ArticlePDF AvailableLiterature Review

Host Range Restriction and Pathogenicity in the Context of Influenza Pandemic

Authors:

Abstract

Influenza A viruses cause pandemics at random intervals. Pandemics are caused by viruses that contain a hemagglutinin (HA) surface glycoprotein to which human populations are immunologically naive. Such an HA can be introduced into the human population through reassortment between human and avian virus strains or through the direct transfer of an avian influenza virus to humans. The factors that determine the interspecies transmission and pathogenicity of influenza viruses are still poorly understood; however, the HA protein plays an important role in overcoming the interspecies barrier and in virulence in avian influenza viruses. Recently, the RNA polymerase (PB2) protein has also been recognized as a critical factor in host range restriction, while the nonstructural (NS1) protein affects the initial host immune responses. We summarize current knowledge of viral factors that determine host range restriction and pathogenicity of influenza A viruses.
Influenza A viruses cause pandemics at random inter-
vals. Pandemics are caused by viruses that contain a
hemagglutinin (HA) surface glycoprotein to which human
populations are immunologically naive. Such an HA can be
introduced into the human population through reassortment
between human and avian virus strains or through the
direct transfer of an avian influenza virus to humans. The
factors that determine the interspecies transmission and
pathogenicity of influenza viruses are still poorly under-
stood; however, the HA protein plays an important role in
overcoming the interspecies barrier and in virulence in
avian influenza viruses. Recently, the RNA polymerase
(PB2) protein has also been recognized as a critical factor
in host range restriction, while the nonstructural (NS1) pro-
tein affects the initial host immune responses. We summa-
rize current knowledge of viral factors that determine host
range restriction and pathogenicity of influenza A viruses.
O
f the 3 types of influenza viruses (A, B, and C), only
influenza A viruses are established in animals other
than humans. Influenza pandemics are caused by viruses
that have a hemagglutinin (HA) to which most humans
have no immune memory. The strains of the 1957 Asian
and 1968 Hong Kong pandemics had HAs derived from an
avian virus. Although little information exists about avian
influenza viruses at the time of the Spanish influenza pan-
demic, the HA of the virus responsible for that outbreak is
also thought to be of avian origin. Since avian influenza
viruses do not replicate efficiently in humans and nonhu-
man primates, they must overcome host range restriction
for the avian virus HA to be introduced into human popu-
lations. The molecular basis for host range restriction is not
well understood; however, HA plays a key role in the
restriction of interspecies transmission.
The Spanish influenza was among the most devastating
infectious diseases in history. At least 20 million people
died worldwide. Antimicrobial agents were not available
in 1918; however, existing evidence suggests that this high
death toll was due to the extreme virulence of the virus.
Although all 8 RNA segments of the Spanish influenza
virus have been sequenced, these sequences offer no
explanation for the high virulence. The Spanish influenza
exemplifies how the magnitude of a pandemic can be
determined by the pathogenicity of the virus.
In this review, we focus on 2 properties of influenza A
viruses as they relate to pandemics, host range restriction
and pathogenicity. Viral factors that affect these properties
are examined.
Viral Proteins Responsible for
Host Range Restriction
Viral Glycoproteins
The HA protein mediates virus binding to sialic acid
(SA)–containing host cell surface molecules and promotes
the release of viral ribonucleoprotein complexes through
membrane fusion. By contrast, the sialidase activity of the
neuraminidase (NA) protein removes SA to liberate newly
synthesized viruses from infected cells. Thus, efficient
virus replication requires the balanced actions of HA
receptor-binding specificity and NA sialidase activity.
HA Receptor Specificity
Influenza virus infectivity is influenced by 2 entities:
SA species (N-acetylneuraminic acid [NeuAc] and N-gly-
colylneuramic acid [NeuGc]) and the type of linkage to
galactose (sialyloligosaccharides terminated by SA linked
to galactose by an α2,6 linkage [Acα2,6Gal] or an α2,3
linkage [Acα2,3Gal]) on the host cell surface. Human
influenza viruses preferentially recognize sialyloligosac-
chrides containing SAα2,6Gal (1,2), matched by mainly
Host Range Restriction and
Pathogenicity in the Context of
Influenza Pandemic
Gabriele Neumann* and Yoshihiro Kawaoka*†‡
Emerging Infectious Diseases • www.cdc.gov/eid • Vol. 12, No. 6, June 2006 881
*University of Wisconsin-Madison, Madison, USA; †University of
Tokyo, Tokyo, Japan; and ‡Japan Science and Technology
Agency, Saitama, Japan
NeuAcα2,6Gal linkages on the epithelial cells of the
human trachea (3). By contrast, avian viruses preferential-
ly recognize SAα2,3Gal sialic acids (1,2), in accordance
with the predominance of sialyoligosaccharides with
SAα2,3Gal linkages on the epithelial cells of duck intes-
tine. The epithelial cells of pig trachea contain both types
of SAs and both types of linkages (4), which likely
explains the high susceptibility of these animals to both
human and avian influenza viruses (5). Pigs may therefore
serve as a “mixing vessel” for reassortment between these
2 viruses and the source of pandemic strains, although no
evidence exists that the 1957 or 1968 pandemic viruses
originated in pigs.
Despite these differences in receptor specificity, avian
viruses can infect humans and have caused lethal infec-
tions (6–8). This fact may be explained by the recent find-
ing that in differentiated cultures of human tracheo-
bronchial epithelium, α2,3-linked SAs were found on cili-
ated cells, whereas α2,6-linked SAs were present on non-
ciliated cells (9). The prevalence of cells that possess α2,6
and/or α2,3-linked SAs in the lower respiratory tract
remains unknown; however, ciliated cells support avian
virus infection. Despite the presence of α2,6-linked SA-
bearing cells in differentiated human tracheobronchial
epithelium, viruses with avian-type receptor specificity
can infect humans, since the index 1997 H5N1 virus iso-
lated from a human preferentially recognized an avian
receptor (10). Nevertheless, for efficient human-to-human
transmission, HA derived from an avian virus must prefer-
entially recognize the human receptor. This notion is sup-
ported by the finding that the earliest isolates in the 1918
(11,12), 1957, and 1968 pandemics preferentially recog-
nized NeuAcα2,6Gal–containing sialyloligosaccharides
(13), even though their HAs were derived from avian
viruses. Conversion of receptor specificity from
SAα2,3Gal to SAα2,6Gal may therefore be critical for
generating pandemic influenza viruses.
Sequence comparison, receptor specificity assays, and
crystallographic analysis have identified amino acid
residues that determine receptor specificity: Gln-226
(found in avian viruses) determines specificity for
SAα2,3Gal, whereas Leu-226 correlates with SAα2,6Gal
specificity in human H2 and H3, but not H1, viruses
(2,13). In all human viruses (with the few exceptions of
early isolates from the Asian influenza outbreak [13]),
Leu-226 is associated with Ser-228, while Gln-226 is asso-
ciated with Gly-228 in avian viruses. For H1 viruses, Asp-
190 (found in human and swine virus isolates) or Glu-190
(found in avian virus isolates) determines preferential
binding to α2,6 or α2,3 linkages, respectively (11–14).
Land-based poultry are thought to play a critical role in
the emergence of pandemic influenza viruses. Compared
to H5N1 viruses isolated from aquatic birds, those isolated
from chickens have significantly lower affinity for
NeuAcα2,3Gal (10), similar to human virus isolates; how-
ever, H5N1 chicken isolates have not acquired preferential
specificity for NeuAcα2,6Gal. H5N1 chicken isolates with
reduced avian receptor specificity share 2 characteristic
features of human viruses, namely, an additional glycosy-
lation site in the globular head region of HA and a deletion
in the NA stalk (see below). Similarly, the receptor speci-
ficity of H9N2 viruses isolated from land-based poultry,
but not of those isolated from aquatic birds, is similar to
that of human isolates (15). Hence, land-based poultry may
serve as an intermediate host that facilitates the conversion
of avian to human-type receptors. Avian viruses in land-
based poultry may, therefore, pose a greater threat to
humans than previously thought.
NA Properties
Since efficient release of virus from infected cells
requires the removal of SA by NA, the receptor-binding
and receptor-destroying properties of HA and NA, respec-
tively, must be balanced. When an avian virus with an N2
NA was introduced into the human population, its SAα2,6
cleavage activity increased (16,17), which suggests it had
adapted to the SAα2,6 receptor specificity of human HAs.
The NA stalk, which separates the head region with the
enzymatic center from the transmembrane and cytoplas-
mic domains, varies in sequence and length, depending on
the virus (18). Typically, shortened stalks result in less effi-
cient virus release since the active site in the head region
cannot efficiently access its substrate (19,20). However,
naturally occurring avian viruses with shortened stalks are
virulent in poultry, and the 1997 H5N1 viruses isolated
from patients in Hong Kong (which are believed to have
been transmitted to humans from poultry) are character-
ized by a deletion in the NA stalk (10). Moreover, most
recent highly pathogenic H5N1 viruses isolated from ter-
restrial poultry possess short NA stalks (21).
In avian species, the intestinal tract is the primary site
of replication, whereas in humans, influenza virus replica-
tion is typically restricted to the respiratory tract. The NA
activity of avian H1N1 viruses is more resistant to the low
pH environment in the upper digestive tract than is its
human or swine-derived counterpart (22). In line with this
finding, highly pathogenic H5N1 viruses can replicate in
the human intestine, causing gastrointestinal symptoms
(23), and are shed in large amounts in stool.
Internal Proteins
Classical coinfection experiments, or reverse genetics
experiments that tested multiple gene combinations of 2
parental viruses, suggest that the genes encoding the
“internal proteins”—namely, RNApolymerase (PB2, PB1,
PA), nucleoprotein (NP), matrix protein (M1, M2), and
PERSPECTIVE
882 Emerging Infectious Diseases • www.cdc.gov/eid • Vol. 12, No. 6, June 2006
nonstructural protein (NS1, NS2/NEP)—also contribute to
host range. The contribution of individual proteins to host
range restriction, however, likely varies, depending on the
test system and the virus strains under investigation.
PB2
PB2 is a component of the viral polymerase complex
and, as such, is essential for viral replication. The 1997
H5N1 human virus isolates in Hong Kong have been
divided into 2 groups on the basis of their pathogenicity in
mice; this classification also generally corresponds to dis-
ease severity in humans (24,25). Reverse genetics studies
have shown that Lys at position 627 of PB2 (found in all
human isolates) determines high pathogenicity in mice,
while Glu at this position (found in all avian isolates)
determines low pathogenicity (26). However, the nature of
the amino acid at position 627 of PB2 does not affect the
cell tropism of the virus but rather its replicative ability in
mice and probably in humans.
Several other findings underline the importance of
residue 627 of PB2: 1) an H7N7 virus isolated from a
patient with fatal pneumonia in the Netherlands in 2003
contained Lys at this position, in contrast to viruses isolat-
ed from nonfatal cases and from chickens (27); 2) some of
the H5N1 viruses isolated from patients in Vietnam are
characterized by Lys-627 in PB2 (28); 3) a single reassor-
tant virus bearing an avian virus PB2 gene against a human
virus background replicated efficiently in avian but not
human cells, a feature that could be traced to the nature of
the amino acid at position 627 of PB2 (29); and 4) ribonu-
cleoprotein complexes reconstituted from human or avian
polymerase and NP proteins identified residue 627 of PB2
as the major determinant of replication efficiency in mam-
malian cells (30). Collectively, these findings suggest that
a Glu-to-Lys mutation at position 627 of the PB2 protein
allows avian viruses to efficiently grow in humans and
implicates Lys at this position as an important host range
determinant.
Other Components of the Replication Complex
In addition to PB2, the remaining 2 polymerase pro-
teins (PB1 and PA) and the nucleoprotein NP may also
contribute to host range. In a minireplicon system, replica-
tion in mammalian cells was more efficient with avian than
with human virus PB1 proteins (30), which suggests that
avian PB1 may have greater activity that could provide a
replicative advantage in mammalian systems. This sce-
nario is especially appealing in light of the finding that
both the 1957 and 1968 pandemic viruses possessed avian
PB1 genes, in addition to avian HA, NA, or both genes
(31,32). In one study, however, an avian PB1 gene severe-
ly restricted replication of a human virus in mammalian
cells and squirrel monkeys (33). These findings seem to
contradict a role of avian PB1 in replication in mammalian
cells. PAand NP proteins have also been implicated in host
range restriction; for example, an avian virus NP segment
against a background of a human virus resulted in attenua-
tion in squirrel monkeys (33). However, because of the
limited data available, whether these findings indicate a
contribution of these gene products to host range restric-
tion or simply reflect incompatibility among the viral gene
segments is unclear.
M Segment
Segment 7 of influenza A viruses encodes the M1
matrix and the M2 ion channel proteins. In coinfection
experiments that selected for reassortants containing a
human virus M gene and an avian virus HA gene, the M
segment of an early human virus (A/PR/8/34, H1N1)
cooperated efficiently with avian virus HAs, whereas M
segments derived from more recent isolates have gradual-
ly lost this ability (34). This finding may suggest that cur-
rently circulating human viruses are less likely to reassort
with avian viruses than their predecessors. If this is the
case, the risk for a global pandemic caused by reassortants
possessing avian HA, NA, or both segments against a
human virus background would be reduced.
Molecular Basis of Pathogenicity
HA Cleavability
The HA protein is synthesized as a precursor protein
that is cleaved into 2 subunits (HA1 and HA2) by host cell
proteases. HA cleavage is a prerequisite for fusion of the
viral and endosomal membranes and, therefore, for viral
infectivity (35). Low pathogenic avian influenza viruses
possess a single Arg residue at the cleavage site, recog-
nized by extracellular, trypsinlike proteases. These pro-
teases are thought to be secreted only by cells of the
respiratory and intestinal tract and consequently limit
infections to these organs. By contrast, highly pathogenic
avian viruses possess multiple basic amino acids at the
cleavage that are recognized by ubiquitous, intracellular,
subtilisin-like proteases that thus trigger systemic infec-
tion. In addition, HAcleavability is affected by the absence
or presence of a carbohydrate side chain near the cleavage
site that may interfere with the accessibility of host pro-
teases to the cleavage site (36). The acquisition of a high-
ly cleavable HA converted an avirulent strain to virulence
in Pennsylvania in 1983 (H5N2), Mexico in 1994 (H5N2),
Italy in 1997 (H7N1), Chile in 2002 (H7N3), and Canada
in 2004 (H7N3) (Table). HA cleavability is, therefore, con-
sidered the major determinant of tissue tropism of avian
influenza viruses (41). This correlation seems to extend to
humans, since all avian viruses that have killed humans
possess a highly cleavable HA (6,7,27), and an H5N1
Host Range and Pathogenicity in Influenza Pandemic
Emerging Infectious Diseases • www.cdc.gov/eid • Vol. 12, No. 6, June 2006 883
mutant virus whose HA cleavage site had been changed to
an avirulent type was attenuated in mice (26).
Role of NS1 in Antagonizing Cellular
Immune Responses
Pathogenesis depends partly on the ability of a virus to
evade or suppress the host immune response. The NS1 pro-
tein, encoded by segment 8, plays a central role in this
process by counteracting the cellular interferon (IFN)
response in a 2-pronged approach: 1) by binding to double-
stranded RNA, thereby suppressing the activation of dou-
ble-stranded RNA-activated protein kinase, a known
stimulator of type I IFN, and 2) by preventing the activa-
tion of transcription factors such as ATF-2/c-Jun, NFkB,
and IRF-3/5/7, all of which stimulate IFN production
(42,43). The NS gene of the 1918 Spanish flu blocked the
expression of IFN-regulated genes in human cells more
efficiently than did the NS gene of the A/PR/8/34 (H1N1)
virus (44), which suggests that the NS genes of highly
pathogenic viruses may be more proficient in counteract-
ing the host immune response than those of less pathogen-
ic viruses.
Viruses containing the NS gene of the 1997 H5N1 virus
are potent inducers of proinflammatory cytokine genes,
particularly tumor necrosis factor-α (TNF-α) and IFN-β in
human primary monocyte-derived macrophages (45).
Similarly, 2003 human H5N1 isolates induce high levels of
proinflammatory cytokines in primary human macro-
phages (46). These in vitro findings are substantiated by
reports of unusually high serum concentrations of
chemokines in patients infected with H5N1 influenza
viruses. High levels of macrophage-derived chemokines
and cytokines were also induced by a recombinant virus
containing a gene segment of the 1918 Spanish flu; in this
case, however, the HA segment stimulated the increased
levels of chemokines and cytokines (14,47). This upregu-
lation of cytokine function at later phases of infection may
account for the unusual clinical signs and symptoms and
the degree of disease severity associated with human infec-
tions of highly pathogenic influenza viruses.
Highly pathogenic H5N1 viruses not only trigger the
overproduction of proinflammatory cytokines but also are
resistant to the antiviral effects of IFN and TNF-α.
Pretreatment of porcine lung epithelial cells with IFN-α,
IFN-γ, or TNF-α has no effect on the replication of a
recombinant human H1N1 virus possessing the NS gene of
the 1997 H5N1 virus but abolishes replication of the
parental human H1N1 virus (48,49). Resistance to the
antiviral effects of IFN and TNF-α is associated with glu-
tamic acid at position 92 of the NS1 protein, as demon-
strated by reverse genetics studies. These in vitro data
extend to in vivo findings, since pigs infected with a virus
containing Glu-92 in NS1 experience higher virus titers
and body temperatures than those infected with a control
virus (48,49). Collectively, these findings indicate that
NS1 induces a cytokine imbalance that likely contributes
to the extreme pathogenicity of avian influenza viruses in
humans.
Conclusions
One might speculate that the next pandemic may be
caused by highly pathogenic H5N1 viruses that acquire the
ability to be efficiently transmitted among humans, or by
H9N2 viruses, which are as prevalent as H5N1 viruses in
Asia and in some cases already recognize human recep-
tors. Further investigation of the molecular basis of host
range restriction is therefore important. In addition, a bet-
ter understanding of the mechanisms and consequences of
PERSPECTIVE
884 Emerging Infectious Diseases • www.cdc.gov/eid • Vol. 12, No. 6, June 2006
chemokine/cytokine imbalance caused by highly patho-
genic avian viruses is essential, as is a greater appreciation
for the contributions of other viral properties, such as
replicative ability, to pathogenesis.
Dr Neumann is a research associate professor at the
University of Wisconsin-Madison, Madison, Wisconsin. Her
research interests are the pathogenicity of influenza and Ebola
viruses and the development of reverse genetics techniques for
negative-strand RNA viruses.
Dr Kawaoka is a professor at the University of Wisconsin-
Madison and at the University of Tokyo, Tokyo, Japan. His
research focuses on the molecular biology and pathogenesis of
influenza and Ebola viruses.
References
1. Rogers GN, Paulson JC. Receptor determinants of human and animal
influenza virus isolates: differences in receptor specificity of the H3
hemagglutinin based on species of origin. Virology. 1983;127:
361–73.
2. Rogers GN, Paulson JC, Daniels RS, Skehel JJ, Wilson IA, Wiley
DC. Single amino acid substitutions in influenza haemagglutinin
change receptor binding specificity. Nature. 1983;304:76–8.
3. Couceiro JN, Paulson JC, Baum LG. Influenza virus strains selective-
ly recognize sialyloligosaccharides on human respiratory epithelium;
the role of the host cell in selection of hemagglutinin receptor speci-
ficity. Virus Res. 1993;29:155–65.
4. Ito T, Couceiro JN, Kelm S, Baum LG, Krauss S, Castrucci MR, et al.
Molecular basis for the generation in pigs of influenza A viruses with
pandemic potential. J Virol. 1998;72:7367–73.
5. Kida H, Ito T, Yasuda J, Shimizu Y, Itakura C, Shortridge KF, et al.
Potential for transmission of avian influenza viruses to pigs. J Gen
Virol. 1994;75:2183–8.
6. Claas EC, Osterhaus AD, Van Beek R, de Jong JC, Rimmelzwaan GF,
Senne DA, et al. Human influenza A H5N1 virus related to a highly
pathogenic avian influenza virus. Lancet. 1998;351:472–7.
7. Subbarao K, Klimov A, Katz J, Regnery H, Lim W, Hall H, et al.
Characterization of an avian influenza A (H5N1) virus isolated from
a child with a fatal respiratory illness. Science. 1998;279:393–6.
8. Fouchier RA, Schneeberger PM, Rozendaal FW, Broekman JM,
Kemink SA, Munster V, et al. Avian influenza A virus (H7N7) asso-
ciated with human conjunctivitis and a fatal case of acute respiratory
distress syndrome. Proc Natl Acad Sci U S A. 2004;101:1356–61.
9. Matrosovich MN, Matrosovich TY, Gray T, Roberts NA, Klenk HD.
Human and avian influenza viruses target different cell types in cul-
tures of human airway epithelium. Proc Natl Acad Sci U S A.
2004;101:4620–4.
10. Matrosovich M, Zhou N, Kawaoka Y, Webster R. The surface glyco-
proteins of H5 influenza viruses isolated from humans, chickens, and
wild aquatic birds have distinguishable properties. J Virol.
1999;73:1146–55.
11. Gamblin SJ, Haire LF, Russell RJ, Stevens DJ, Xiao B, Ha Y, et al.
The structure and receptor-binding properties of the 1918 influenza
hemagglutinin. Science. 2004;303:1838–42.
12. Stevens J, Corper AL, Basler CF, Taubenberger JK, Palese P, Wilson
IA. Structure of the uncleaved human h1 hemagglutinin from the
extinct 1918 influenza virus. Science. 2004;303:1866–70.
13. Matrosovich M, Tuzikov A, Bovin N, Gambaryan A, Klimov A,
Castrucci MR, et al. Early alterations of the receptor-binding proper-
ties of H1, H2, and H3 avian influenza virus hemagglutinins after
their introduction into mammals. J Virol. 2000;74:8502–12.
14. Kobasa D, Takada A, Shinya K, Hatta M, Halfmann P, Theriault S, et
al. Enhanced virulence of influenza A viruses with the haemagglu-
tinin of the 1918 pandemic virus. Nature. 2004;431:703–7.
15. Matrosovich MN, Krauss S, Webster RG. H9N2 influenza A viruses
from poultry in Asia have human virus-like receptor specificity.
Virology. 2001;281:156–62.
16. Baum LG, Paulson JC. The N2 neuraminidase of human influenza
virus has acquired a substrate specificity complementary to the
hemagglutinin receptor specificity. Virology. 1991;180:10–5.
17. Kobasa D, Kodihalli S, Luo M, Castrucci MR, Donatelli I, Suzuki Y,
et al. Amino acid residues contributing to the substrate specificity of
the influenza A virus neuraminidase. J Virol. 1999;73:6743–51.
18. Blok J, Air GM. Variation in the membrane-insertion and “stalk”
sequences in eight subtypes of influenza type A virus neuraminidase.
Biochemistry. 1982;21:4001–7.
19. Els MC, Air GM, Murti KG, Webster RG, Laver WG. An 18-amino
acid deletion in an influenza neuraminidase. Virology.
1985;142:241–7.
20. Luo G, Chung J, Palese P. Alterations of the stalk of the influenza
virus neuraminidase: deletions and insertions. Virus Res. 1993;29:
141–53.
21. Li KS, Guan Y, Wang J, Smith GJ, Xu KM, Duan L, et al. Genesis of
a highly pathogenic and potentially pandemic H5N1 influenza virus
in eastern Asia. Nature. 2004;430:209–13.
22. Takahashi T, Suzuki Y, Nishinaka D, Kawase N, Kobayashi Y, Hidari
KI, et al. Duck and human pandemic influenza A viruses retain siali-
dase activity under low pH conditions. J Biochem (Tokyo).
2001;130:279–83.
23. de Jong, Bach VC, Phan TQ, Vo MH, Tran TT, Nguyen BH, et al.
Fatal avian influenza A (H5N1) in a child presenting with diarrhea
followed by coma. N Engl J Med. 2005;352:686–91.
24. Gao P, Watanabe S, Ito T, Goto H, Wells K, McGregor M, et al.
Biological heterogeneity, including systemic replication in mice, of
H5N1 influenza A virus isolates from humans in Hong Kong. J Virol.
1999;73:3184–9.
25. Lu X, Tumpey TM, Morken T, Zaki SR, Cox NJ, Katz JM. A mouse
model for the evaluation of pathogenesis and immunity to influenza
A (H5N1) viruses isolated from humans. J Virol. 1999;73:5903–11.
26. Hatta M, Gao P, Halfmann P, Kawaoka Y. Molecular basis for high
virulence of Hong Kong H5N1 influenza A viruses. Science.
2001;293:1840–2.
27. Fouchier RA, Schneeberger PM, Rozendaal FW, Broekman JM,
Kemink SA, Munster V, et al. Avian influenza A virus (H7N7) asso-
ciated with human conjunctivitis and a fatal case of acute respiratory
distress syndrome. Proc Natl Acad Sci U S A. 2004;101:1356–61.
28. Puthavathana P, Auewarakul P, Charoenying PC, Sangsiriwut K,
Pooruk P, Boonnak K, et al. Molecular characterization of the com-
plete genome of human influenza H5N1 virus isolates from Thailand.
J Gen Virol. 2005;86:423–33.
29. Subbarao EK, London W, Murphy BR. A single amino acid in the
PB2 gene of influenza A virus is a determinant of host range. J Virol.
1993;67:1761–4.
30. Naffakh N, Massin P, Escriou N, Crescenzo-Chaigne B, van der Werf
S. Genetic analysis of the compatibility between polymerase proteins
from human and avian strains of influenza A viruses. J Gen Virol.
2000;81 (Part 5):1283–91.
31. Scholtissek C, Rohde W, Von Hoyningen V, Rott R. On the origin of
the human influenza virus subtypes H2N2 and H3N2. Virology.
1978;87:13–20.
Host Range and Pathogenicity in Influenza Pandemic
Emerging Infectious Diseases • www.cdc.gov/eid • Vol. 12, No. 6, June 2006 885
32. Kawaoka Y, Krauss S, Webster RG. Avian-to-human transmission of
the PB1 gene of influenza A viruses in the 1957 and 1968 pandemics.
J Virol. 1989;63:4603–8.
33. Snyder MH, Buckler-White AJ, London WT, Tierney EL, Murphy
BR. The avian influenza virus nucleoprotein gene and a specific con-
stellation of avian and human virus polymerase genes each specify
attenuation of avian-human influenza A/Pintail/79 reassortant viruses
for monkeys. J Virol. 1987;61:2857–63.
34. Scholtissek C, Stech J, Krauss S, Webster RG. Cooperation between
the hemagglutinin of avian viruses and the matrix protein of human
influenza A viruses. J Virol. 2002;76:1781–6.
35. Garten W, Klenk HD. Understanding influenza virus pathogenicity.
Trends Microbiol. 1999;7:99–100.
36. Kawaoka Y, Naeve CW, Webster RG. Is virulence of H5N2 influenza
viruses in chickens associated with loss of carbohydrate from the
hemagglutinin? Virology. 1984;139:303–16.
37. Garcia M, Crawford JM, Latimer JW, Rivera-Cruz E, Perdue ML.
Heterogeneity in the haemagglutinin gene and emergence of the high-
ly pathogenic phenotype among recent H5N2 avian influenza viruses
from Mexico. J Gen Virol. 1996;77:1493–504.
38. Banks J, Speidel ES, Moore E, Plowright L, Piccirillo A, Capua I, et
al. Changes in the haemagglutinin and the neuraminidase genes prior
to the emergence of highly pathogenic H7N1 avian influenza viruses
in Italy. Arch Virol. 2001;146:963–73.
39. Suarez DL, Senne DA, Banks J, Brown IH, Essen SC, Lee CW, et al.
Recombination resulting in virulence shift in avian influenza out-
break, Chile. Emerg Infect Dis. 2004;10:693–9.
40. Pasick J, Handel K, Robinson J, Copps J, Ridd D, Hills K, et al.
Intersegmental recombination between the haemagglutinin and
matrix genes was responsible for the emergence of a highly pathogen-
ic H7N3 avian influenza virus in British Columbia. J Gen Virol.
2005;86:727–31.
41. Horimoto T, Kawaoka Y. Reverse genetics provides direct evidence
for a correlation of hemagglutinin cleavability and virulence of an
avian influenza A virus. J Virol. 1994;68:3120–8.
42. Garcia-Sastre A. Identification and characterization of viral antago-
nists of type I interferon in negative-strand RNA viruses. Curr Top
Microbiol Immunol. 2004;283:249–80.
43. Krug RM, Yuan W, Noah DL, Latham AG. Intracellular warfare
between human influenza viruses and human cells: the roles of the
viral NS1 protein. Virology. 2003;309:181–9.
44. Geiss GK, Salvatore M, Tumpey TM, Carter VS, Wang X, Basler CF,
et al. Cellular transcriptional profiling in influenza A virus-infected
lung epithelial cells: the role of the nonstructural NS1 protein in the
evasion of the host innate defense and its potential contribution to
pandemic influenza. Proc Natl Acad Sci U S A. 2002;99:10736–41.
45. Cheung CY, Poon LL, Lau AS, Luk W, Lau YL, Shortridge KF, et al.
Induction of proinflammatory cytokines in human macrophages by
influenza A (H5N1) viruses: a mechanism for the unusual severity of
human disease? Lancet. 2002;360:1831–7.
46. Guan Y, Poon LL, Cheung CY, Ellis TM, Lim W, Lipatov AS, et al.
H5N1 influenza: a protean pandemic threat. Proc Natl Acad Sci
U S A. 2004;101:8156–61.
47. Kash JC, Basler CF, Garcia-Sastre A, Carter V, Billharz R, Swayne
DE, et al. Global host immune response: pathogenesis and transcrip-
tional profiling of type A influenza viruses expressing the hemagglu-
tinin and neuraminidase genes from the 1918 pandemic virus. J Virol.
2004;78:9499–511.
48. Seo SH, Hoffmann E, Webster RG. Lethal H5N1 influenza viruses
escape host anti-viral cytokine responses. Nat Med. 2002;8:950–4.
49. Seo SH, Hoffmann E, Webster RG. The NS1 gene of H5N1 influenza
viruses circumvents the host anti-viral cytokine responses. Virus Res.
2004;103:107–13.
Address for correspondence: Yoshihiro Kawaoka, Department of
Pathobiological Sciences, School of Veterinary Medicine, University of
Wisconsin-Madison, 2015 Linden Dr, Madison, WI 53706, USA; email:
kawaokay@svm.vetmed.wisc.edu
PERSPECTIVE
886 Emerging Infectious Diseases • www.cdc.gov/eid • Vol. 12, No. 6, June 2006
Search
past issues
... This preference contributes to virus attachment and efficient replication in the human upper respiratory tract [38]. The human lower respiratory tract contains SAα-2,3Gal, which could explain why avian influenza viruses are able to infect humans and cause a strong host response [39]. ...
Article
Full-text available
Influenza A viruses continue to be a serious health risk to people and result in a large-scale socio-economic loss. Avian influenza viruses typically do not replicate efficiently in mammals, but through the accumulation of mutations or genetic reassortment, they can overcome interspecies barriers, adapt to new hosts, and spread among them. Zoonotic influenza A viruses sporadically infect humans and exhibit limited human-to-human transmission. However, further adaptation of these viruses to humans may result in airborne transmissible viruses with pandemic potential. Therefore, we are beginning to understand genetic changes and mechanisms that may influence interspecific adaptation, cross-species transmission, and the pandemic potential of influenza A viruses. We also discuss the genetic and phenotypic traits associated with the airborne transmission of influenza A viruses in order to provide theoretical guidance for the surveillance of new strains with pandemic potential and the prevention of pandemics.
... The pathogenicity, virulence, and host speci city of in uenza viruses can be primarily ascribed to the functions of three proteins: hemagglutinin (HA), polymerase basic protein 2 (PB2), and nonstructural protein 1 (NS1) 13 . Hemagglutinin plays a critical role in host species restriction, mediating viral entry through receptor-binding speci city 14 . PB2 is a pivotal element of the viral RNA polymerase complex, which also includes polymerase basic protein 1 (PB1), polymerase acidic protein (PA), and nucleoprotein (NP), orchestrating the synthesis of viral RNA 13 . ...
Preprint
Full-text available
The recent expansion of HPAIV H5N1 infections in terrestrial mammals in the Americas, most recently including the outbreak in dairy cattle, emphasizes the critical need for better epidemiological monitoring of zoonotic diseases. In this work, we detected, isolated and characterized the HPAIV H5N1 from environmental swab samples collected from a dairy farm in the state of Kansas (USA). Genomic sequencing of these samples uncovered two distinctive mutations in the PB2 (E249G) and NS1 (R21Q) genes which are rare and absent in recent 2024 isolates of H5N1 circulating in the mammalian avian species. Additionally, approximately 1.7% of the sequence reads indicated a PB2 (E627K) mutation, commonly associated with the virus's adaptation to mammalian hosts. Phylogenetic analyses of the PB2 and NS genes demonstrated more genetic identity between this environmental isolate and the 2024 human isolate (A/Texas/37/2024) of H5N1. Conversely, HA and NA gene analyses revealed a closer relationship between our isolates and those found in other dairy cattle with almost 100% identity, sharing a common phylogenetic subtree. These findings underscore the rapid evolutionary progression of HPAIV H5N1 among dairy cattle and reinforces the need for more epidemiological monitoring which can easily be done using environmental sampling.
... Previous reports showed that acquiring swine-origin FLUAV internal genes is critical [7,8]. Since the temperature of the lower respiratory tract of pigs is slightly higher than that of humans [5], the polymerase complex must be adapted to higher temperatures to overcome this host barrier, potentially through reassortment or gain of adaptative mutations [9,10]. It has also been shown that the HA gene can adapt to the new host via introduction of changes increasing receptor-binding affinity of the HA protein [11,12]. ...
Article
Full-text available
Influenza A virus (FLUAV) infects a wide range of hosts and human-to-swine spillover events are frequently reported. However, only a few of these human viruses have become established in pigs and the host barriers and molecular mechanisms driving adaptation to the swine host remain poorly understood. We previously found that infection of pigs with a 2:6 reassortant virus (hVIC/11) containing the hemagglutinin (HA) and neuraminidase (NA) gene segments from the human strain A/Victoria/361/2011 (H3N2) and internal gene segments of an endemic swine strain (sOH/04) resulted in a fixed amino acid substitution in the HA (A138S, mature H3 HA numbering). In silico analysis revealed that S138 became predominant among swine H3N2 virus sequences deposited in public databases, while 138A predominates in human isolates. To understand the role of the HA A138S substitution in the adaptation of a human-origin FLUAV HA to swine, we infected pigs with the hVIC/11 A138S mutant and analyzed pathogenesis and transmission compared to hVIC/11 and sOH/04. Our results showed that the hVIC/11 A138S virus had an intermediary pathogenesis between hVIC/11 and sOH/04. The hVIC/11 A138S infected the upper respiratory tract, right caudal, and both cranial lobes while hVIC/11 was only detected in nose and trachea samples. Viruses induced a distinct expression pattern of various pro-inflammatory cytokines such as IL-8, TNF-α, and IFN-β. Flow cytometric analysis of lung samples revealed a significant reduction of porcine alveolar macrophages (PAMs) in hVIC/11 A138S -infected pigs compared to sOH/04 while a MHCII low CD163 neg population was increased. The hVIC/11 A138S showed a higher affinity for PAMs than hVIC/11, noted as an increase of infected PAMs in bronchoalveolar lavage fluid (BALF), and showed no differences in the percentage of HA-positive PAMs compared to sOH/04. This increased infection of PAMs led to an overexpression of granulocyte-monocyte colony-stimulating factor (GM-CSF) stimulation but a reduced expression of peroxisome proliferator-activated receptor gamma (PPARγ) in the sOH/04-infected group. Analysis using the PAM cell line 3D4/21 revealed that the A138S substitution improved replication and apoptosis induction in this cell type compared to hVIC/11 but at lower levels than sOH/04. Overall, our study indicates that adaptation of human viruses to the swine host involves an increased affinity for the lower respiratory tract and alveolar macrophages.
... In this review, we refer to these viruses as 2013 low pathogenic avian influenza H7N9 virus (2013 LPAI H7N9 virus). After four years of circulation in nature, a few strains isolated from chickens in Guangdong province in 2017 were found to have acquired 12 extra nucleotides (-aaacggactgcg-) encoding four amino acids (-KGKRTAR/G-) in the cleavage site of HA , a motif for increased virulence of H5 and H7 viruses in chickens (Neumann and Kawaoka, 2006). In subsequent surveillance, four more different motifs were detected in the HA cleavage sites of H7N9 strains: -KRKRAAR/G-, -KRKRTAR/G-, -KGKRIAR/G-, and -KRRRTAR/G-(the four amino acids underlined were insertions, whereas the amino acid R shown in italics was a mutation of the amino acid G at that position), and viruses bearing the motif "-KRKRTAR/G-" were widely detected among viruses isolated from birds and humans Yang et al., 2017). ...
Article
Full-text available
Background: One Health approach is crucial to tackling complex global public health threats at the interface of humans, animals, and the environment. As outlined in the One Health Joint Plan of Action, the international One Health community includes stakeholders from different sectors. Supported by the Bill & Melinda Gates Foundation, an academic community for One Health action has been proposed with the aim of promoting the understanding and real-world implementation of One Health approach and contribution towards the Sustainable Development Goals for a healthy planet. Main text: The proposed academic community would contribute to generating high-quality scientific evidence, distilling local experiences as well as fostering an interconnected One Health culture and mindset, among various stakeholders on different levels and in all sectors. The major scope of the community covers One Health governance, zoonotic diseases, food security, antimicrobial resistance, and climate change along with the research agenda to be developed. The academic community will be supported by two committees, including a strategic consultancy committee and a scientific steering committee, composed of influential scientists selected from the One Health information database. A workplan containing activities under six objectives is proposed to provide research support, strengthen local capacity, and enhance global participation. Conclusions: The proposed academic community for One Health action is a crucial step towards enhancing communication, coordination, collaboration, and capacity building for the implementation of One Health. By bringing eminent global experts together, the academic community possesses the potential to generate scientific evidence and provide advice to local governments and international organizations, enabling the pursuit of common goals, collaborative policies, and solutions to misaligned interests.
... Like most previously detected H5N8 viruses in Egypt, the current HAs of the Egyptian H5N8 viruses had PLREKR-RKR/GLF motif at the HA cleavage site. The viral surface HA glycoprotein is a major determinant of influenza virus and contributed in the virulence , host range and is greatly influenced by HA receptor-binding properties (Matrosovich et al., 2000;Neumann and Kawaoka, 2006). Our results were similar to the previous study in HA glycoprotein of H5N1 in which four mutations (Q222L, G224S, T156A, and H103Y) known to facilitate the transmission of the virus in ferrets via respiratory droplets were reported (Herfst et al., 2012). ...
... Although HA plays a key role in the restriction of interspecies transmission, efficient virus infection and transmission require balanced actions of HA receptor-binding and NA sialidase activity [43]. The HA protein of IAV is the primary target of protective immune responses and is a major component of vaccines. ...
Article
Full-text available
Human-to-swine transmission of influenza A (H3N2) virus occurs repeatedly and plays a critical role in swine influenza A virus (IAV) evolution and diversity. Human seasonal H3 IAVs were introduced from human-to-swine in the 1990s in the United States and classified as 1990.1 and 1990.4 lineages; the 1990.4 lineage diversified into 1990.4.A–F clades. Additional introductions occurred in the 2010s, establishing the 2010.1 and 2010.2 lineages. Human zoonotic cases with swine IAV, known as variant viruses, have occurred from the 1990.4 and 2010.1 lineages, highlighting a public health concern. If a variant virus is antigenically drifted from current human seasonal vaccine (HuVac) strains, it may be chosen as a candidate virus vaccine (CVV) for pandemic preparedness purposes. We assessed the zoonotic risk of US swine H3N2 strains by performing phylogenetic analyses of recent swine H3 strains to identify the major contemporary circulating genetic clades. Representatives were tested in hemagglutination inhibition assays with ferret post-infection antisera raised against existing CVVs or HuVac viruses. The 1990.1, 1990.4.A, and 1990.4.B.2 clade viruses displayed significant loss in cross-reactivity to CVV and HuVac antisera, and interspecies transmission potential was subsequently investigated in a pig-to-ferret transmission study. Strains from the three lineages were transmitted from pigs to ferrets via respiratory droplets, but there were differential shedding profiles. These data suggest that existing CVVs may offer limited protection against swine H3N2 infection, and that contemporary 1990.4.A viruses represent a specific concern given their widespread circulation among swine in the United States and association with multiple zoonotic cases.
... Perbedaan kepekaan protein HA VAI terhadap protease hospes akan berhubungan dengan tingkat virulensi (Shangguan et al, 1998;Callan et al 1997;Puthavathana, 2005 (NeuGc)); 2). Tipe hubungan glikosidik ke galaktosa paling ujung (Acα2-3Gal atau Acα2-6Gal) dan susunan fragmen yang terletak lebih dalam dari sialil-oligosakharida yang terdapat di permukaan sel hospes (Roger & Paulson, 1983;Herrier, 1995;Gambaryan, 2005;Neumann & Kawaoka, 2006). Sebuah varietas dari sialil-oligosakharida yang lain diekspresikan dengan pembatasan (restriksi) ke jaringan dan asal spesies di dalam hospes VAI lainnya. ...
Article
Dengan adanya kejadian luar biasa yang pertama virus avian influenza H5N1 tahun 1997, semakin jelas bahwa potensi virulensi virus H5N1 telah meluas ke manusia. Review ini disusun untuk memahami karakteristik virus, siklus replikasi virus, mekanisme virus masuk ke dalam hospes, peran hemaglutinin sebagai determinan patogenisitas, urutan basa hemaglutinin yang berperan dalam memicu peningkatan virulensi dan fungsi dari 6 segmen gen lainnya pada virus avian influenza. Review juga dibuat untuk memahami gambaran gen Hemaglutinin avian influenza virus H5N1 yang bertujuan untuk mengetahui penularan secara efisien dan replikasi virus avian influenza pada manusia, sehingga penularan selanjutnya dapat diantisipasi dengan baik. Kerja sama lintas sektor antara kementerian kesehatan, kementerian koordinator kesejateraan rakyat, kementerian lain, universitas dan organisasi yang berkompeten sangat dibutuhkan untuk mendukung pencegahan penyebaran virus avian influenza H5N1 di Indonesia.
Preprint
The nature of the interplay between immunity and viral variation is infinitely adaptive. Infection frequently induces immune responses against variation-prone epitopes, rather than against spatially hidden conserved epitopes. It thus remains a substantial challenge to elicit the immune responses to the conserved epitopes providing broad-spectrum immunity. We developed an approach of scaffold-mediated mosaic display to present monomeric influenza virus hemagglutinins (HAs), which exposes highly conserved stem and interface epitopes. Stable monomers were rationally engineered from H1 and H3 subtypes and B type HA trimers, with amino acid mutations at the monomer-monomer interface and for disulfide bond formation, and fused to a self-assembling scaffold, to generate a mosaic HA monomer-displaying nanoparticle, 3HA-np. Immunization with 3HA-np induced broadly neutralizing antibodies (bnAbs) in mice and ferrets and protected against challenges with H1N1 and H3N2 viruses. Competitive immunoassays revealed that 3HA-np induced high interface- and stem-binding Ab titers as compared to head Ab titers, indicating that the monomeric and mosaic nature of 3HA-np elicit cross-reactive Abs. Our results suggest that exposure of the hidden conserved epitope by monomer-displaying nanoparticles is a promising approach to generate a universal influenza vaccine.
Article
A review of the theories of the existence of sexes, genetic diversity, and the distribution of mutations among organisms shows that all these concepts are not a product of random evolution and cannot be explained within the framework of Darwinism. Most mutations are the result of the genome acting on itself. This is an organized process that is implemented very differently in different species, in different places in the genome. Because of the fact that it is not random, this process must be directed and regulated, albeit with complex and not fully understood laws. This means that an additional reason must be included in order to model such mutations during evolution. The assumption of directionality must not only be explicitly included in evolutionary theory but must also occupy a central place in it. In this study an updated model of partially directed evolution is constructed, which is capable of qualitatively explaining the indicated features of evolution. Experiments are described that can confirm or disprove the proposed model.
Article
Full-text available
A highly pathogenic avian influenza virus, H5N1, caused disease outbreaks in poultry in China and seven other east Asian countries between late 2003 and early 2004; the same virus was fatal to humans in Thailand and Vietnam. Here we demonstrate a series of genetic reassortment events traceable to the precursor of the H5N1 viruses that caused the initial human outbreak in Hong Kong in 1997 (refs 2-4) and subsequent avian outbreaks in 2001 and 2002 (refs 5, 6). These events gave rise to a dominant H5N1 genotype (Z) in chickens and ducks that was responsible for the regional outbreak in 2003-04. Our findings indicate that domestic ducks in southern China had a central role in the generation and maintenance of this virus, and that wild birds may have contributed to the increasingly wide spread of the virus in Asia. Our results suggest that H5N1 viruses with pandemic potential have become endemic in the region and are not easily eradicable. These developments pose a threat to public and veterinary health in the region and potentially the world, and suggest that long-term control measures are required.
Article
Full-text available
Reassortant viruses which possessed the hemagglutinin and neuraminidase genes of wild-type human influenza A viruses and the remaining six RNA segments (internal genes) of the avian A/Pintail/Alberta/119/79 (H4N6) virus were previously found to be attenuated in humans. To study the genetic basis of this attenuation, we isolated influenza A/Pintail/79 X A/Washington/897/80 reassortant viruses which contained human influenza virus H3N2 surface glycoprotein genes and various combinations of avian or human influenza virus internal genes. Twenty-four reassortant viruses were isolated and first evaluated for infectivity in avian (primary chick kidney [PCK]) and mammalian (Madin-Darby canine kidney [MDCK]) tissue culture lines. Reassortant viruses with two specific constellations of viral polymerase genes exhibited a significant host range restriction of replication in mammalian (MDCK) tissue culture compared with that in avian (PCK) tissue culture. The viral polymerase genotype PB2-avian (A) virus, PB1-A virus, and PA-human (H) virus was associated with a 900-fold restriction, while the viral polymerase genotype PB2-H, PB1-A, and PA-H was associated with an 80,000-fold restriction of replication in MDCK compared with that in PCK. Fifteen reassortant viruses were subsequently evaluated for their level of replication in the respiratory tract of squirrel monkeys, and two genetic determinants of attenuation were identified. First, reassortant viruses which possessed the avian influenza virus nucleoprotein gene were as restricted in replication as a virus which possessed all six internal genes of the avian influenza A virus parent, indicating that the nucleoprotein gene is the major determinant of attenuation of avian-human A/Pintail/79 reassortant viruses for monkeys. Second, reassortant viruses which possessed the viral polymerase gene constellation of PB2-H, PB1-A, and PA-H, which was associated with the greater degree of host range restriction in vitro, were highly restricted in replication in monkeys. Since the avian-human influenza reassortant viruses which expressed either mode of attenuation in monkeys replicated to high titer in eggs and in PCK tissue culture, their failure to replicate efficiently in the respiratory epithelium of primates must be due to the failure of viral factors to interact with primate host cell factors. The implications of these findings for the development of live-virus vaccines and for the evolution of influenza A viruses in nature are discussed.
Article
During 1997 in Hong Kong, 18 human cases of respiratory illness, including 6 fatalities, were caused by highly pathogenic avian influenza A (H5N1) viruses. Since H5 viruses had previously been isolated only from avian species, the outbreak raised questions about the ability of these viruses to cause severe disease and death in humans. To better understand the pathogenesis and immunity to these viruses, we have used the BALB/c mouse model. Four H5N1 viruses replicated equally well in the lungs of mice without prior adaptation but differed in lethality for mice. H5N1 viruses that were highly lethal for mice were detected in multiple organs, including the brain. This is the first demonstration of an influenza A virus that replicates systemically in a mammalian species and is neurotropic without prior adaptation. The mouse model was also used to evaluate a strategy of vaccination against the highly pathogenic avian H5N1 viruses, using an inactivated vaccine prepared from nonpathogenic A/Duck/Singapore-Q/F119-3/97 (H5N3) virus that was antigenically related to the human H5N1 viruses. Mice administered vaccine intramuscularly, with or without alum, were completely protected from lethal challenge with H5N1 virus. Protection from infection was also observed in 70% of animals administered vaccine alone and 100% of mice administered vaccine with alum. The protective effect of vaccination correlated with the level of virus-specific serum antibody. These results suggests a strategy of vaccine preparedness for rapid intervention in future influenza pandemics that uses antigenically related nonpathogenic viruses as vaccine candidates.
Article
The complement of sialyloligosaccharides present on the surface of human tracheal epithelium has been implicated as an important factor in the selection of hemagglutinin receptor specificity of human influenza A virus. Human strains of influenza A virus preferentially recognize host cell receptors bearing SAalpha2,6Gal sequences, a sequence which is found on the surface of ciliated tracheal epithelium. A fluorescently-labelled H3 human virus strain bound avidly to the apical surface of human tracheal epithelium, while a fluorescently-labelled receptor variant strain, which preferentially binds SAalpha2,3Gal sequences, showed little binding to the epithelial surface and localized primarily to intracellular mucin droplets. Extracts of human bronchial mucin, which is known to contain sialic acid primarily in the SAalpha2,3Gal linkage, was a potent inhibitor of the binding of the receptor variant strain to trachea sections, while the binding of the parent strain was unaffected by the presence of mucin. Human bronchial mucin also inhibited the binding of the receptor variant strains, but not the parent virus strains, to human erythrocytes derivatized to contain SAalpha2,6Gal sequences. These results suggest that a combination of selection pressures present in the respiratory tract environment have resulted in the evolution of a hemagglutinin receptor specificity in human influenza A virus strains which optimizes recognition of, binding to and infection of host cells.
Article
We determined the origin and evolutionary pathways of the PB1 genes of influenza A viruses responsible for the 1957 and 1968 human pandemics and obtained information on the variable or conserved region of the PB1 protein. The evolutionary tree constructed from nucleotide sequences suggested the following: (i) the PB1 gene of the 1957 human pandemic strain, A/Singapore/1/57 (H2N2), was probably introduced from avian species and was maintained in humans until 1968; (ii) in the 1968 pandemic strain, A/NT/60/68 (H3N2), the PB1 gene was not derived from the previously circulating virus in humans but probably from another avian virus; and (iii) a current human H3N2 virus inherited the PB1 gene from an A/NT/60/68-like virus. Nucleotide sequence analysis also showed that the avian PB1 gene was introduced into pigs. Hence, transmission of the PB1 gene from avian to mammalian species is a relatively frequent event. Comparative analysis of deduced amino acid sequences disclosed highly conserved regions in PB1 proteins, which may be key structures required for PB1 activities.
Article
The neuraminidase (NA) of influenza viruses cleaves sialic acids from receptors, prevents self-aggregation and facilitates release of virus during budding from host cells. Although the structure and function of the globular head of the influenza virus NA has been well studied, much less is known about the stalk of the NA, the region between the viral membrane and the globular head. Applying a reverse genetics system, we altered the stalk of the influenza A/WSN/33 virus NA by making deletions, insertions and mutations in this region of the gene. Our data show that the length of the NA stalk can be variable. Deletions of up to 28 amino acids and insertions of up to 41 amino acids in the stalk region did not abolish formation of infectious progeny virus. The data also indicate that the cysteine at position 76 is essential for formation of infectious virus, and that deletions beyond the cysteine did not result in infectious virus. Interestingly, shortening of the length of the stalk region by 28 amino acids resulted in a virus with a markedly reduced growth rate in MDCK cells as compared to that in MDBK cells. An insertion of 41 extra amino acids into the stalk did not significantly interfere with viral growth in MDCK or MDBK cells, which suggests that the stalk region would tolerate the introduction of long foreign sequences.
Article
The haemagglutinin (HA) glycoproteins of influenza virus membranes are responsible for binding viruses to cells by interacting with membrane receptor molecules which contain sialic acid (for review see ref. 1). This interaction is known to vary in detailed specificity for different influenza viruses (see, for example, refs 2-4) and we have attempted to identify the sialic acid binding site of the haemagglutinin by comparing the amino acid sequences of haemagglutinins with different binding specificities. We present here evidence that haemagglutinins which differ in recognizing either NeuAc alpha 2 leads to 3Gal- or NeuAc alpha 2 leads to 6Gal- linkages in glycoproteins also differ at amino acid 226 of HA1. This residue is located in a pocket on the distal tip of the molecule, an area previously proposed from considerations of the three-dimensional structure of the haemagglutinin to be involved in receptor binding.
Article
The RNA of the human influenza virus Singapore (H2N2) strain has been labeled in vivo by phosphorus-32 and separated by polyacrylamide gel electrophoresis into eight segments, which were correlated to the corresponding gene functions and/or proteins. The base sequence homology between the individual genes (segments) of the H2N2 virus and those of different influenza A strains has been determined by molecular hybridization. Segments 1, 5, 7, and 8 of the Singapore strain exhibit a base sequence homology of almost 100% as compared to the FM1 strain (HlNl), while the homology between the other segments is significantly lower (24–76%). For the Singapore and Hong Kong (H3N2) strains all segments except that coding for the hemagglutinin (HA, 24%) exhibit a homology close to 100%. The 32P-labeled segment 4 (HA-gene) of the avian influenza A strain duck Ukraine (Hav7Neg2) shows a homology of 92% to Hong Kong, while the homology of at least two other segments is significantly lower. These results are taken as an indication that the H2N2 subtype is derived from the HlN1 subtype by a recombination event retaining four H1N1 segments, while the other four segments are gained from another yet unknown strain. The H3N2 subtype is presumably derived from a H2N2 subtype, retaining seven segments of the H2N2 subtype, while the gene coding for the HA is obtained from the duck Ukraine or another highly related strain.
Article
A survey of 10 human influenza A viruses of the N2 serotype, isolated between 1957 and 1987, has revealed a drift in neuraminidase linkage specificity. While the earliest N2 strains examined exhibit strict specificity for cleavage of the NeuAc alpha 2,3Gal sequence, N2 isolates from 1967 to 1968 also show limited activity towards the NeuAc alpha 2,6Gal linkage. In strains isolated in 1972 and later, the N2 neuraminidase has approximately equal activity towards both types of linkages. The NeuAc alpha 2,6Gal linkage cleaved by the later N2 neuraminidases is the preferred receptor determinant of human H2 and H3 hemagglutinins. Thus, the acquired neuraminidase specificity of the later isolates allows elution of bound virus from erythrocytes derivatized to contain the NeuAc alpha 2,6Gal linkage, while earlier isolates, which cleave only the NeuAc alpha 2,3Gal sequence, fail to elute from these erythrocytes. These results suggest that the observed drift in N2 neuraminidase specificity in the direction of the preferred H2 and H3 receptor determinant may facilitate release of progeny virus from host cells.
Article
We determined the origin and evolutionary pathways of the PB1 genes of influenza A viruses responsible for the 1957 and 1968 human pandemics and obtained information on the variable or conserved region of the PB1 protein. The evolutionary tree constructed from nucleotide sequences suggested the following: (i) the PB1 gene of the 1957 human pandemic strain, A/Singapore/1/57 (H2N2), was probably introduced from avian species and was maintained in humans until 1968; (ii) in the 1968 pandemic strain, A/NT/60/68 (H3N2), the PB1 gene was not derived from the previously circulating virus in humans but probably from another avian virus; and (iii) a current human H3N2 virus inherited the PB1 gene from an A/NT/60/68-like virus. Nucleotide sequence analysis also showed that the avian PB1 gene was introduced into pigs. Hence, transmission of the PB1 gene from avian to mammalian species is a relatively frequent event. Comparative analysis of deduced amino acid sequences disclosed highly conserved regions in PB1 proteins, which may be key structures required for PB1 activities.