized as live carriers for effi- is endemic for NDV, but its current AI dis-
cient delivery of either DNA vaccine constructs ease status is unknown. Outbreaks of HP A/
or vaccine antigens [170] directly into professional H5N1 were reported in 2004, 2006 and 2007
APCs [171] . This strategy can elicit humoral and in poultry, although none have been reported
cellular responses against the pathogens from since. The country borders Indonesia, which is
which the target genes are derived [172–175] . endemic for A/H5N1, and Thailand, which has
Live oral attenuated Salmonella typhimurium also had outbreaks of HP A/H5N1 AI. In addi-
is a well-known, useful carrier for the expres- tion, in countries such as Thailand, Indonesia,
sion and delivery of heterologous antigens to and China where pigs are not fully protected
mucosal lymphoid tissue, including the HA of from acquiring infection with any possible avian
AIV [176] . The viable bacteria multiply inside the influenza viruses, the generation of pandemic

12 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

influenza strains is a possibility. It was recently immune responses, thus, search for new AI vac-
reported that the unprotected transport of pigs cine delivery mechanisms should be advocated
and the breach of biosecurity measures could such as nanovaccines. Another drawback of cur-
facilitate the mixed infection of humans and pigs rently applied vaccines used in poultry is the low
with avian viruses and the generation of newer antigen stability in the vaccines and the short
strains of greater pandemic potential [187, 188] . duration of immunity. Worryingly, it has been
This could also explain the emergence of influ- recently reported that independent recombina-
enza A strains of pandemic potential in China tion events between distinct attenuated infec-
and southeast Asia. Because of the lack of active tious laryngotracheitis virus vaccine strains can
surveillance, particularly in duck populations, result in virulent recombinant viruses [190] . This
the true disease status in such regions remains prospect might arise in AIVs as well. Although
undetermined. the use of nanotechnology to deliver AI vaccines
Although the use of influenza vaccines to may improve vaccine efficacy and overcome the
protect the poultry industry and humans is an recombination challenges [191,192] , for now, we
effective procedure to control the disease, the advocate strict biosecurity procedures coupled
current use of vaccination encounters several with the implementation of worldwide intera-

of
challenges [189] . A major hurdle is the increased gency surveillance efforts modeled on those of
chance of having asymptomatically infected the USA and Europe in high-risk regions, such
poultry spreading the virus. In addition, poor as in developing countries, to identify potentially
administration is the most common cause of pandemic strains [193–195] . Surveillance and diag-

ro
vaccine failure in poultry making the available nosis of AI in humans and birds are not similar
AI vaccines are not efficient to elicit protective and involve different techniques. Thus, AIV
Executive summary
Background & history
rP
„„Avian influenza virus (AIV), which is closely related to human influenza virus, appeared nearly 150 years go and has recently re-
emerged, causing grave concern among certain populations.
„„Several pandemics of AIV have afflicted human populations with devastating impact.

„„The recently emerged viruses, mainly A/H5N1 and A/H7N9, arose from several reassortment events in different hosts and pose a

significant threat to human public health.


ho

Genome & virology of AIV


„„Although the structure of AIV has been fully elucidated, it remains difficult to predict when the next pandemic will occur.
„„Hemagglutinin and neuraminidase are the major antigenic determinants of influenza A viruses.

„„Acquisition of a polybasic hemagglutinin cleavage site is not the only necessary step for the evolution of low-pathogenicity avian

influenza strains into high-pathogenicity avian influenza viruses. There are other factors that are also required.
ut

Epidemiology of AIV
„„AIVs are natural inhabitants of waterfowl, mainly migratory ducks.
„„Several interspecies barriers determine the transmissibility of AIVs among different species and, finally, to humans.

„„Surveillance is of importance in order to monitor the current status of avian influenza in populations and to update vaccine databases.
A

„„Global programmed surveillance programs in Asia, a potential origin of the next pandemic, should be promptly initiated.

Diagnosis of AIV
„„Diagnosis of avian influenza infection in waterfowl is different from diagnosis in humans.
„„Suitable and reliable diagnostic techniques in real time will guard against severe infection in humans and allow monitoring of AIVs

spreading in their natural reservoirs.


„„Virus isolation in embryonating chicken eggs should be advocated as the cornerstone diagnostic technique of isolation of live AIVs for

subsequent characterization and updating of vaccine databases.


„„Nucleic acid-based tests are useful techniques for the screening of AIVs during surveillance studies.

„„The specificity of quantitative PCR is subject to several factors, such as the primers, RNA extraction method, RNA integrity and presence

of inhibitors, among others.


„„Virologic and serological techniques, along with rapid molecular methods (e.g., quantitative PCR), will provide information and suitable

intervention strategies to limit the pandemic potential of avian influenza in humans and poultry.
Vaccines
„„Vaccines are useful tools to minimize the threat of avian influenza in poultry, but a concern that is associated with the use of vaccines in
poultry is that vaccines could increase the chance of having asymptomatically infected poultry spreading the virus.
„„DNA vaccines and nanovaccines are promising tools to improve currently applied avian influenza vaccines in poultry.

future science group www.futuremedicine.com 13


Review El Zowalaty, Bustin, Husseiny & Ashour

surveillance in AIV’s natural reservoirs of wild advisable to use complementary methods for
birds is of high importance in order to monitor the identification of AIV, since this approach is
the silent circulating strains and protect poultry most likely to yield a reliable and comprehensive
from being infected with LPAI strains, as well as evaluation of circulating viruses [13,107] .
HPAI. Biosecurity measures, environmental pro-
tection and handling of poultry in at-risk farms Financial & competing interests disclosure
or locations of endemic infections will limit and The authors have no relevant affiliations or financial
control the spread of the disease in humans and involvement with any organization or entity with a
will minimize poultry-to-human transmission. financial interest in or financial conflict with the sub-
Virologic and serological monitoring, along ject matter or materials discussed in the manuscript.
with the use of rapid molecular methods (e.g., This includes employment, consultancies, honoraria,
quantitative PCR), will provide information in stock ownership or options, expert testimony, grants or
real time in order to enable suitable intervention patents received or pending, or royalties.
strategies to limit the AI pandemic and disease No writing assistance was utilized in the production
potential in humans and poultry. Finally, it is of this manuscript.

of
10. Saif YM. Diseases of Poultry. Iowa State Press, 18. Nichols JE, Leduc JW. Influenza.
References IA, USA (2003). In: Vaccines for Biodefense and Emerging and
Papers of special note have been highlighted as:
11. Arzt J, White W, Thomsen B, Brown C. Neglected Diseases. Barrett AT, Stanberry LR
n of interest

Agricultural diseases on the move early in the (Eds). Academic Press, Waltham, MA, USA,
nn of considerable interest

ro
third millennium. Vet. Pathol. 47(1), 15–27 497-525 (2008).
1. World Organization for Animal Health. Avian
(2010). 19. Taubenberger JK, Reid AH, Krafft AE,
influenza. In: Manual of Diagnostic Tests and
12. El Zowalaty ME, Abin M, Chander Y, Redig Bijwaard KE, Fanning TG. Initial genetic
Vaccines for Terrestrial Animals: Mammals,
PT, Goyal SM. Isolation of H5 avian characterization of the 1918 ‘Spanish’
Birds, and Bees. (6th Edition, Volume 1). Office
influenza viruses from waterfowl in the upper influenza virus. Science 275(5307),
rP
International des Epizooties, France, 465–481
midwest region of the United States. Avian 1793–1796 (1997).
(2008).
Dis. 55(2), 259–262 (2011). 20. Taubenberger J, Morens D. 1918 Influenza:
2. Swayne DE. The global nature of avian
13. El Zowalaty ME, Abin M, Raju S et al. the mother of all pandemics. Emerg. Infect.
influenza. In: Avian Influenza. Wiley-
Isolation of avian influenza virus from Dis. 12(1), 15–22 (2006).
Blackwell, NJ, USA, 126–127 (2009).
polymerase chain reaction-negative cloacal 21. Cox NJ, Tamblyn SE, Tam T. Influenza
3. Kurtz J, Manvell RJ, Banks J. Avian influenza
samples of waterfowl. J. Vet. Diagn. Invest. pandemic planning. Vaccine 21(16),
ho

virus isolated from a woman with


23(1), 87–90 (2011). 1801–1803 (2003).
conjunctivitis. Lancet 348(9031), 901–902
(1996). n First comprehensive study in literature on 22. Hehme N, Engelmann H, Künzel W,
the isolation of AI viruses from waterfowl Neumeier E, Sänger R. Pandemic
4. Webster RG, Geraci J, Petursson G, Skirnisson
after passage in embryonating chicken preparedness: lessons learnt from H2N2 and
K. Conjunctivitis in human beings caused by
eggs and prior to rapid RT-qPCR to H9N2 candidate vaccines. Med. Microbiol.
influenza A virus of seals. N. Engl. J. Med.
ut

improve the detection and isolation of live Immunol. 191(3–4), 203–208 (2002).
304(15), 911 (1981).
influenza viruses for further 23. Newcastle disease. In: Manual of Standards for
5. Webster RG, Bean WJ, Gorman OT,
characterization. It is imperative to Diagnostic Tests and Vaccines. Office
Chambers TM, Kawaoka Y. Evolution and
perform virus isolation during AI International des Epizooties, France, 161–169
ecology of influenza A viruses. Microbiol. Mol.
surveillance efforts to obtain a clear (1996).
A

Biol. Rev. 56(1), 152–179 (1992).


picture on the circulating AIV and to 24. Garten RJ, Davis CT, Russell CA et al.
6. Kilbourne ED. Influenza immunity: new
understand that depending solely on Antigenic and genetic characteristics of
insights from old studies. J. Infect. Dis. 193(1),
qRT-PCR may yield spurious results. swine-origin 2009 A (H1N1) influenza
7–8 (2006).
viruses circulating in humans. Science
14. Garten RJ, Davis CT, Russell CA et al.
7. Lindstrom S, Cox N, Klimov A. Genetic 325(5937), 197–201 (2009).
Antigenic and genetic characteristics of
analysis of human H2N2 and early H3N2
swine-origin 2009 A(H1N1) influenza viruses 25. Igarashi M, Ito K, Yoshida R, Tomabechi D,
influenza viruses, 1957–1972: evidence
circulating in humans. Science 325(5937), Kida H, Takada A. Predicting the antigenic
for genetic divergence and multiple
197–201 (2009). structure of the pandemic (H1N1) 2009
reassortment events. Virology 328(1), 101–119
influenza virus hemagglutinin. PLoS ONE
(2004). 15. Mounts AW, Kwong H, Izurieta HS et al.
5(1), e8553 (2010).
Case–control study of risk factors for avian
8. Palese P Shaw ML Orthomyxoviridae: the
influenza A (H5N1) disease, Hong Kong, 26. Smith GJ, Vijaykrishna D, Bahl J et al.
viruses and their replication. In: Fields’
1997. J. Infect. Dis. 180(2), 505–508 (1999). Origins and evolutionary genomics of the
Virology. Knipe DM, Howley PM, Griffin
2009 swine-origin H1N1 influenza A
DM, Lamb RA, Martin MA (Eds). 16. Alexander DJ. Avian influenza: historical
epidemic. Nature 459(7250), 1122–1125
Lippincott, Williams & Wilkins, PA, USA, aspects. Avian Dis. 47, 4–13 (2003).
(2009).
1647–1689 (2007). 17. Lupiani B, Reddy SM. The history of avian
27. Zhang W, Qi J, Shi Y et al. Crystal structure
9. Capua I, Alexander DJ. Avian influenza and influenza. Comp. Immunol. Microbiol. Infect.
of the swine-origin A (H1N1)-2009 influenza
human health. Acta Tropica 83(1), 1–6 (2002). Dis. 32(4), 311–323 (2009).

14 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

A virus hemagglutinin (HA) reveals similar (H7N9) outbreak in China. N. Engl. J. Med. 55. Hay AJ, Gregory V, Douglas AR, Lin YP.
antigenicity to that of the 1918 pandemic doi:10.1056/NEJMoa1304617 (2013) (Epub The evolution of human influenza viruses.
virus. Protein Cell 1(5), 459–467 (2010). ahead of print). Phil. Trans. R. Soc. Lond. 356(1416), 1861–
28. Perez-Padilla R. Pneumonia and respiratory 43. Liu D, Shi W, Shi Y et al. Origin and 1870 (2001).
failure from swine-origin influenza A (H1N1) diversity of novel avian influenza A H7N9 56. Bouvier NM, Palese P. The biology of
in Mexico. N. Engl. J. Med. 361(7), 680 viruses causing human infection: influenza viruses. Vaccine 26, D49–D53
(2009). phylogenetic, structural, and coalescent (2008).
29. Smith GJ, Vijaykrishna D, Bahl J et al. analyses. Lancet 381(9881), 1926–1932 57. Steinhauer D, Skehel J. Genetics of influenza
Origins and evolutionary genomics of the (2013). viruses. Annu. Rev. Genet. 36, 305–332
2009 swine-origin H1N1 influenza A 44. Zhu H WD, Kelvin Dj, Li L et al. Infectivity, (2002).
epidemic. Nature 459(7250), 1122–1125 transmission, and pathology of human H7N9 58. García-Sastre A. The neuraminidase of bat
(2009). influenza in ferrets and pigs. Science influenza viruses is not a neuraminidase.
30. Centanni ESE. La peste aviaria I & II. 341(6142), 183–186 (2013). Proc. Natl Acad. Sci. USA 109(46), 18635–
Communicazione fatta all’accademia delle nn Recent outbreaks caused by avian H7N9 18636 (2012).
scienze mediche e naturali de Ferrara. (1901). influenza viruses could be the beginning 59. Tong S, Li Y, Rivailler P et al. A distinct
31. Stubs EL, Biester HE, Devries L. Fowl pest. of a human avain influenza pandemic with lineage of influenza A virus from bats. Proc.
In: Diseases of Poultry. Iowa State Press, IA, possible human-to-human spread. The Natl Acad. Sci. USA 109(11), 4269–4274
USA, 493–502 (1952). finding that efficient transmissibility of (2012).

of
32. Perroncito E. Epizoozia tifoide nei gallinacei. H7N9 viruses in ferrets by direct contact 60. A revision of the system of nomenclature for
Ann. Accad. Agric. Torino. 126, 21–87 (1878). raises grave public health concerns. influenza viruses: a WHO memorandum.
33. Becker WB. The isolation and classification of 45. Uyeki TM, Cox NJ. Global concerns Bull. World Health Organ. 58(4), 585–591
tern virus: influenza virus A/tern/South regarding novel influenza A (H7N9) virus (1980).

ro
Africa/1961. J. Hyg. (Lond.) 64(3), 309–320 infections. N. Engl. J. Med. 368(20), 61. Fouchier RA, Munster V, Wallensten A et al.
(1966). 1862–1864 (2013). Characterization of a novel influenza A virus
34. Subbarao K, Klimov A, Katz J et al. 46. Easterday BC. Animals in the influenza hemagglutinin subtype (H16) obtained from
Characterization of an avian influenza A world. Phil. Trans. R. Soc. Lond. 288(1029), black-headed gulls. J. Virol. 79(5),
rP
(H5N1) virus isolated from a child with a 433–437 (1980). 2814–2822 (2005).
fatal respiratory illness. Science 279(5349), 47. Daly JM, Blunden AS, Macrae S et al. 62. [A revised system of nomenclature for
393–396 (1998). Transmission of equine influenza virus to influenza A viruses: WHO report]. Bull.
35. Webster RG. Predictions for future human English foxhounds. Emerg. Infect. Dis. 14(3), World Health Organ. 57(4), 611–617 (1979).
influenza pandemics. J. Infect. Dis. 461–464 (2008). 63. Fauci AS. Emerging and re-emerging
176(Suppl. 1), S14–S19 (1997). 48. Song D, Kang B, Lee C et al. Transmission of infectious diseases: influenza as a prototype
ho

36. Guo Y, Xu X, Wan X. [Genetic avian influenza virus (H3N2) to dogs. Emerg. of the host–pathogen balancing act. Cell
characterization of an avian influenza A Infect. Dis. 14(5), 741–746 (2008). 124(4), 665–670 (2006).
(H5N1) virus isolated from a sick goose 49. Herfst S, Schrauwen EJ, Linster M et al. 64. Zambon MC. Epidemiology and
in China]. Zhonghua Shi Yan He Lin Airborne transmission of influenza A/H5N1 pathogenesis of influenza. J. Antimicrob.
Chuang Bing Du Xue Za Zhi 12(4), 322–325 virus between ferrets. Science 336(6088), Chemother. 44(Suppl. B), 3–9 (1999).
(1998). 1534–1541 (2012). 65. Desselberger U, Nakajima K, Alfino P et al.
ut

37. Claas EC, Osterhaus AD, van Beek R et al. 50. Imai M, Watanabe T, Hatta M et al. Biochemical evidence that new influenza
Human influenza A H5N1 virus related to a Experimental adaptation of an influenza H5 virus strains in nature may arise by
highly pathogenic avian influenza virus. HA confers respiratory droplet transmission recombination (reassortment). Proc. Natl
Lancet 351(9101), 472–477 (1998). to a reassortant H5 HA/H1N1 virus in Acad. Sci. USA 75(7), 3341–3345 (1978).
A

38. Mounts AW, Kwong H, Izurieta HS et al. ferrets. Nature 486(7403), 420–428 (2012). 66. Olsen B, Munster VJ, Wallensten A,
Case–control study of risk factors for avian 51. Berns KI, Casadevall A, Cohen ML et al. Waldenström J, Osterhaus AD, Fouchier
influenza A (H5N1) disease, Hong Kong, Adaptations of avian flu virus are a cause for RA. Global patterns of influenza A virus in
1997. J. Infect. Dis. 180(2), 505–508 (1999). concern. Science 335(6069), 660–661 (2012). wild birds. Science 312(5772), 384–388
39. Peiris JS, Yu WC, Leung CW et al. (2006).
52. Baltimore D. Expression of animal virus
Re‑emergence of fatal human influenza A genomes. Bacteriol. Rev. 35(3), 235–241 67. Capua I, Alexander DJ. Avian influenza
subtype H5N1 disease. Lancet 363(9409), (1971). infections in birds a moving target. Influenza
617–619 (2004). Other Respi. Viruses 1(1), 11–18 (2007).
53. Virology Division International Union of
40. Banner D, Kelvin AA. The current state of Microbiologival Societies. Virus Taxonomy. 68. Galloway SE, Reed ML, Russell CJ,
H5N1 vaccines and the use of the ferret Eighth Report of the International Committee Steinhauer DA. Influenza HA subtypes
model for influenza therapeutic and on the Taxonomy of Viruses. Fauquet CM, demonstrate divergent phenotypes for
prophylactic development. J. Infect. Dev. Mayo MA, Maniloff J, Desselberger, Ball LA cleavage activation and pH of fusion:
Ctries 6(06), 465–469 (2012). (Eds). Academic Press, MA, USA (2005). implications for host range and adaptation.
41. Bartlett JG. Planning for avian influenza. PLoS Pathog. 9(2), e1003151 (2013).
54. MacLachlan NJ, Dubovi EJ.
Ann. Intern. Med. 145(2), 141–144 (2006). Orthomyxoviridae. In: Fenner’s Veterinary 69. Senne DA, Panigrahy B, Kawaoka Y et al.
42. Li Q, Zhou L, Zhou M et al. Preliminary Virology (4th Edition). Academic Press, MA, Survey of the hemagglutinin (HA) cleavage
report. epidemiology of the avian influenza A USA, 353–370 (2011). site sequence of H5 and H7 avian influenza
viruses: amino acid sequence at the HA

future science group www.futuremedicine.com 15


Review El Zowalaty, Bustin, Husseiny & Ashour

cleavage site as a marker of pathogenicity health. Clin. Microbiol. Rev. 20(2), 243–267 virus in different host species: large-scale
potential. Avian Dis. 40(2), 425–437 (2007). sequence analysis. Virol. J. 6, 67 (2009).
(1996). 83. Webby R, Hoffmann E, Webster R. 97. Munster VJ, Fouchier RA. Avian influenza
70. Vey M, Orlich M, Adler S, Klenk HD, Rott Molecular constraints to interspecies virus: of virus and bird ecology. Vaccine
R, Garten W. Hemagglutinin activation of transmission of viral pathogens. Nat. Med. 10, 27(45), 6340–6344 (2009).
pathogenic avian influenza viruses of S77–S81 (2004). 98. Pedersen K, Swafford SR, Deliberto TJ. Low
serotype H7 requires the protease 84. Couceiro J, Paulson JC, Baum LG. Influenza pathogenicity avian influenza subtypes
recognition motif R-X-K/R-R. Virology virus strains selectively recognize isolated from wild birds in the United States,
188(1), 408–413 (1992). sialyloligosaccharides on human respiratory 2006–2008. Avian Dis. 54(1 Suppl.),
71. Wood GW, McCauley JW, Bashiruddin JB, epithelium; the role of the host cell in 405–410 (2010).
Alexander DJ. Deduced amino acid sequences selection of hemagglutinin receptor 99. Hua YP, Chai HL, Yang SY, Zeng XW, Sun
at the haemagglutinin cleavage site of avian specificity. Virus Res. 29(2), 155–165 (1993). Y. Primary survey of avian influenza virus and
influenza A viruses of H 5 and H 7 subtypes. 85. Shinya K, Ebina M, Yamada S, Ono M, Kasai Newcastle disease virus infection in wild birds
Arch. Virol. 130(1–2), 209–217 (1993). N, Kawaoka Y. Avian flu: influenza virus in some areas of Heilongjiang Province,
72. Stieneke-Gröber A, Vey M, Angliker H. receptors in the human airway. Nature China. J. Vet. Sci. 6(4), 311–315 (2005).
Influenza virus hemagglutinin with 440(7083), 435–436 (2006). 100. Stallknecht DE, Shane SM, Zwank PJ, Senne
multibasic cleavage site is activated by furin, 86. Matrosovich M, Zhou N, Kawaoka Y, Webster DA, Kearney MT. Avian influenza viruses
a subtilisin-like endoprotease. EMBO J. 11(7), R. The surface glycoproteins of H5 influenza from migratory and resident ducks of coastal

of
2407–2414 (1992). viruses isolated from humans, chickens, and Louisiana. Avian Dis. 34(2), 398–405 (1990).
73. Alexander DJ. An overview of the wild aquatic birds have distinguishable 101. Douglas KO, Lavoie MC, Kim LM, Afonso
epidemiology of avian influenza. Vaccine properties. J. Virol. 73(2), 1146–1155 (1999). CL, Suarez DL. Isolation and genetic
25(30), 5637–5644 (2007). 87. Beare AS. Replication of avian influenza characterization of avian influenza viruses and

ro
74. Bogs J, Veits J, Gohrbandt S et al. Highly viruses in humans. Arch. Virol. 119(1–2), 37 a Newcastle disease virus from wild birds in
pathogenic H5N1 influenza viruses carry (1991). Barbados: 2003–2004. Avian Dis. 51(3),
virulence determinants beyond the polybasic 88. Hinshaw VS, Webster RG, Naeve CW, 781–787 (2007).
hemagglutinin cleavage site. PLoS ONE 5(7), Murphy BR. Altered tissue tropism of 102. El Zowalaty ME, Chander Y, Redig PT,
e11826 (2010).
rP
human–avian reassortant influenza viruses. El Latif HKA, El Sayed MA, Goyal SM.
75. Gohrbandt S, Veits J, Hundt J et al. Amino Virology 128(1), 260–263 (1983). Selective isolation of Avian influenza virus
acids adjacent to the haemagglutinin cleavage 89. Ito T, Couceiro JN, Kelm S et al. Molecular (AIV) from cloacal samples containing AIV
site are relevant for virulence of avian basis for the generation in pigs of influenza A and Newcastle disease virus. J. Vet. Diagn.
influenza viruses of subtype H5. J. Gen. Virol. viruses with pandemic potential. J. Virol. Invest. 23(2), 330–332 (2011).
92(1), 51–59 (2011). 72(9), 7367–7373 (1998). 103. Hinshaw VS, Wood JM, Webster RG, Deibel
76. Hatta M, Kawaoka Y. The NB protein of R, Turner B. Circulation of influenza viruses
ho

90. Slemons RD, Easterday BC. Virus replication


influenza B virus is not necessary for virus in the digestive tract of ducks exposed by and paramyxoviruses in waterfowl originating
replication in vitro. J. Virol. 77(10), aerosol to type-A influenza. Avian Dis. 22(3), from two different areas of North America.
6050–6054 (2003). 367–377 (1978). Bull. World Health Organ. 63(4), 711–719
77. Seo SH, Hoffmann E, Webster RG. Lethal (1985).
91. Kim JK, Negovetich NJ, Forrest HL, Webster
H5N1 influenza viruses escape host anti-viral RG. Ducks: the trojan horses of H5N1 104. Jahangir A, Ruenphet S, Ueda S et al. Avian
ut

cytokine responses. Nat. Med. 8(9), 950–954 influenza. Influenza Other Respi. Viruses 3(4), influenza and Newcastle disease viruses from
(2002). 121–128 (2009). northern pintail in Japan: isolation,
78. Hinshaw VS, Webster RG, Turner B. The characterization and inter-annual
92. Janies D, Hill AW, Guralnick R, Habib F,
perpetuation of orthomyxoviruses and comparisons during 2006–2008. Virus Res.
Waltari E, Wheeler WC. Genomic analysis
paramyxoviruses in Canadian waterfowl. 143(1), 44–52 (2009).
A

and geographic visualization of the spread of


Can. J. Microbiol. 26(5), 622–629 (1980). avian influenza (H5N1). Syst. Biol. 56(2), 105. Slemons RD, Johnson DC, Osborn JS, Hayes
79. El Zowalaty ME, Abin M, Chander Y, Redig 321–329 (2007). F. Type-A influenza viruses isolated from wild
PT, Goyal SM. Improved method for the free-flying ducks in California. Avian Dis.
93. Ollier L, Caramella A, Giordanengo V,
isolation and sub-typing of avian influenza 18(1), 119–124 (1974).
Lefebvre JC. High permissivity of human
viruses from oropharyngeal samples of ducks. HepG2 hepatoma cells for influenza viruses. 106. Slemons RD, Shieldcastle MC, Heyman LD,
Avian Dis. 55(3), 439–442 (2011). J. Clin. Microbiol. 42(12), 5861–5865 (2004). Bednarik KE, Senne DA. Type A influenza
80. Stallknecht DE. Ecology and epidemiology of viruses in waterfowl in Ohio and implications
94. Perez DR, Sorrell EM, Donis RO. Avian
avian influenza viruses in wild bird for domestic turkeys. Avian Dis. 35(1),
influenza: an omnipresent pandemic threat.
populations: waterfowl, shorebirds, pelicans, 165–173 (1991).
Pediatr. Infect. Dis. J. 24(11 Suppl.),
cormorants, etc. Avian Dis. 47(Special issue), S208–S216 (2005). 107. Lira J, Moresco KA, Stallknecht DE, Swayne
61–69 (2003). DE, Fisher DS. Single and combination
95. Ferro PJ, El-Attrache J, Fang X et al. Avian
81. Charlton B, Crossley B, Hietala S. diagnostic test efficiency and cost analysis for
influenza surveillance in hunter-harvested
Conventional and future diagnostics for avian detection and isolation of avian influenza
waterfowl from the gulf coast of Texas
influenza. Comp. Immunol. Microbiol. Infect. virus from wild bird cloacal swabs. Avian Dis.
(November 2005–January 2006). J. Wildl.
Dis. 32(4), 341–350 (2009). 54(Suppl. 1), 606–612 (2010).
Dis. 44(2), 434–439 (2008).
82. Peiris JM, de Jong MD, Guan Y. Avian 108. Weinberg A, Mettenbrink CJ, Ye D, Yang CF.
96. Furuse Y, Suzuki A, Kamigaki T, Oshitani H.
influenza virus (H5N1): a threat to human Sensitivity of diagnostic tests for influenza
Evolution of the M gene of the influenza A

16 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

varies with the circulating strains. J. Clin. vaccination. Clin. Vaccine Immunol. 10(3), continuous cell line (Vero) and human lung
Virol. 33(2), 172–175 (2005). 481–482 (2003). embryonated cells (MRC-5) in the isolation
109. Philippa JD. Avian influenza. In: Zoo and 122. Julkunen I, Pyhala R, Hovi T. Enzyme of influenza A virus from nasopharyngeal
Wild Animal Medicine Current Therapy immunoassay, complement fixation and aspirates by shell vial culture. J. Clin.
(6th Edition). Fowler ME (Ed.). Saunders, hemagglutination inhibition tests in the Microbiol. 35(7), 1900–1901 (1997).
PA, USA, 79–87 (2008). diagnosis of influenza A and B virus 134. Schultz-Cherry S, Dybdahl-Sissoko N,
110. Swayne DE, Suarez DL. Highly pathogenic infections. Purified hemagglutinin in McGregor M, Hinshaw VS. Mink lung
avian influenza. Rev. Sci. Tech. 19(2), subtype-specific diagnosis. J. Virol. Methods epithelial cells: unique cell line that supports
463–482 (2000). 10(1), 75–84 (1985). influenza A and B virus replication. J. Clin.
123. Pearson JE, Senne DA. Diagnostic procedures Microbiol. 36(12), 3718–3720 (1998).
111. Suarez DL, Das A, Ellis E. Review of rapid
molecular diagnostic tools for avian influenza for avian influenza. Avian Dis. 47, 222–227 135. Shope RE. Swine influenza : III. Filtration
virus. Avian Dis. 51(1 Suppl.), 201–208 (2003). experiments and etiology. J. Exp. Med. 54(3),
(2007). 124. Smith W, Andrewes CH, Laidlaw PP. A virus 373–385 (1931).

112. Ellström P, Latorre-Margalef N, from influenza patients. Lancet 2, 66 (1933). 136. Webster RG. Influenza virus: transmission
Griekspoor P et al. Sampling for low- 125. Swayne DE, Senne DA, Suarez DL. Avian between species and relevance to emergence of
pathogenic avian influenza A virus in wild influenza. In: A Laboratory Manual for the the next human pandemic. Arch. Virol. 13,
mallard ducks: oropharyngeal versus cloacal Isolation, Identification, and Characterization 105–113 (1997).
swabbing. Vaccine 26(35), 4414–4416 of Avian Pathogens. Dufour-Zavala L (Ed.). 137. Fouchier RA, Bestebroer TM, Herfst S, van

of
(2008). American Association of Avian Pathologists, der Kemp L, Rimmelzwaan GF, Osterhaus
113. Jindal N, De Abin M, Primus AE et al. GA, USA, 128–134 (2008). AD. Detection of influenza A viruses from
Comparison of cloacal and oropharyngeal n Diagnsotic principles followed in the different species by PCR amplification of
samples for the detection of avian influenza conserved sequences in the matrix gene.
identification and characcterization of

ro
virus in wild birds. Avian Dis. 54(1), 115–119 J. Clin. Microbiol. 38(11), 4096–4101 (2000).
avian influenza viruses as well as other
(2010). avian pathogens. 138. Allwinn R, Preiser W, Rabenau H, Buxbaum
114. Roelandt S, Outtrim L, Browning C, S, Stürmer M, Doerr HW. Laboratory
126. Cattoli G, Drago A, Maniero S et al.
Alexander DJ, Brown IH, Irvine RM. diagnosis of influenza – virology or serology?
Comparison of three rapid detection systems
Med. Microbiol. Immunol. 191(3–4), 157–160
rP
Evaluation of two different swab transport for type A influenza virus on tracheal swabs of
systems in the detection of avian influenza (2002).
experimentally and naturally infected birds.
virus excretion from infected pekin ducks Avian Pathol. 33(4), 432–437 (2004). 139. Espy MJ, Smith TF, Harmon MW, Kendal
Anas platyrhynchos. J. Virol. Methods AP. Rapid detection of influenza virus by shell
127. Cattoli G, Capua I. Molecular diagnosis of
184(1–2), 8–14 (2012). vial assay with monoclonal antibodies. J. Clin.
avian influenza during an outbreak. Dev. Biol.
115. WHO. Laboratory procedures. In: WHO Microbiol. 24(4), 677–679 (1986).
(Basel) 124, 99–105 (2006).
Manual on Animal Influenza Diagnosis and 140. Stokes CE, Bernstein JM, Kyger SA, Hayden
ho

128. Carman S, Stansfield C, Weber J, Bildfell R,


Surveillance. WHO Press, Switzerland, 98, FG. Rapid diagnosis of influenza A and B by
Van Dreumel T. H3N2 influenza A virus
15–19 (2002). 24-h fluorescent focus assays. J. Clin.
recovered from a neonatal pig in Ontario –
116. Docherty DE, Slota PG. Use of muscovy Microbiol. 26(7), 1263–1266 (1988).
1997. Can. Vet. J. 40(12), 889–890 (1999).
duck embryo fibroblasts for the isolation of 141. Dunn JJ, Gordon C, Kelley C, Carroll KC.
129. Clavijo A, Tresnan DB, Jolie R, Zhou EM.
viruses from wild birds. Methods Cell Sci. Comparison of the Denka-Seiken INFLU
Comparison of embryonated chicken eggs
ut

11(3), 165–170 (1988). A.B-Quick and BD Directigen Flu A+B kits


with MDCK cell culture for the isolation of
117. Taubenberger JK, Layne SP. Diagnosis of with direct fluorescent-antibody staining and
swine influenza virus. Can. Vet. J. 66(2),
influenza virus: coming to grips with the shell vial culture methods for rapid detection
117–121 (2002).
molecular era. Mol. Diagn. 6(4), 291–305 of influenza viruses. J. Clin. Microbiol. 41(5),
130. Lee CW, Jung K, Jadhao S, Suarez D. 2180–2183 (2003).
(2001).
A

Evaluation of chicken-origin (DF-1) and


118. Potter C, Zuckerman AJ, Banatvala JE, 142. Chander Y, Jindal N, Stallknecht DE,
quail-origin (QT-6) fibroblast cell lines for
Pattison JR. Influenza. In: Principles and Sreevatsan S, Goyal SM. Full length
replication of avian influenza viruses. J. Virol.
Practice of Clinical Virology. Wiley, NY, USA, sequencing of all nine subtypes of the
Methods 153(1), 22–28 (2008).
199–223 (1987). neuraminidase gene of influenza A viruses
131. Moresco KA, Stallknecht DE, Swayne DE. using subtype specific primer sets. J. Virol.
119. Newton DW, Treanor JJ, Menegus MA. Evaluation and attempted optimization of Methods 165(1), 116–120 (2010).
Clinical and laboratory diagnosis of influenza avian embryos and cell culture methods for
virus infections. Am. J. Manag. Care 143. Hoffmann E, Stech J, Guan Y, Webster RG,
efficient isolation and propagation of low
6(Suppl. 5), S265–S275 (2000). Perez DR. Universal primer set for the full
pathogenicity avian influenza viruses. Avian
length amplification of all influenza A viruses.
120. Petric M, Comanor L, Petti CA. Role of the Dis. 54(1 Suppl.), 622–626 (2010).
Arch. Virol. 146(12), 2275–2289 (2001).
laboratory in diagnosis of influenza during 132. Munster VJ, Baas C, Lexmond P et al.
seasonal epidemics and potential pandemics. 144. Jindal N, Chander Y, de Abin M et al.
Practical considerations for high-throughput
J. Infect. Dis. 194(Suppl. 2), S98–S110 Amplification of four genes of influenza A
influenza A virus surveillance studies of wild
(2006). viruses using a degenerate primer set in a one
birds by use of molecular diagnostic tests.
step RT-PCR method. J. Virol. Methods
121. Prince HE, Leber AL. Comparison of J. Clin. Microbiol. 47(3), 666–673 (2009).
160(1–2), 163–166 (2009).
complement fixation and hemagglutination 133. Reina J, Fernandez-Baca V, Blanco I, Munar
inhibition assays for detecting antibody 145. Lee MS. Identification and subtyping of avian
M. Comparison of Madin–Darby canine
responses following influenza virus influenza viruses by reverse transcription-
kidney cells (MDCK) with a green monkey

future science group www.futuremedicine.com 17


Review El Zowalaty, Bustin, Husseiny & Ashour

PCR. J. Virol. Methods 97(1–2), 13–22 transcriptase-polymerase chain reaction 168. Lai WC, Bennett M. DNA vaccines. Crit.
(2001). control and its application to avian influenza Rev. Immunol. 18(5), 449–484 (1998).
146. Bustin SA. Quantification of mRNA using diagnosis and quantification. Avian Dis. 169. Gentschev I, Dietrich G, Spreng S et al.
real-time reverse transcription PCR 51(s1), 213–220 (2007). Recombinant attenuated bacteria for the
(RT‑PCR): trends and problems. J. Mol. 158. Das A, Spackman E, Senne D, Pedersen J, delivery of subunit vaccines. Vaccine
Endocrinol. 29(1), 23–39 (2002). Suarez DL. Development of an internal 19(17–19), 2621–2628 (2001).
147. Wacker MJ, Godard MP. Analysis of one-step positive control for rapid diagnosis of avian 170. Mollenkopf H, Dietrich G, Kaufmann SH.
and two-step real-time RT-PCR using influenza virus infections by real-time reverse Intracellular bacteria as targets and carriers
SuperScript III. J. Biomol. Tech. 16(3), transcription-PCR with lyophilized reagents. for vaccination. Biol. Chem. 382(4), 521–532
266–271 (2005). J. Clin. Microbiol. 44(9), 3065–3073 (2006). (2001).
148. Munch M, Nielsen LP, Handberg KJ, 159. Bustin SA, Benes V, Garson JA et al. The 171. Husseiny MI, Hensel M. Evaluation of an
Jorgensen PH. Detection and subtyping (H5 MIQE guidelines: minimum information for intracellular-activated promoter for the
and H7) of avian type A influenza virus by publication of quantitative real-time PCR generation of live Salmonella recombinant
reverse transcription-PCR and PCR-ELISA. experiments. Clin. Chem. 55(4), 611–622 vaccines. Vaccine 23(20), 2580–2590 (2005).
Arch. Virol. 146(1), 87–97 (2001). (2009).
172. Husseiny MI, Hensel M. Rapid method for
149. Dybkær K, Munch M, Handberg KJ, nn Comprehensive set of guidelines that the construction of Salmonella enterica serovar
Jørgensen PH. Application and evaluation of describe the minimum information Typhimurium vaccine carrier strains. Infect.
RT-PCR-ELISA for the nucleoprotein and necessary for evaluating qPCR experiments Immun. 73(3), 1598–1605 (2005).

of
RT-PCR for detection of low-pathogenic H5 to encourage better experimental practice, 173. Li L, Fang W, Li J, Huang Y, Yu L. Oral
and H7 subtypes of avian influenza virus. allowing more reliable and unequivocal DNA vaccination with the polyprotein gene
J. Vet. Diagn. Invest. 16(1), 51–56 (2004). interpretation of qPCR results. of infectious bursal disease virus (IBDV)
150. Collins RA, Ko LS, So KL, Ellis T, Lau LT, 160. Couch RB. Prevention and treatment of delivered by the attenuated Salmonella elicits

ro
Yu AC. Detection of highly pathogenic and influenza. N. Engl. J. Med. 343(24), protective immune responses in chickens.
low pathogenic avian influenza subtype H5 1778–1787 (2000). Vaccine 24(33–34), 5919–5927 (2006).
(Eurasian lineage) using NASBA. J. Virol. 161. Barenfanger J. Clinical and financial benefits 174. Michael A, Stratford R, Khan S, Dalgleish A,
Methods 103(2), 213–225 (2002). of rapid detection of respiratory viruses: Pandha H. Novel strains of Salmonella
rP
151. Imai M, Ninomiya A, Minekawa H et al. an outcomes study. J. Clin. Microbiol. 38(8), typhimurium as potential vectors for gene
Development of H5-RT-LAMP (loop- 2824–2828 (2000). delivery. FEMS Microbiol. Lett. 238(2),
mediated isothermal amplification) system for 162. Rahman M, Vandermause MF, Kieke BA,
345–351 (2004).
rapid diagnosis of H5 avian influenza virus Belongia EA. Performance of Binax NOW 175. Liljebjelke KA, Petkov DI, Kapczynski DR.
infection. Vaccine 24(44–46), 6679–6682 Flu A and B and direct fluorescent assay in Mucosal vaccination with a codon-optimized
(2006). comparison with a composite of viral culture hemagglutinin gene expressed by attenuated
152. Spackman E, Senne DA, Myers TJ et al. or reverse transcription polymerase chain Salmonella elicits a protective immune
ho

Development of a real-time reverse reaction for detection of influenza infection response in chickens against highly
transcriptase PCR assay for type A influenza during the 2006 to 2007 season. Diagn. pathogenic avian influenza. Vaccine 28(27),
virus and the avian H5 and H7 hemagglutinin Microbiol. Infect. Dis. 62(2), 162–166 (2008). 4430–4437 (2010).
subtypes. J. Clin. Microbiol. 40(9), 3256– 163. Chua TH, Ellis TM, Wong CW et al. 176. Husseiny MI, Hensel M. Evaluation of
3260 (2002). Performance evaluation of five detection tests Salmonella live vaccines with chromosomal
ut

153. Chomczynski P, Sacchi N. Single-step method for avian influenza antigen with various avian expression cassettes for translocated fusion
of RNA isolation by acid guanidinium samples. Avian Dis. 51(1), 96–105 (2007). proteins. Vaccine 27(28), 3780–3787 (2009).
thiocyanate-phenol-chloroform extraction. 164. Cui S, Tong G. A chromatographic strip test 177. Husseiny MI, Wartha F, Hensel M.
Anal. Biochem. 162(1), 156–159 (1987). for rapid detection of one lineage of the H5 Recombinant vaccines based on translocated
154. Song DS, Lee YJ, Jeong OM et al. Evaluation subtype of highly pathogenic avian influenza. effector proteins of Salmonella pathogenicity
A

of a competitive ELISA for antibody detection J. Vet. Diagn. Invest. 20(5), 567–571 (2008). island 2. Vaccine 25(1), 185–193 (2007).
against avian influenza virus. J. Vet. Sci. 10(4), 165. Ho HT, Qian HL, He F et al. Rapid detection 178. Pan Z, Zhang X, Geng S et al. Priming with a
323–329 (2009). of H5N1 subtype influenza viruses by antigen DNA vaccine delivered by attenuated
155. Muyal JP, Muyal V, Kaistha BP, Seifart C, capture enzyme-linked immunosorbent assay Salmonella typhimurium and boosting with a
Fehrenbach H. Systematic comparison of using H5- and N1-specific monoclonal killed vaccine confers protection of chickens
RNA extraction techniques from frozen and antibodies. Clin. Vaccine Immunol. 16(5), against infection with the H9 subtype of
fresh lung tissues: checkpoint towards gene 726–732 (2009). avian influenza virus. Vaccine 27(7),
expression studies. Diagn. Pathol. 4(1), 9 166. Wada A, Sakoda Y, Oyamada T, Kida H.
1018–1023 (2009).
(2009). Development of a highly sensitive 179. Russmann H, Shams H, Poblete F, Fu Y,
156. van Elden LJ, van Kraaij MG, Nijhuis M et al. immunochromatographic detection kit for H5 Galan JE, Donis RO. Delivery of epitopes by
Polymerase chain reaction is more sensitive influenza virus hemagglutinin using silver the Salmonella type III secretion system for
than viral culture and antigen testing for the amplification. J. Virol. Methods 178(1), 82–86 vaccine development. Science 281(5376),
detection of respiratory viruses in adults with (2011). 565–568 (1998).
hematological cancer and pneumonia. Clin. 167. Wolff JA, Malone RW, Williams P et al. 180. Evans DT, Chen LM, Gillis J et al. Mucosal
Infect. Dis. 34(2), 177–183 (2002). Direct gene transfer into mouse muscle priming of simian immunodeficiency virus-
157. Borm SV, Steensels M, Ferreira H et al. in vivo. Science 247(4949 Pt 1), 1465–1468 specific cytotoxic T-lymphocyte responses in
A universal avian endogenous real-time reverse (1990). rhesus macaques by the Salmonella type III

18 Future Microbiol. (2013) 8(9) future science group


Avian influenza viruses: virology, diagnosis & surveillance Review

secretion antigen delivery system. J. Virol. source for virus reassortment between a and replication of influenza virus. Annu. Rev.
77(4), 2400–2409 (2003). human and an avian influenza viral strain Biochem. 52(1), 467–506 (1983).
181. Nishikawa H, Sato E, Briones G et al. In vivo might lead to the generation of a pandemic
antigen delivery by a Salmonella typhimurium strain. Websites
type III secretion system for therapeutic 188. Lee CW, Suarez DL. Avian influenza virus: 201. World Organization for Animal Health
cancer vaccines. J. Clin. Investig. 116(7), prospects for prevention and control by (OIE). Infection with viruses of notifiable
1946–1954 (2006). vaccination. Anim. Health Res. Rev. 6(1), 1–15 avian influenza. Terrestrial Animal Health
182. Xiong G, Husseiny MI, Song L et al. Novel (2005). Code Chapter 10.4.
cancer vaccine based on genes of Salmonella 189. Lee SW, Markham PF, Coppo MJC et al. www.oie.int/fileadmin/Home/eng/
pathogenicity island 2. Int. J. Cancer 126(11), Attenuated vaccines can recombine to form Internationa_Standard_Setting/docs/pdf/A_
2622–2634 (2010). virulent field viruses. Science 337(6091), TAHSC_Feb_2012_Part_A.pdf
183. Xu X, Husseiny MI, Goldwich A, Hensel M. 188–188. (Accessed 22 May 2012)
Efficacy of intracellular activated promoters 190. Fernando GJ, Chen X, Prow TW et al. Potent 202. WHO. Cumulative number of confirmed
for generation of Salmonella-based vaccines. immunity to low doses of influenza vaccine by human cases of avian influenza A (H5N1)
Infect. Immun. 78(11), 4828–4838 (2010). probabilistic guided micro-targeted skin reported to WHO.
184. Swayne DE, Kapczynski D. Strategies and delivery in a mouse model. PLoS ONE 5(4), www.who.int/influenza/human_animal_
challenges for eliciting immunity against e10266 (2010). interface/HAI_Risk_Assessment/en/index.
avian influenza virus in birds. Immunol. Rev. html

of
191. Poinern GREJ, Le XT, Shan S et al.
225(1), 314–331 (2008). (Accessed 5 May 2013)
Ultrasonic synthetic technique to
manufacture a pHEMA nanopolymeric-based 203. US Interagency Working Group. An early
nn Comprehensive review of AI vaccines and
vaccine against the H6N2 avian influenza detection system for highly pathogenic H5N1
vaccination techniques to maximize
virus: a preliminary investigation. Int. avian influenza in wild migratory birds:
immune responses in poultry with future

ro
J. Nanomed. 6, 2167–2174 (2011). US Interagency Strategic Plan. Washington,
prospectives to imporve the efficiency of
DC, USA (2006).
available AI vaccines. 192. Comin A, Stegeman J, Klinkenberg D,
www.nwhc.usgs.gov/publications/other/
Busani L, Marangon S. Design and results of
185. Swayne DE. Avian influenza vaccines and aisurveillanceplan.pdf
an intensive monitoring programme for avian
therapies for poultry. Comp. Immunol. (Accessed 5 September 2012)
influenza in meat-type turkey flocks during
rP
Microbiol. Infect. Dis. 32(4), 351–363
four epidemics in northern Italy. Zoonoses 204. US Department of the Interior and US
(2009).
Public Health 58(4), 244–251 (2012). Geological Survey. Surveillance plan for the
186. 187. Wuethrich B: Chasing the fickle swine early detection of H5N1 highly pathogenic
193. Kelvin AA, Meloni D, Sansonetti P et al.
flu. Science 299(5612), 1502-1505 (2003). avian influenza virus in migratory birds in the
Influenza monitoring in Sardinia, Italy
187. 188. Zhu Y, Qi X, Cui L, Zhou M, Wang United States: surveillance year 2009 (Edited
identifies H3 subtype in Mediterranean wild
H: Human co-infection with novel avian by Brand CJ) (2009).
migratory birds. J. Infect. Dev. Ctries 6(11),
ho

influenza A H7N9 and influenza A H3N2 http://pubs.usgs.gov/gip/92/pdf/gip-92.pdf


786–797 (2012).
viruses in Jiangsu province, China. The (Accessed 24 September 2012)
194. Lebarbenchon C, Chang C-M, van der Werf
Lancet 381(9883), 2134 (2013). 205. London Laboratory Services Group (2013).
S et al. Influenza A virus in birds during
n First reprot on the dual infection of www.lhsc.on.ca/lab/labstracts_llsg.htm
spring migration in the Camargue, France.
human with both avian and seasonal (Accessed 18 May 2012)
J. Wildl. Dis. 43(4), 789–793 (2007).
influenza viruses suggesting a potential 206. www.cdc.gov/ H1N1flu/images.htm
ut

195. Lamb RA, Choppin PW. The gene structure


(Accessed 5 May 2013)
A

future science group www.futuremedicine.com 19

View publication stats

Potrebbero piacerti anche