Sei sulla pagina 1di 5

CHAPTER 18: INFLUENZA VIRUS • Acute stage lasts about three days, but cough and

malaise may last for several weeks.


Orthomyxoviridae: Greek ortho (correct or normal) and myxa - More severe pandemics occur at irregular 10- to 50-year
(mucus); virions bind to mucoproteins intervals:
VIRION • “Spanish flu” pandemic in 1918–1920
- Enveloped particles, quasi-spherical or filamentous. • “Asian flu” in 1956 (60,000 deaths in North America)
- Diameter: 80–120 nm.
• “Hong Kong flu” in 1967.
- Envelope is derived from host cell plasma membrane by
• “Swine flu” pandemic of 2009–2010, which caused
budding.
- Compact helical nucleocapsids. upward of 16,000 deaths worldwide, did not lead to
GENOME widespread serious illness as was feared.
- Linear sSRNA, negative sense.
- 6-8 different segments. INFLUENZA VIRUS INFECTIONS OF THE RESPIRATORY TRACT CAN
- Total genome length 10–15 kb. LEAD TO SECONDARY BACTERIAL INFECTIONS
GENES AND PROTEINS
Each genome segment codes for one or two proteins - Initially infects epithelial cells in the upper respiratory tract
• Envelope glycoproteins: hemagglutinin (HA) and neuraminidase - Virus infection: causes loss of the ciliated epithelium, leading
(NA) to loss of the ability of the respiratory tract to clear viruses or
• Integral membrane protein (M2) with ion channel activity bacteria by mucociliary flow that normally traps these agents
• Matrix protein (M1) in mucus and disposes of them.
• Nucleocapsid protein (NP) - Virus replication: induces production of interferons,
• Three RNA polymerase proteins (PA, PB1, and PB2) cytokines, and other soluble response factors, leading to
• Nonstructural protein (NS1) local and systemic inflammatory responses.
• Minor structural protein (NS2)
• Results in the symptoms fever, headache, chills, malaise,
• Additional nonstructural protein (PB1-F2)
and muscle aches.
VIRUSES AND HOSTS
- Resulting secondary bacterial pneumonias cause many of the
- Influenza types A, B, and C: hosts include birds, various
mammals and humans. deaths attributed to influenza virus infection.
- Thogotovirus: transmitted by tick
- Isavirus: infects fish, particularly salmon ORTHOMYXOVIRUSES ARE NEGATIVE-STRAND RNA VIRUSES
DISEASES WITH SEGMENTED GENOMES
- Symptoms: high fever, sore throat, cough, headache, muscular
pain. Myxoviruses: split into two families→ orthomyxoviruses and
- Can be fatal in elderly, infants, and chronically ill, often by paramyxoviruses
secondary bacterial infections.
- 1918: influenza pandemic killed 20 million people worldwide. Orthomyxoviridae family:
- H1N1 swine flu: new pandemic strain spread worldwide in 2009. - Influenza virus types A, B, and C
- Emerging avian influenza virus strains threaten domestic fowl - Thogotovirus: thickborne mammalian virus
and another possible human pandemic. - Isavirus: infects Atlantic salmon
DISTINCTIVE CHARACTERISTICS
- Replicate in the nucleus, unlike most RNA viruses. Orthomyxoviruses
- Viral mRNA synthesis is primed by stealing capped 5' ends of - Negative-strand RNA genome composed of bet 6 and 8
cellular pre-mRNAs in the nucleus.
distinct gene segments.
- Can undergo reassortment (“antigenic shift”) by exchanging
• Individually wrapped within helical nucleocapsids, which
genome segments between related strains.
- Reassortment generates new viruses that can cause pandemics
are packaged in a lipid envelope derived from the
due to changed surface antigens (HA and NA). plasma membrane to form the virion.
• Virions can be either roughly spherical or filamentous
INFLUENZA VIRUSES CAUSE SERIOUS ACUTE DISEASE IN
HUMANS, AND OCCASIONAL PANDEMICS - Influenza type A viruses: responsible for most flu epidemics
and are widespread in many avian and mammalian species.
- Responsible for one of the worst human epidemics - Influenza type B: limited to humans
- Continue to kill hundreds of thousands of humans worldwide - Influenza type C: can infect both pigs and humans.
every year.
• Reason: ability of influenza viruses to escape immune Multiple subtypes are distinguished within each influenza virus
surveillance by changing the amino acid sequences of type by means of variation in the structure of their two surface
their surface glycoproteins. glycoproteins.
- Flu: (the “common cold”) caused not by influenza virus but
by a number of upper respiratory viruses including Filamentous virions:
rhinoviruses and coronaviruses. - Can grow as long as the diameter of an epithelial cell
- Real influenza: severe acute disease in all age groups with - Responsible for cell-to-cell transmission of virus
symptoms of sore throat, cough, high fever, headache, and - Could infect a neighboring cell while still being extruded.
muscular aches and pains. - Seen on fresh isolates
Quasi-spherical virions:
- Smaller, elongated and may be more readily incorporated M2 protein:
into tiny droplets in respiratory aerosols - Small (97 aa protein); has N-terminal external domain, a
- responsible for person-to-person transmission. single transmembrane domain, and a larger internal domain
- Seen on virus repeatedly passaged in eggs or cell cultures - Has the ability to form a highly selective transmembrane ion
channel that allows H+ ions to penetrate the membrane.
EIGHT INFLUENZA VIRUS GENOME SEGMENTS CODE FOR A - Creates a small pore in the viral envelope due to the
TOTAL OF 11 DIFFERENT VIRAL PROTEINS formation of tetramers by lining up four parallel
transmembrane α-helices
- 9 out of 11 viral proteins are packaged in virions
- Genome segments 1 to 6 code for a single protein: M2 H+ion channel:
• Three RNA polymerase subunits (PB2, PB1, and PA) - Facilitates the release of viral nucleocapsids from the virion
• Envelope glycoprotein hemagglutinin (HA) - Allows protons to enter the interior of the virion during
• Nucleocapsid protein (NP) acidification within the endosome
• Second envelope glycoprotein, neuraminidase (NA). • Low pH within virions weakens the interaction of M1
protein w/ viral nucleocapsids, facilitating their release
- PB1-F2: additional short protein coded by second RF on RNA into the cytoplasm upon membrane fusion.
2 by influenza A
- Messenger RNAs: can be spliced and gives rise to two distinct Influenza A virus M2 protein:
viral proteins: - contains a His- XXX-Trp motif (where X can be any amino acid)
• Genome segment 7: codes for the matrix protein (M1) that appears to constitute the main functional element of the
and a third envelope protein (M2). M2 channels.
• Genome segment 8: codes for two nonstructural - only conserved region shared with the similarly functioning
proteins (NS1 and NS2), M2 protein of influenza B virus
- also found in the p7 proteins of some genotypes of hepatitis
HEMAGGLUTININ PROTEIN BINDS TO CELL RECEPTORS AND C virus (aflavivirus).
MEDIATES FUSION OF THE ENVELOPE WITH THE ENDOSOMAL
MEMBRANE NUCLEOCAPSIDS ENTER THE NUCLEUS, WHERE mRNA
SYNTHESIS AND RNA REPLICATION OCCUR
Hemagglutinin protein (HA):
- Type I transmembrane protein w/c has the ability to Nucleocapsids
agglutinate (clump) RBC. - Contain multiple copies of the NP protein that wrap the
- Binds to sialic acid-containing receptors found on RBC and ssRNAs in an unusual twin helical conformation with a central
other cell types. loop
- Also responsible for entry of the virus genome into the cell - Also contain a trimer of RNA polymerase proteins PA, PB1,
via fusion pathway and PB2.
Synthesis of HA:
- Synthesized as a fusion-incompetent protein w/c is inserted Orthomyxoviruses replicate in the nucleus
into the membrane of the ER and transported via the Golgi - Transport and transcription of virions into nucleus → delivery
apparatus to the PM and translation of viral mRNA to cytoplasm → delivery of viral
- Activated by the cleavage of cellular proteases into two proteins to nucleus → direct genome replication and
subunits (HA1 and HA2) formation of nucleocapsid → nucleocapsid transported back
• HA1: surface subunit w/c contains the sialic acid binding to cytoplasm for assembly
domain.
• HA2: anchored in the virus envelope and contains a - Nucleocapsids are imported from the cytoplasm to the
hydrophobic fusion peptide near the amino terminus nucleus via the nuclear pore complexes
created by protease cleavage. - NP protein and polymerase proteins contain nuclear
- Virions will then enter cell w/in an endosomal vesicle after localization signals that mediate interactions with cellular
binding to cell receptor importin-α.
- HA will undergo a conformational change as the pH of the **importin-α: docks with nuclear pore complexes and promotes entry
of its cargo into the nucleus.
endosome drops.
• N-terminal part of HA2 forms a rigid alpha-helical
CAPPED 5' ENDS OF CELLULAR PREMESSENGER RNAS ARE USED
structure, moving the fusion peptide out of the
AS PRIMERS FOR SYNTHESIS OF VIRAL mRNAS
hydrophobic region of HA and toward the endosomal
membrane, where it inserts itself into the membrane
Capped cellular premessenger RNAs are synthesized in the
• One end of HA1 is now bound to the endosomal
nucleus by cellular RNA polymerase II and undergo
membrane, and the other end is bound to the virus
polyadenylation and splicing before being transported as mature
envelope
mRNAs to the cytoplasm.
M2 IS AN ION CHANNEL THAT FACILITATES RELEASE OF
PB2 protein:
NUCLEOCAPSIDS FROM THE VIRION
- Contains a domain that recognizes the 5' cap structure found - Enables the replication machinery to begin to produce full-
on all eukaryotic mRNAs. length, plus-strand copies of the 8 viral genome segments
- Binds to the capped end of a cellular premessenger RNA - full-length transcripts contain 5' triphosphate groups and
PB1: result from unprimed transcription initiation.
- Acts as a nuclease, cleaving the bound premessenger RNA at
an A or G residue Mechanism of switching from primed to unprimed RNA
- Also acts as an RNA polymerase by adding ribonucleotides synthesis:
complementary to the genome RNA sequence, proceeding Interaction of with RNA polymerase complex or with the 5' and
toward its 5' end. 3' ends in nucleocapsid → Begins copying the genome RNA
PA: role in transcription is not well understood directly from its 3' terminus, plus strand copy synthesized →
Plus-strand copy complexed with NP protein, forming
- Capped RNA fragment bound to PB2 is used as a primer to nucleocapsids containing plus-strand antigenome RNAs.
begin copying the genome RNA. - RNA polymerase does not stutter and terminate at the
- Resulting capped viral mRNAs have heterogeneous poly(U) stretch
sequences - Occurs because the PB1 subunit has been displaced
- Orthomyxoviruses can make capped messenger RNAs even
though they do not code for a capping enzyme. - Plus-strand antigenome RNAs are copied in a similar fashion
by the RNA polymerase complex to generate nucleocapsids
- Reason why the replication of influenza virus is blocked by containing full-length, minus-strand genomes
treatment of infected cells with inhibitors of cellular RNA - Minus strand genomes can be transcribed to produce more
synthesis such as actinomycin D or α-amanitin. viral mRNAs or can generate more full-length, plus-strand
- Cellular premessenger RNAs are rapidly spliced and exported RNAs for genome replication
to the cytoplasm - NP does not bind to influenza virus mRNA since they are
- Influenza virus cannot make its own mRNAs. capped and contain cellular mRNA sequences, not viral
sequences, at their 5' ends.
VIRAL mRNAS TERMINATE IN POLY(A) TAILS GENERATED BY
“STUTTERING” TRANSCRIPTION NUCLEOCAPSIDS ARE EXPORTED FROM THE NUCLEUS IN A
COMPLEX WITH MATRIX PROTEIN AND NS2
Transcription of the viral RNA polymerase stutters or pauses when
poly(U) sequence is reached Nucleocapsids must be transported back out of the nucleus to the
- Stuttering reads through the poly(U) sequence a number of cytoplasm, so that they can be incorporated into progeny virions
times and repeatedly adding complementary A residues to
the transcript to generate a poly(A) tail - Matrix protein (M1): imported into the nucleus and binds to
- RNA polymerase eventually terminates transcription at that nucleocapsids to form a complex
position. - NS2:
- Results from blockage of further movement of the RNA • Piggybacks onto nucleocapsid/matrix protein complexes
polymerase by PB1 itself since it remains bound to the and signals the cellular protein export system to
terminal 12 to 13 nt at the 5' end of the same genome RNA it transport the complexes through the nuclear pores to
is copying. the cytoplasm
• Contains a nuclear export signal, and it can also bind to
TWO INFLUENZA A mRNAS UNDERGO ALTERNATIVE SPLICING the matrix protein.
IN THE NUCLEUS
THE NS1 PROTEIN INTERFERES WITH POLYADENYLATION OF
Transcription generates a complete set of 8 viral mRNAs CELLULAR mRNAS
- 6 of these are exported directly to the cytoplasm where
they are translated by ribosomes. NS1 protein: most abundant viral protein in virus-infected cells
- RNAs generated from genome segments 7 and 8 contain - can bind to both single- and double-stranded RNA and to two
splicing consensus sequences that are recognized by the host cell proteins including:
cellular splicing machinery in the nucleus. • Cleavage and polyadenylation specificity factor (CPSF):
- Only a fraction of these two viral mRNAs are spliced, and responsible for the cleavage of cellular pre-mRNAs
both unspliced (M1 and NS1) mRNAs and spliced (M2 and shortly downstream of the consensus AAUAAA
NS2) mRNAs are exported from the nucleus. polyadenylation signal
- Ensures that M2 and NS2 are made in lesser amounts than • poly(A)-binding protein II (PABII): facilitates production
the highly abundant M1 and NS1 proteins. of full-length poly(A) chains on the 3' terminus created
by cleavage.
GENOME REPLICATION BEGINS WHEN NEWLY SYNTHESIZED NP
PROTEIN ENTERS THE NUCLEUS - Binding of NS1 inhibits the normal 3' end cleavage and
polyadenylation of cellular mRNAs; synthesis of viral mRNA is
Viral proteins (NP and the RNA polymerase subunits) are not affected
transported from their site of synthesis in the cytoplasm back
into the nucleus
Influenza PB2 and PB1: its cap-binding and endonuclease - Interact directly with the cytoplasmic tail of M2 to promote
activities leads to the degradation of many cellular pre-mRNAs in recruitment of internal viral proteins and viral nucleocapsids
virus-infected cells. to the plasma membrane, facilitates

- Most cellular pre-mRNAs are degraded in the nucleus, and - Orthomyxovirus virions must contain at least one copy of
cellular mRNA production is therefore suppressed. each genome segment in order to be able to initiate an
infection.
THE NS1 PROTEIN ALSO SUPPRESSES A VARIETY OF HOST CELL - 7 nucleocapsids are arranged parallel to each other as a
ANTIVIRAL RESPONSE PATHWAYS cylindrical barrel and an 8th nucleocapsid lies in the center of
the bundle.
NS1: • One copy of each of the eight different nucleocapsids is
- Binds and inhibits signaling by RIG-1 incorporated into a bundle either before or during virus
**RIG-1: responds to RNA virus infections by the induction of budding.
interferon synthesis via interferon response factor-3. - Viral proteins recognize and interact with specific RNA
- Activates the phosphatidylinositol 3-kinase (PI3K)/Akt sequences in the nucleocapsids to incorporate them, one by
pathway w/c leads to caspase-9 activity inhibition and one, into the bundles that are packaged into virions during
suppression of apoptosis. budding.
- Binds and sequesters dsRNA w/c suppresses the activity of
two major interferon stimulated antiviral mechanisms: NEURAMINIDASE CLEAVES SIALIC ACID, THE CELLULAR
double-stranded, RNA-dependent protein kinase (PKR) and RECEPTOR THAT BINDS TO HA
2', 5'-oligo(A) synthetase/Ribonuclease L.
- Prevents the maturation of human primary dendritic cells, Influenza neuraminidase (NA):
thereby limiting host T-cell activation considered as a - type II transmembrane protein w/ a short cytoplasmic N-
potential target for chemotherapeutic agents to suppress terminus, a membrane-spanning domain, and a long C-
influenza virus infection terminal domain that extends outward from the virus
- Its four C-terminal constitutes PDZ ligand domain of the envelope.
consensus sequence X-Ser/Thr-X-Val (where X is any amino - Has the ability to cleave terminal N-acetyl neuraminic acid
acid). (sialic acid) from oligosaccharides located on mucoproteins,
• Interacts w/ PDZ-binding protein(s) and modulate cell-surface glycoproteins, and glycolipids.
pathogenicity through additional mechanisms - Can reverse binding of HA to sialic acid residues by cleaving
• PDZ domains: protein–protein recognition modules the bound sialic acid residue from the remainder of the
oligosaccharide
PB1-F2 MAY CONTRIBUTE IN SUPPRESSION OF THE HOST
IMMUNE RESPONSE INFLUENZA VIRUS STRAINS VARY IN BOTH TRANSMISSIBILITY
AND PATHOGENICITY
PB1-F2 protein:
- Recently discovered in the PB1 polymerase gene segments of - Most influenza A viruses that circulate in wild or domestic
certain influenza A virus strains. animals do not readily infect humans.
- Shown to overcome host defense mechanisms and to - This host specificity resides in HA, NA, PA, PB1, and PB2 that
enhance pathogenicity by inducing increased apoptosis in have adapted to function optimally in a particular species.
host immune cells responding to influenza virus infection. Example:
- HA proteins of avian influenza A: bind preferentially to α-2,3
VIRAL ENVELOPE PROTEINS ASSEMBLE IN THE PLASMA sialic acid
MEMBRANE AND DIRECT BUDDING OF VIRIONS • In contrast w/ human influenza A w/c bind
preferentially to α -2,6 sialic acid
- The cellular protein trafficking machinery translocates HA, - Some HA proteins are cleaved by furin proteases which
neuraminidase and M2 ion channel via the Golgi network to infects many cell types since their fusion mechanism is
the cell surface, where virus assembly takes place. activated upon cleavage.
- Three proteins accumulate in lipid rafts (membrane enriched - Other HA proteins are not cleaved intracellularly during virus
in cholesterol), which are the site of virion formation by formation but require activation by trypsin cleavage after
budding. release of virions from an infected cell.
- The cytoplasmic tails of HA, NA, and M2 are crucial to virion • Trypsin expression is limited to the upper respiratory
formation since they interact with matrix protein (M1) bound tract in humans,
to virus nucleocapsids and transported from the nucleus to • Less likely to infect lung and other tissues in the body;
the cytoplasm. leads to milder and more self-limiting infections.
- Artificial virus-like particles (VLPs) containing influenza
surface proteins can be assembled in the absence of M1. GENETIC VARIABILITY GENERATES NEW VIRUS STRAINS THAT
• However, M1 is required for the production of infectious CAN CAUSE PANDEMICS
VLPs.
M1:
- Functions to package the genome in virions.
Protective immunity after infection with influenza A virus does not • Appeared in China in 1968,
last for a lifetime as a result from the antigenic change in the • “Hong Kong” influenza pandemic, had a new HA
domains of virus envelope glycoproteins, HA and NA.
Up to the present, influenza viruses that causes widespread
Antigenic change occurs in two ways: human disease have been limited to strains that contain HA types
1. Antigenic drift: 1, 2, or 3.
- Slowly but continuously
- Results from the accumulation of point mutations w/in HIGHLY PATHOGENIC AVIAN INFLUENZA A H5N1 STRAINS IN
regions of the envelope glycoprotein genes POULTRY FARMS ARE A POTENTIAL THREAT BUT ARE POORLY
- Variants in antigenic domain of envelope glycoprotein as a TRANSMITTED AMONG HUMANS
result of errors made by RNA polymerase during replication
since it does not possess proofreading mechanisms avian H5N1 strains:
- Can selectively replicate and propagate - Highly pathogenic strains w/c spread through commercial
- Common to a number of RNA viruses, including HIV, and is a poultry farms in Hong Kong (1997); causing 6 deaths
major complicating factor in production of vaccines. - Resulted in serious and often fatal disease in a number of
- Resulted in the generation of numerous influenza A subtypes people directly exposed to infected birds.
with antigenically distinct HA and NA surface glycoproteins - Further spread probably via infected migrating waterfowl.
- High density of domestic animals and migratory wildfowl
2. Antigenic shift: increase the likelihood of generation of mutants or
- Suddenly but episodically reassortant viruses that will acquire the ability to be
- Results from reassortment of influenza virus genes during transmitted among humans.
mixed infections with two or more virus subtypes. - Neuraminidase inhibitors zanamivir and oseltamivir could
- Cells infected simultaneously with two virus strains can slow down the spread of influenza A virus during a pandemic
produce reassortant virions or reduce symptoms in infected people.
- Viruses with many combinations of the different HA and NA
subtypes, as well as other viral genes, are generated in A NEW PANDEMIC STRAIN OF INFLUENZA A VIRUS AROSE BY
multiply infected animals in the wild GENETIC SHIFT AND SPREAD WORLDWIDE IN 2009
- Reassortment bet. animal and human strains can create a
virus that can replicate in humans but has acquired an HA or New strain of human H1N1 influenza A virus was identified in the
NA subtype derived from the animal virus w/c can cause a United States and Mexico in 2009
pandemic - Genomic analysis showed that it was related to common
reassortant swine influenza A viruses isolated in North
THE 1918 PANDEMIC INFLUENZA A VIRUS WAS PROBABLY NOT America, Europe, and Asia,
A REASSORTANT VIRUS - HA protein of this virus strongly resembles the HA protein of
the original 1918 pandemic virus.
Subtype H1N1: - Drifted during > 40 years when it dominated human influenza
- 1918 pandemic virus strain arose from an avian virus by strains
mutation (“drift”), rather than by reassortment accdng to - WHO raised the warning level for this virus to the highest
analysis of its genome sequence available—Phase Six—indicating that influenza had entered a
- showed extremely high pathogenicity for mice and for pandemic stage of worldwide spread on June 11, 2009.
embryonated chicken eggs as shown in an experiment • 16,713 deaths.
• Occurred in young, healthy people
PA, PB1, and PB2 of the 1918 human virus: Acquired the ability • Most patients had only mild symptoms and a short
to bind to the α -2,6 sialic acid present in the human respiratory course of disease without severe complications or
tract by virtue of a single amino acid change from an HA that binds mortality.
to the avian α -2,3 sialic acid. - Initially susceptible to the neuraminidase inhibitor
oseltamivir (Tamiflu), which helped to shorten the fever
HA: lacks a furin cleavage site, NA protein enhances the cleavage period and quickly improved disease symptoms.
of HA in a trypsin-independent fashion, increasing the infectivity • Strains resistant to the drug were reported within
and spread of this virus. months, leading to controversy about its use in patients
with mild disease.
GENOME SEQUENCES FROM SOME PREVIOUS INFLUENZA A - A massive vaccine development campaign allowed the first
VIRUS STRAINS CONFIRM THE ANTIGENIC SHIFT HYPOTHESIS human vaccination campaigns against pandemic H1N1 virus
to begin in October 2009.
subtype H2N2 and subtype H3N2: subtype H2N2: both - Seasonal strains of influenza A H3N1 and H1N1 as well as
reassortant viruses influenza B virus began to cocirculate with pandemic H1N1
- subtype H2N2: virus.
• New influenza A virus w/c appeared in Southeast China
in 1957
• “Asian flu” pandemic.
- subtype H3N2

Potrebbero piacerti anche