ArticlePDF AvailableLiterature Review

Pathogenesis of Semliki Forest Virus Encephalitis

Authors:

Abstract

This article provides a review of the pathogenesis of Semliki Forest virus (SFV) encephalitis. In mice, outcome of infection varies according to age of the mouse and strain of the virus and can include acute encephalitis, subacute demyelinating meningoencephalomyelitis, and persistent subclinical central nervous system (CNS) infection. All strains of virus are virulent in mice infected <12 days of age. The L10 strain is also virulent in mice >14 days age, whereas the A7(74) strain is avirulent. The genetic difference between these strains maps to the nsp3 gene. For A7(74) virus, age-related virulence correlates with ability of CNS neurons to replicate virus and undergo apoptotic cell death. Immature developing neurons support complete virus replication but as neuronal populations and circuits mature in the postnatal brain, virus infection becomes progressively restricted and nonproductive. This restricted replication can be overcome by gold I compounds, which may function by inducing neuronal dedifferentiation to a state permissive for virus replication. Biochemical pathways associated with membrane biogenesis may be an important determinant of this effect. Infection of some developing neuronal populations results in apoptosis, whereas infection of mature neurons results in persistent infection. An active type-I interferon system prevents virus spread in extraneural tissues. An initial high-titer plasma viremia is controlled by immunoglobulin M (IgM) antibodies. Virus enters the brain across cerebral endothelial cells and initiates scattered foci of perivascular infection. The blood-brain barrier is disrupted. Neurons and oligodendrocytes are the cell types most frequently infected. Infectivity in the brain can be eliminated by IgG antibodies, though an active T-cell response is required for virus elimination. Lesions of inflammatory demyelination require the presence of CD8(+) T lymphocytes and probably result from destruction by these cells of virally infected oligodendrocytes.
Journal of NeuroVirology, 8(suppl. 2): 66–74, 2002
c
°2002 Taylor & Francis ISSN 1355–0284/02 $12.00+.00
DOI: 10.1080/135502802901068000
Pathogenesis of Semliki Forest virus encephalitis
John K Fazakerley
Centre for Infectious Diseases, University of Edinburgh, Summerhall, Edinburgh, United Kingdom
This article provides a review of the pathogenesis of Semliki Forest virus (SFV)
encephalitis. In mice, outcome of infection varies according to age of the mouse
and strain of the virus and can include acute encephalitis, subacute demyeli-
nating meningoencephalomyelitis, and persistent subclinical central nervous
system (CNS) infection. All strains of virus are virulent in mice infected <12
days of age. The L10 strain is also virulent in mice >14 days age, whereas the
A7(74) strain is avirulent. The genetic difference between these strains maps
to the nsp3 gene. For A7(74) virus, age-related virulence correlates with ability
of CNS neurons to replicate virus and undergo apoptotic cell death. Immature
developing neurons support complete virus replication but as neuronal pop-
ulations and circuits mature in the postnatal brain, virus infection becomes
progressively restricted and nonproductive. This restricted replication can be
overcome by gold I compounds, which may function by inducing neuronal
dedifferentiation to a state permissive for virus replication. Biochemical path-
ways associated with membrane biogenesis may be an important determinant
of this effect. Infection of some developing neuronal populations results in
apoptosis, whereas infection of mature neurons results in persistent infection.
An active type-I interferon system prevents virus spread in extraneural tissues.
An initial high-titer plasma viremia is controlled by immunoglobulin M (IgM)
antibodies. Virus enters the brain across cerebral endothelial cells and initiates
scattered foci of perivascular infection. The blood-brain barrier is disrupted.
Neurons and oligodendrocytes are the cell types most frequently infected. In-
fectivity in the brain can be eliminated by IgG antibodies, though an active
T-cell response is required for virus elimination. Lesions of inflammatory de-
myelination require the presence of CD8+T lymphocytes and probably result
from destruction by these cells of virally infected oligodendrocytes. Journal of
NeuroVirology (2002) 8(suppl. 2), 66–74.
Keywords: alphavirus; demyelination; encephalitis; Semliki Forest virus; SFV
Strains of SFV
Semliki Forest virus (SFV), an alphavirus of the
Togaviridae family, is naturally found in sub-Saharan
Africa where it is spread principally by Aedes
africanus and Aedes aegypti mosquitoes. SFV is
most closely related to Chikungunya, Getah, and Ma-
yaro alphaviruses. SFV was first isolated in 1942
from a squash of 130 female, Aedes abnormalis
mosquitoes captured in the Semliki Forest in Uganda
(Smithburn and Haddow, 1944). The natural ver-
tebrate host remains unknown but infections have
been documented in horses, monkeys, and man. The
Address correspondence to John K Fazakerley, Centre for Infec-
tious Diseases, University of Edinburgh, Summerhall, Edinburgh,
EH9 1QH, United Kingdom. E-mail: John.Fazakerley@ed.ac.uk
Received 19 August 2002; accepted 29 August 2002.
virus was associated with an epidemic of equine
encephalitis in Senegal (Robin et al, 1974). In a
1987 survey in the Central African Republic, SFV
was isolated from 22 patients with fever, severe per-
sistent headaches, myalgia, and arthralgia (Mathiot
et al, 1990). Virus was also isolated from locally
caught Aedes aegypti mosquitoes. There is one re-
ported case of the death of a scientist working with
the Osterrieth strain of the virus (Willems et al,
1979). The patient had neurological disease and at
postmortem examination, a typical viral meningoen-
cephalomyelitis was observed. SFV was isolated from
both the cerebrospinal fluid and the brain. It is known
that the scientist was working with virus super-
natant from baby hamster kidney (BHK) cells but
the route and dose of infection and the genotype
of this virus are unknown. Seroconversion in labo-
ratory workers is common (Willems et al, 1979). SFV
SFV encephalitis
JK Fazakerley 67
is relatively stable in aerosols of tissue homogenates
(Benbough, 1969), although aerosols of purified virus
are rapidly inactivated (de Jong et al, 1976).
Various strains of virus have been used in lab-
oratories around the world and have been shown
experimentally to infect voles, mice, guinea pigs,
rabbits, and rats (Seamer et al, 1967a, 1967b; Bradish
et al, 1971). Different strains have been designated as
virulent or avirulent according to their virulence in
adult mice. The L10, V13, and Osterrieth strains and
the strain designated prototype are virulent (Bradish
et al, 1971; Glasgow et al, 1991). The A8, A7, A7(74),
and MRS MP 192/7 strains are avirulent (Bradish
et al, 1971; Henderson et al, 1970). Virulence in
mice is age-related because all strains are virulent
in neonatal and suckling mice. The L10 strain was
derived from the original isolate (Smithburn and
Haddow, 1944) by eight intracerebral passages
through adult and two through baby mouse brains
(Bradish et al, 1971). A7(74) was derived from the
AR2066 strain by seven passages through neonatal
mouse brain and colony selection on chick em-
bryo fibroblasts. AR2066 was isolated from Aedes
argenteopunctatus mosquitoes in Namancurra,
Mozambique (McIntosh et al, 1961). A panel of 15
E1 or E2 monoclonal antibodies raised to MRS MP
192/7 can distinguish between some of the virus
strains (Boere et al, 1984, 1986). Several series of
SFV mutants, including temperature-sensitive (ts)
mutants, have been described (Tan et al, 1969;
Keranen and Kaariainen, 1974; Hearne et al, 1987;
Barrett et al, 1980).
A number of strains including prototype, which
is related to L10, A7, and A7(74), have been cloned
and sequenced (Garoff et al, 1980; Takkinen, 1986;
Glasgow et al, 1994; Santagati et al, 1995, 1998;
Tarbatt et al, 1997; Tuittila et al, 2000). The molecu-
lar clone of the prototype strain, known as SFV4, has
been engineered to form a much used viral expres-
sion and vaccine system (Liljestrom and Garoff, 1991;
Tubulekas et al, 1997). Isolated SFV RNA is infec-
tious (Friedman et al, 1966). As with many positive-
stranded RNA viruses, studies of site-directed
mutants have proved to be a powerful approach to
mapping areas of the genome responsible for individ-
ual phenotypic characteristics. Among the SFV4, A7,
and A7(74) strains, there are multiple changes scat-
tered throughout the genome (Tuittila et al, 2000).
The difference in virulence between SFV4 and A7 or
A7(74) appears to be polygenic and changes in the
E2 gene, the 50untranslated region, and the carboxy-
terminal end of the nsP3 gene have been shown to
be determinants of virulence between these particu-
lar strains (Santagati et al, 1995, 1998; Tarbatt et al,
1997; Tuittila et al, 2000).
SFV replication
SFV is a relatively simple virus, it has an approx-
imately 12-kb genome of single-stranded positive-
sense RNA. Replication is via a negative-sense inter-
mediate that gives rise to full-length genomic RNA
and a subgenomic message, which represents the 30
third of the genomic RNA and contains the genes
for the viral structural proteins. Both the genomic
and subgenomic RNAs are capped and polyadeny-
lated. From 50to 30along the genomic RNA, there
are four nonstructural replicase proteins, designated
nsP1 to nsP4, followed by the capsid protein C and
the envelope glycoproteins E2, E3, and E1. Many
of the functions of the replicase proteins have been
elucidated. NsP1 is involved in capping the viral
RNAs and has methyl and guanylyl transferase ac-
tivities; it participates in initiation of negative-strand
RNA synthesis and targets the replicase complex to
membranes (Ahola and Kaariainen, 1995; Laakkonen
et al, 1994; Ahola et al, 1997; Peranen and Kaariainen,
1991). The large nsP2 protein has single-stranded
RNA-stimulated ATPase and GTPase activities, RNA
triphosphatase activity, RNA helicase activity, reg-
ulates synthesis of the 26S subgenomic RNA, is
involved in the cessation of negative-strand syn-
thesis, and contains a papain-like proteinase do-
main responsible for processing the nonstructural
polyprotein (Rikkonen et al, 1994; Vasiljeva et al,
2000; Gomez de Cedron et al, 1999; Suopanki et al,
1998; Merits et al, 2001). In infected cells, about half
the nsP2 synthesized is translocated to the nucleus
(Kujala et al, 1997; Peranen et al, 1990; Rikkonen
et al, 1992). The protein contains a nuclear-targeting
sequence that is present in all three cloned and se-
quenced strains of the virus. Disruption of this se-
quence has minimal effect on virus replication in
continuously cultured cells but severely attenuates
replication in mouse brain (Fazakerley et al, 2002).
The role of nsP3 is less well known. It is a phos-
phoprotein, with a role in the regulation of RNA
synthesis (Peranen et al, 1988; Wang et al, 1991).
NsP4 is the catalytic subunit of the RNA polymerase
(Keranen and Kaariainen, 1979). The polymerase
complex is associated with plasma membrane and
endo/lysosomes by specific interactions of nsP1 and
nsP2. SFV virions are enveloped and on budding
have a spike composed of three E1-E2 heterodimers.
Within the lipid envelope, the RNA is associated
with the capsid protein, which is in contact with the
C-terminal domain of the E2 protein. Budding of viri-
ons is driven by spike capsid interactions and does
not require the presence of full-length genomic RNA
(Suomalainen et al, 1992).
The receptor remains undetermined but virus en-
try and fusion are via the endosomal system and have
been much studied (Helenius et al, 1986). Fusion
of the viral and endosomal membranes is a func-
tion of the E1 glycoprotein and is dependent upon
the presence of cholesterol and reorganization of the
envelope spike at acid pH (Omar and Koblet, 1988;
Helenius et al, 1980; Kielian and Helenius, 1984;
Wahlberg and Garoff, 1992). Upon fusion, the capsid
is liberated into the cytoplasm. RNA replication is
SFV encephalitis
68 JK Fazakerley
associated with smooth membrane structures termed
cytopathic vacuoles (CPVs), types I and II (Grimley
et al, 1968; Grimley and Friedman, 1970; Kujala et al,
2001). These structures can be separated from other
subcellular structures on sucrose gradients, and bio-
chemical markers indicate they are derived from late
endosomes and lysosomes (Friedman et al, 1972;
Froshauer et al, 1988; Mehta et al, 1990; Kujala et al,
2001). Confocal microscopy and electronmicroscopy
(EM) studies reveal that the surfaces of CPV-I are cov-
ered by small invaginations termed spherules, which
contain the viral nsP’s and are the likely sites of vi-
ral RNA replication (Grimley et al, 1968; Kujala et al,
2001). In vitro studies of cells in continuous culture
have generally observed virus budding at the plasma
membrane. However, EM observations on brain sec-
tions from SFV-infected neonatal mice indicate that
in neurons, virions frequently bud into the lumen of
type-II vacuoles, which then fuse with the cell mem-
brane to release mature virions (Pathak et al, 1976).
In brain sections, clumps of electron-dense material
are observed throughout the cytoplasm early in in-
fection. These contain capsid protein and are proba-
bly RNA capsid complexes. They migrate to under-
lie membranes, including the plasma membrane at
sites of virus budding (Pathak et al, 1976; Pathak and
Webb, 1978, 1983a, 1988).
Neuroinvasion, CNS tropism,
and age-related virulence
Following intraperitoneal (IP) inoculation into adult
mice, SFV replicates in muscles, including skele-
tal, smooth, and cardiac muscle (Pusztai et al, 1971;
Amor et al, 1996). A high-titer plasma viremia is de-
tectable within 24 h. This usually falls after 48 h
and in the blood, virus is generally undetectable
by infectivity assay by 4 days (Pusztai et al, 1971;
Fazakerley et al, 1993). All strains that have been
studied (L10, A7, A7(74)) are neuroinvasive. The
much studied A7(74) and L10 strains have been
shown to replicate in and probably enter the brain
across cerebral endothelial cells (Pathak and Webb,
1974; SoiluHanninen et al, 1994), initiating perivas-
cular foci of infection (Fazakerley et al, 1993). It
is likely that other strains also enter the central
nervous system (CNS) by this route. Following in-
tranasal inoculation, infection of the brain via olfac-
tory nerves can also occur (Kaluza et al, 1987; Oliver
and Fazakerley, 1998). Once in the CNS, all strains
of the virus infect neurons and oligodendrocytes,
but not astrocytes, and only very rarely meningeal,
ependymal, or choroid plexus cells (Balluz et al,
1993; Pathak and Webb, 1983b).
From the original perivascular foci, the virulent
L10 strain spreads rapidly around the brain, pro-
ducing a fatal panencephalitis (Figure 1) (Fazakerley
et al, 1993). This occurs irrespective of the age of
the mice and so rapidly that the immune response
Figure 1 Autoradiographic images showing distribution of SFV
RNA by in situ hybridization in brains of 3- to 4-week-old BALB/c
mice at 2, 3, 4, and 5 days post inoculation (IP). Mice were inocu-
lated with 5000 PFU of SFV A7(74) or L10. One group of mice also
received 10 mg of sodium aurothiomalate (GSTM) IP 3 to 4 hours
prior to virus infection. Infection with SFV A7(74) results in dis-
crete scattered foci of perivascular infection first apparent here at
4 days post infection. These foci do not enlarge with time even in
SCID mice (not shown). Prior treatment with GSTM results in rapid
spread of A7(74) infection to produce a panencephalitis similar to
that seen with the L10 strain of the virus. Modified from Fazakerley
et al (1993) and Scallan et al (1999), with permission.
has no time to intervene. In neonatal and suckling
mice, many neurons are destroyed. In adult mice, at
the time of death, most infected neurons have a rel-
atively normal morphology and the exact cause of
death is unclear. Destruction of a vital group of neu-
rons, toxic levels of cytokines, neuronal dysfunction,
or extraneural pathology are possibilities. In contrast,
the dynamics of A7(74) infection of the CNS varies
according to age of the mice. In neonatal mice, A7(74)
virus spreads rapidly around the brain and infection
is, as with the L10 strain, rapidly fatal. As the mice
age, the A7(74) virus is less able to spread in the CNS,
and there is a sharp age-related virulence: mice in-
fected at 12 days of age or less die, whereas those
infected at 14 days of age or more survive (Fleming,
1977; Oliver et al, 1997; Oliver and Fazakerley,
1998). In the mature adult mouse CNS, A7(74) virus
has a restricted replication and remains confined to
perivascular foci (Figure 1) (Pathak and Webb, 1978;
Fazakerley et al, 1993; Oliver et al, 1997). This age-
related virulence is not a function of the maturity
of specific immune responses, as this virus also re-
mains confined to small, predominantly perivascu-
lar foci in athymic nu/nu mice or mice with severe
combined immunodeficiency (SCID) (Fazakerley and
Webb, 1987b; Fazakerley et al, 1993; Amor et al,
1996).
The A7(74) age-related virulence is a function of
the maturity of CNS cells. In the first two weeks
after birth, in the mouse a number of major matura-
tional events are ongoing in the CNS, these include
SFV encephalitis
JK Fazakerley 69
Figure 2 Distribution of SFV A7(74) RNA (brown staining) by in
situ hybridization at 40 h post infection in the brains of mice in-
oculated (IP) at (A) P4, (B) P8, and (C) P12. In (A), virus can be
observed to infect columns of interconnected neurons. These are
broader in the frontal and fore- and hind-limb areas and narrower
in the occipital cortex, consistent with the known banding patterns
of these functional motor and sensory neuronal groupings (Purves
et al, 1992). At this age (P4), there is widespread axonogenesis
and synaptogenesis occurring in these interconnecting columns.
This connectivity is progressively completed between P4 and P10
and is associated with a progressive curtailment in the number of
columns infected by the virus (Band C). In the animals inoculated
at P12 (C), only cells in deep layer VI of the cortex, the cingulated
gyrus, and the corpus callosum are infected (arrowheads). Modi-
fied from Oliver et al (1997), with permission.
axonogenesis, synaptogenesis, gliogenesis, and
myelination (reviewed by Fazakerley, 2001). The
A7(74) strain of SFV is able to replicate productively
in highly active maturing neurons undergoing
axonogenesis and synaptogenesis (Figure 2). As
these processes are completed, a number of major
changes occur that restrict virus replication (Pathak
and Webb, 1978; Fazakerley et al, 1993; Oliver
et al, 1997; Oliver and Fazakerley, 1998). Restriction
may be linked to shutdown of metabolic processes
required by the virus, for example, production and
transport of smooth membrane vesicles (Oliver
et al, 1997) or to up-regulation of inhibitory pro-
cesses, for example, antiapoptotic genes (Scallan
et al, 1997). Of relevance here is the changed
course of events following treatment of infected
mice with gold compounds. Treatment of adult
mice with sodium aurothiomalate and other gold I
compounds results in conversion of this normally
avirulent infection to a virulent infection (Allner
et al, 1974; Bradish et al, 1975; Mehta and Webb,
1982; Gates et al, 1984; Scallan and Fazakerley,
1999). The gold salt is transported to the CNS where
it induces smooth membrane production in neurons
Figure 3 A discrete lesion of inflammatory demyelination in a
white matter tract in the cerebellum. SFV A7(74), 18 days post
infection (IP), 5-week-old BALB/c mouse.
(Pathak and Webb, 1983a). In mice treated with gold
compounds, A7(74) virus replication is no longer
restricted in neurons but is productive and spreads
rapidly around the brain (Figure 1), resulting in a
panencephalitis characteristic of that observed with
the virulent L10 virus (Pathak and Webb, 1983a;
Scallan and Fazakerley, 1999).
Infection of neurons in the neonate can result in
death of specific neuronal populations by apopto-
sis (Allsopp et al, 1998), whereas infection of ma-
ture (adult) neurons is generally nondestructive and
can result in virus persistence (Fazakerley et al, 1993;
Amor et al, 1996). In the normal course of postnatal
development of the CNS, as immature neurons of the
neonatal mouse brain make their connections, they
become more resistant to apoptotic death, whereas
those neurons that do not form correct connections
die by this process (reviewed by Fazakerley, 2001).
In culture, SFV A7(74) and L10 both kill continu-
ously growing cell lines by apoptosis, but expres-
sion of antiapoptotic genes prolongs survival (Scallan
et al, 1997). SFV L10 and A7(74) also kill primary cul-
tures of mouse embryonic sensory neurons by apop-
tosis (Allsopp et al, 1998), but A7(74) does not de-
stroy primary cultures of neurons from adult mice
whereas L10 does. For renewable cell populations,
apoptotic cell death upon infection can be consid-
ered to be a highly successfully, altruistic antiviral
defense mechanism. For mature, nonrenewable, vital
cell populations such as neurons this would not be
the case, and selective pressures may have dictated
that mature neurons are a specialized case and do not
readily undergo apoptosis (Allsopp and Fazakerley,
2000; Fazakerley and Allsopp, 2001).
Taken together, the foregoing studies suggest the
following course of events: In the developing mouse
brain, both strains of SFV, L10 and A7(74), repli-
cate efficiently in immature neurons. It is likely that
SFV encephalitis
70 JK Fazakerley
these cells contain suitable membranes and biochem-
ical pathways for complete virus replication. Given
their susceptibility to apoptosis, some infected pop-
ulations of neurons undergo apoptotic cell death. As
neuronal circuits and cells mature in the first 2 post-
natal weeks, neuronal physiology changes such that
A7(74) replication becomes restricted with viral RNA
replication and protein synthesis but no, or minimal,
virus budding and spread of infection. These more
mature neurons are also less susceptible to apop-
tosis and the result, in the absence of specific im-
munity (SCID mice), is a persistent nondestructive
neuronal infection. Given that gold compounds re-
lieve the restriction on viral replication and induce
smooth membrane synthesis, and given the require-
ment for suitable membranes for virus replication
(CPVs, spherules) and budding, it seems likely that
a change in membrane synthesis, or associated bio-
chemical pathways, may be the age-related change
responsible for switching the outcome of A7(74) in-
fection from productive and destructive to restricted
and persistent. Given that replication of the L10 strain
is not restricted in mature neurons, the genetic locus
responsible for this phenotypic difference between
the L10 and A7(74) strains may be informative as to
the mechanisms involved. This difference appears to
reside in the carboxy-terminus domain of the nsP3
protein (Tuittila et al, 2000), but the function of this
remains unknown. It also remains unknown how SFV
triggers the apoptotic response in cells, including
neurons, and what changes occur on neuronal matu-
ration, at least in some populations of neurons, that
reduce susceptibility to apoptosis.
Apart from CNS disease, another interesting prop-
erty of some strains, for example, A7 but not L10, is
their ability to cross the mouse placenta and induce
abortions (Atkins et al, 1982; Milner and Marshall,
1984). The A7 ts22 mutant is teratogenic, inducing
skeletal, skin, and neural tube defects in develop-
ing fetuses (Mabruk et al, 1988, 1989). This is of
interest given the teratogenic effects of the related
rubella virus. As with most RNA viruses, SFV read-
ily generates defective interfering particles on high-
multiplicity passage in culture (Bruton and Kennedy,
1976; Barrett et al, 1984). In a series of experiments,
defective interfering (DI) particles administered ei-
ther intraperitoneally or intranasally at the same time
as virulent virus have been shown to inhibit virus
replication, delay the time of death, completely pro-
tect animals, or convert the infection from lethal to
persistent (Dimmock and Kennedy, 1978; Barrett and
Dimmock, 1984; Atkinson et al, 1986).
Role of immune response
Infection of SCID mice with SFV A7(74) results in
persistent CNS infection without any apparent neu-
ronal loss (Amor et al, 1996). In contrast, in im-
munocompetent mice, virus is eliminated from the
CNS. This comparison demonstrates two important
points, firstly there is no, or at most only rare, direct
A7(74) virus-induced death of infected cells in the
adult mouse brain, and secondly, specific immune
responses are required to eliminate this infection. In
immunocompetent mice, infectious virus cannot be
detected by plaque assay after day 8, by the more sen-
sitive ICLD50 after day 11, and by in situ hybridiza-
tion after day 14 (Suckling et al, 1978; Jagelman et al,
1978; Fazakerley et al, 1993). Reverse transcriptase–
polymerase chain reaction (RT-PCR) indicates that vi-
ral RNA can be detected at later time points (Donnelly
et al, 1997). After the clearance of detectable infectiv-
ity, from day 14 onwards, immunocompetent mice
develop lesions of inflammatory demyelination in
the white matter. These lesions are apparent through-
out the CNS (Suckling et al, 1978; Kelly et al, 1982).
No demyelinating lesions are observed in SCID mice
(Amor et al, 1996), despite CNS virus persistence.
Specific immune responses therefore have a role both
in clearing this infection and in the generation of
lesions of demyelination.
The roles of different effector functions of the im-
mune response have been determined over the years
by studying naturally arising mutant mice, genet-
ically engineered mice, mice treated with general
or selective immunosuppressive regimens, and by
adoptive transfer experiments. Following IP inocu-
lation of virus, plasma levels of type-I interferons
parallel those of the viremia (Bradish et al, 1975). In
mice with a genetic deletion of functional type-I inter-
feron receptors (Muller et al, 1994), IP infection with
SFV is fatal within 48 h, with widespread infection
of many organs and tissues (JKF, unpublished data).
Type-I interferons are therefore crucial in protecting
against widespread SFV infection in numerous tis-
sues and their absence leads to widespread infection
and death. The role of other cytokines has not been
investigated in detail, but interleukin (IL)-1α, IL-1β,
IL-3, IL-6, IL-10, and tumor necrosis factor (TNF)α
can be readily detected in the CNS following SFV
A7(74) infection (Morris et al, 1997).
Following IP inoculation of SFV, A7(74) BALB/c
mice produce a rapid and neutralizing serum im-
munoglobulin M (IgM) antibody response followed
rapidly by an IgG2a response and a much slower IgG1
response (Fazakerley et al, 1993). During the nor-
mal course of infection in immunocompetent mice,
the blood-brain barrier is leaky from days 4 to 10,
allowing passage of immunoglobulins (Parsons and
Webb, 1982a; SoiluHanninen et al, 1994). Antibody-
producing plasma cells can be detected in the brain
and intrathecal antibodies can be detected for months
post infection (Parsons and Webb, 1984). SCID mice,
which have no serum antiviral antibodies, have both
a persistent viremia and a persistent CNS infection
(Amor et al, 1996), whereas nu/nu mice control the
viremia but have a persistent CNS infection. The
nu/nu mice produce serum antiviral IgM but not IgG
(Suckling et al, 1982), suggesting that this may be
SFV encephalitis
JK Fazakerley 71
controlling the viremia but unable to eliminate the
CNS infection. That this is the case is confirmed by
resolution of the persistent viremia but not the CNS
infection on transfer of day 7 nu/nu mouse serum to
infected SCID mice (Amor et al, 1996). In contrast,
transfer of high-titer IgG sera from immunocompe-
tent BALB/c mice to SCID mice was able to abolish
both the viremia and titers of infectious virus from the
CNS (Amor et al, 1996). Whether this was complete
eradication of all viral sequences was not checked
by more sensitive techniques such as RT-PCR. Anti-
bodies are highly effective in protecting mice from
challenge with a lethal dose of SFV. A number of
B-cell epitopes have been mapped on the viral glyco-
proteins (Boere et al, 1984; Snijders et al, 1991) and
even a single monoclonal antibody directed to the
E2 envelope glycoprotein has been shown to protect
mice against a virulent infection (Boere et al, 1983).
Furthermore, a non-neutralizing anti-E2 monoclonal
has been shown to be able to clear A7(74) virus from
persistently infected SCID mouse brains (Amor et al,
1996). Combinations of linear B-cell epitopes and
B- and T-cell epitopes have been tested as potential
vaccine candidates (Snijders et al, 1991).
Following IP infection of BALB/c mice with SFV
A7(74), perivascular and infiltrating mononuclear
cells are apparent in the CNS from 3 days onwards
(Parsons and Webb, 1982b). Histopathological stud-
ies show that the areas of inflammatory infiltrates
correspond to the areas of infection (Subak-Sharpe
et al, 1993). Perivascular cuffs are a prominent fea-
ture of the neuropathology and are maximal be-
tween 7 and 10 days. Cerebral endothelial cells up-
regulate adhesion molecule expression after infection
(SoiluHanninen et al, 1997; Morris et al, 1997), and
some of these, ependymal cells, meningeal cells, most
infiltrating mononuclear cells and some parenchy-
mal cells with a dendritic morphology, are major
histocompatibility complex (MHC)-I+(Morris et al,
1997). The parenchymal MHC-I+cells form a network
References
Ahola T, Kaariainen L (1995). Reaction in alphavirus
mRNA capping: formation of a covalent complex of non-
structural protein nsP1 with 7-methyl-GMP. Proc Natl
Acad Sci USA 92: 507–511.
Ahola T, Laakkonen P, Vihinen H, Kaariainen L (1997).
Critical residues of Semliki Forest virus RNA cap-
ping enzyme involved in methyltransferase and
guanylyltransferase-like activities. J Virol 71: 392–397.
Allner K, Bradish CJ, Fitzgeorge R, Nathanson N (1974).
Modifications by sodium aurothiomalate of the expres-
sion of virulence in mice by defined strains of Semliki
Forest virus. J Gen Virol 24: 221–228.
Allsopp TE, Fazakerley JK (2000). Altruistic cell suicide
and the specialized case of the virus-infected nervous
system. Trends Neurosci 23: 284–290.
Allsopp TE, Scallan MF, Williams A, Fazakerley JK
(1998). Virus infection induces neuronal apoptosis:
surrounding the foci of infection in which the level
of MHC-I staining decreases with distance from the
infected cells. MHC-II+cells, probably pericytes, can
be observed adjacent to blood vessels in areas of in-
fection, and MHC-II+astrocytes and microglial cells
are present in the areas of infection and inflamma-
tion (Morris et al, 1997). A few of the infiltrating in-
flammatory cells are MHC-II+, the majority are not.
CD45/B220+B cells are also present in the inflamma-
tory lesions, including those showing demyelination.
Mice infected with SFV have good delayed-type
hypersensitivity responses to the virus (Kraaijeveld
et al, 1983), and T-cell epitopes for some hap-
lotypes have been mapped (Snijders et al, 1991,
1992a, 1992b). Adoptive-transfer studies in SCID and
nu/nu mice and studies in mice immunosuppressed
with total body irradiation, cyclophosphamide, cy-
closporine, or cycloleucine all demonstrate that
T-cell responses are pathogenic and are required to
generate the lesions of demyelination (Fazakerley
and Webb, 1987a, 1987c; Amor and Webb, 1987;
Amor et al, 1996). Depletion of CD8+cells with a
monoclonal antibody greatly reduces demyelination,
whereas depletion of CD4+cells reduces the extent
of inflammation but not of demyelination (Subak-
Sharpe et al, 1993). Given the requirement for CD8+
cells for demyelination and the tropism of A7(74)
virus for oligodendrocytes, the most likely mecha-
nism of demyelination would seem to be a CD8+
T cell–mediated destruction of virally infected oligo-
dendrocytes, though this remains to be proven. Mice
infected with SFV A7(74) generate CNS antigen-
specific autoantibodies, are more sensitive to the
induction of experimental allergic encephalomyeli-
tis, and have T cells that cross-react between vi-
ral and CNS epitopes (Amor and Webb, 1988;
Mokhtarian and Swoveland, 1987), though any role
for autoimmune responses in the generation of
SFV A7(74)-induced demyelinating lesions remains
unclear.
a comparison with trophic factor withdrawal. Cell Death
Differ 5: 50–59.
Amor S, Scallan MF, Morris MM, Dyson H, Fazakerley
JK (1996). Role of immune responses in protection and
pathogenesis during Semliki Forest virus encephalitis.
J Gen Virol 77: 281–291.
Amor S, Webb HE (1987). The effect of cycloleucine on
SFV A7(74) infection in mice. Br J Exp Pathol 68: 225–
235.
Amor S, Webb HE (1988). CNS pathogenesis following a
dual viral infection with Semliki Forest (alphavirus) and
Langat (flavivirus). Br J Exp Pathol 69: 197–208.
Atkins GJ, Carter J, Sheahan BJ (1982). Effect of alphavirus
infection on mouse embryos. Infect Immun 38: 1285–
1290.
Atkinson T, Barrett ADT, MacKenzie A, Dimmock NJ
(1986). Persistence of virulent Semliki Forest virus in
SFV encephalitis
72 JK Fazakerley
mouse-brain following co-inoculation with defective in-
terfering particles. J Gen Virol 67: 1189–1194.
Balluz IM, Glasgow GM, Killen HM, Mabruk MJ, Sheahan
BJ, Atkins GJ (1993). Virulent and avirulent strains of
Semliki Forest virus show similar cell tropism for the
murine central nervous system but differ in the severity
and rate of induction of cytolytic damage. Neuropathol
Appl Neurobiol 19: 233–239.
Barrett ADT, Crouch CF, Dimmock NJ (1984). Defective in-
terfering Semliki Forest virus populations are biolog-
ically and physically heterogeneous. J Gen Virol 65:
1273–1283.
Barrett ADT, Dimmock NJ (1984). Modulation of a systemic
Semliki Forest virus infection in mice by defective in-
terfering virus. J Gen Virol 65: 1827–1831.
Barrett PN, Sheahan BJ, Atkins GJ (1980). Isolation and pre-
liminary characterization of Semliki Forest virus mu-
tants with altered virulence. J Gen Virol 49: 141–147.
Benbough JE (1969). The effect of relative humidity on the
survival of airborne Semliki Forest virus. J Gen Virol 4:
473–477.
Boere WA, Benaissa-Trouw BJ, Harmsen M, Kraaijeveld
CA, Snippe H (1983). Neutralizing and non-neutralizing
monoclonal antibodies to the E2 glycoprotein of Semliki
Forest virus can protect mice from lethal encephalitis.
J Gen Virol 64: 1405–1408.
Boere WA, Harmsen M, Kraaijeveld CA, Snippe H (1986).
Antigenic differences between virulent and avirulent
strains of Semliki Forest viruses detected with mono-
clonal antibodies. Brief report. Arch Virol 88: 105–112.
Boere WA, Harmsen T, Vinje J, Benaissa-Trouw BJ,
Kraaijeveld CA, Snippe H (1984). Identification of dis-
tinct antigenic determinants on Semliki Forest virus by
using monoclonal antibodies with different antiviral ac-
tivities. J Virol 52: 575–582.
Bradish CJ, Allner K, Fitzgeorge R (1975). Immunomodifi-
cation and the expression of virulence in mice by de-
fined strains of Semliki Forest virus: the effects of cy-
clophosphamide. J Gen Virol 28: 225–237.
Bradish CJ, Allner K, Maber HB (1971). The virulence of
original and derived strains of Semliki Forest virus for
mice, guinea-pigs and rabbits. J Gen Virol 12: 141–160.
Bruton CJ, Kennedy SIT (1976). Defective interfering par-
ticles of Semliki Forest virus: structural differences be-
tween standard virus and defective-interfering particles.
J Gen Virol 31: 383–395.
de Jong JC, Harmsen M, Plantinga AD, Trouwbrost T (1976).
Inactivation of Semliki Forest virus in aerosols. Appl
Environ Microbiol 32: 315–319.
Dimmock NJ, Kennedy SI (1978). Prevention of death in
Semliki Forest virus-infected mice by administration of
defective-interfering Semliki Forest virus. J Gen Virol
39: 231–242.
Donnelly SM, Sheahan BJ, Atkins GJ (1997). Long-term ef-
fects of Semliki Forest virus infection in the mouse cen-
tral nervous system. Neuropathol Appl Neurobiol 23:
235–241.
Fazakerley JK (2001). Neurovirology and developmental
neurobiology. Adv Virus Res 56: 73–124.
Fazakerley JK, Allsopp TE (2001). Programmed cell death
in virus infections of the nervous system. Curr Top Mi-
crobiol Immunol 253: 95–119.
Fazakerley JK, Boyd A, Mikkola ML, Kaariainen L (2002).
A single amino acid change in the nuclear localization
sequence of the nsP2 protein affects the neurovirulence
of Semliki Forest virus. J Virol 76: 392–396.
Fazakerley JK, Pathak S, Scallan M, Amor S, Dyson H
(1993). Replication of the A7(74) strain of Semliki Forest
virus is restricted in neurons. Virology 195: 627–637.
Fazakerley JK, Webb HE (1987a). Cyclosporine enhances
virally induced T-cell-mediated demyelination. The ef-
fect of cyclosporine on a demyelinating virus infection.
J Neurol Sci 78: 35–50.
Fazakerley JK, Webb HE (1987b). Semliki Forest virus-
induced, immune-mediated demyelination: adoptive
transfer studies and viral persistence in nude mice.
J Gen Virol 68: 377–385.
Fazakerley JK, Webb HE (1987c). Semliki Forest virus in-
duced, immune mediated demyelination: the effect of
irradiation. Br J Exp Pathol 68: 101–113.
Fleming P (1977). Age-dependent and strain-related differ-
ences of virulence of Semliki Forest virus in mice. J Gen
Virol 37: 93–105.
Friedman RM, Levin JG, Grimley PM, Berezesky IK (1972).
Membrane-associated replication complex in arbovirus
infection. J Virol 10: 504–515.
Friedman RM, Levy HB, Carter WB (1966). Replication of
Semliki Forest virus: three forms of viral RNA produced
during infection. Proc Natl Acad Sci USA 56: 440–446.
Froshauer S, Kartenbeck J, Helenius A (1988). Alphavirus
RNA replicase is located on the cytoplasmic surface of
endosomes and lysosomes. J Cell Biol 107: 2075–2086.
Garoff H, Frischauf A, Simons K, Lehrach H, Delius H
(1980). Nucleotide sequence of cDNA coding for Semliki
Forest virus membrane glycoproteins. Nature 288: 236–
241.
Gates MC, Sheahan BJ, Atkins GJ (1984). The pathogenicity
of the M9 mutant of Semliki Forest virus in immune-
compromised mice. J Gen Virol 65(Pt 1): 73–80.
Glasgow GM, Killen HM, Liljestrom P, Sheahan BJ, Atkins
GJ (1994). A single amino acid change in the E2 spike
protein of a virulent strain of Semliki Forest virus atten-
uates pathogenicity. J Gen Virol 75: 663–668.
Glasgow GM, Sheahan BJ, Atkins GJ, Wahlberg JM,
Salminen A, Liljestrom P (1991). Two mutations in the
envelope glycoprotein E2 of Semliki Forest virus affect-
ing the maturation and entry patterns of the virus alter
pathogenicity for mice. Virology 185: 741–748.
Gomez de Cedron M, Ehsani N, Mikkola M, Garcia JA,
Kaariainen L (1999). RNA helicase activity of Semliki
Forest virus replicase protein NSP2. FEBS Lett 448: 19–
22.
Grimley PM, Berezesky IK, Friedman RM (1968). Cytoplas-
mic structures associated with an Arbovirus infection:
loci of viral ribonucleic acid synthesis. J Virol 2: 1326–
1338.
Grimley PM, Friedman RM (1970). Development of Semliki
Forest virus in mouse brain—an electron microscopic
study. Exp Mol Pathol 12: 1–13.
Hearne AM, O’Sullivan MA, Atkins GJ (1987). Isolation
and preliminary characterization of Semliki Forest virus
mutants with altered pathogenicity for mouse embryos.
J Gen Virol 68: 107–113.
Helenius A, Doms R, White J, Kielian M (1986). Entry mech-
anisms of influenza and Semliki Forest viruses. J Cell
Biochem 30: 49–49.
Helenius A, Marsh M, White J (1980). The entry of viruses
into animal cells. Trends Biochem 5: 104–106.
Henderson BE, Metselaar D, Kirya GB, Timms GL (1970).
Investigations into yellow fever virus and other ar-
boviruses in the northern regions of Kenya. Bull World
Health Org 42: 787–795.
SFV encephalitis
JK Fazakerley 73
Jagelman S, Suckling AJ, Webb HE, Bowen ETW (1978). The
pathogenesis of avirulent Semliki Forest virus infections
in athymic nude mice. J Gen Virol 41: 599–607.
Kaluza G, Lell G, Reinacher M, Stitz L, Willems WR (1987).
Neurogenic spread of Semliki Forest virus in mice. Arch
Virol 93: 97–110.
Kelly WR, Blakemore WF, Jagelman S, Webb HE (1982). De-
myelination induced in mice by avirulent Semliki Forest
virus. II. An ultrastructural study of focal demyelination
in the brain. Neuropathol Appl Neurobiol 8: 43–53.
Keranen S, Kaariainen L (1974). Isolation and basic
characterization of temperature-sensitive mutants from
Semliki Forest virus. Acta Pathol Microbiol Scand [B]
Microbiol Immunol 82: 810–820.
Keranen S, Kaariainen L (1979). Functional defects of RNA-
negative temperature-sensitive mutants of Sindbis and
Semliki Forest viruses. J Virol 32: 19–29.
Kielian MC, Helenius A (1984). Role of cholesterol in fusion
of Semliki Forest virus with membranes. J Virol 52: 281–
283.
Kraaijeveld CA, Jansen J, Benaissatrouw B, Snippe H
(1983). Delayed-type hypersensitivity in mice after in-
fection with avirulent Semliki Forest virus. Arch Virol
78: 115–118.
Kujala P, Ikaheimonen A, Ehsani N, Vihinen H, Auvinen
P, Kaariainen L (2001). Biogenesis of the Semliki Forest
virus RNA replication complex. J Virol 75: 3873–3884.
Kujala P, Rikkonen M, Ahola T, Kelve M, Saarma M,
Kaariainen L (1997). Monoclonal antibodies specific for
Semliki Forest virus replicase protein nsP2. J Gen Virol
78: 343–351.
Laakkonen P, Hyvonen M, Peranen J, Kaariainen L (1994).
Expression of Semliki Forest virus nsp1-specific methyl-
transferase in insect cells and in escherichia-coli. J Virol
68: 7418–7425.
Liljestrom P, Garoff H (1991). A new generation of animal
cell expression vectors based on the Semliki Forest virus
replicon. Biotechnology 9: 1356–1361.
Mabruk MJ, Flack AM, Glasgow GM, Smyth JM, Folan JC,
Bannigan JG, O’Sullivan MA, Sheahan BJ, Atkins GJ
(1988). Teratogenicity of the Semliki Forest virus mu-
tant ts22 for the foetal mouse: induction of skeletal and
skin defects. J Gen Virol 69: 2755–2762.
Mabruk MJ, Glasglow GM, Flack AM, Folan JC, Bannigan
JG, Smyth JM, O’Sullivan MA, Sheahan BJ, Atkins
GJ (1989). Effect of infection with the ts22 mutant of
Semliki Forest virus on development of the central ner-
vous system in the fetal mouse. J Virol 63: 4027–4033.
Mathiot CC, Grimaud G, Garry P, Bouquety JC, Mada A,
Daguisy AM, Georges AJ (1990). An outbreak of hu-
man Semliki Forest virus infections in Central African
Republic. Am J Trop Med Hyg 42: 386–393.
McIntosh BM, Brookworth C, Kokernot RH (1961). Isolation
of Semliki Forest virus from Aedes (Aedimorphus) ar-
genteopunctatus (THEOBALD) collected in Portuguese
East Africa. Trans R Soc Trop Med Hyg 55: 192.
Mehta S, Pathak S, Webb HE (1990). Induction of membrane
proliferation in mouse CNS by gold sodium thiomalate
with reference to increased virulence of the avirulent
Semliki Forest virus. Biosci Rep 10: 271–279.
Mehta S, Webb HE (1982). Lysosomal enzyme changes in
macrophages from mice given myocrisin and infected
with avirulent Semliki Forest virus. Br J Exp Pathol 63:
443–446.
Merits A, Vasiljeva L, Ahola T, Kaariainen L, Auvinen P
(2001). Proteolytic processing of Semliki Forest virus-
specific non-structural polyprotein by nsP2 protease.
J Gen Virol 82: 765–773.
Milner AR, Marshall ID (1984). Pathogenesis of in utero
infections with arbortigenic and nonarbortigenic al-
phaviruses in mice. J Virol 50: 66–72.
Mokhtarian F, Swoveland P (1987). Predisposition to EAE
induction in resistant mice by prior infection with
Semliki Forest virus. J Immunol 138: 3264–3268.
Morris MM, Dyson H, Baker D, Harbige LS, Fazakerley JK,
Amor S (1997). Characterization of the cellular and cy-
tokine response in the central nervous system following
Semliki Forest virus infection. J Neuroimmunol 74: 185–
197.
Muller U, Steinhoff U, Reis L, Hemmi S, Pavlovic J,
Zinkernagel RM, Aguet M (1994). Functional role of type
I and type II interferons in antiviral defense. Science 264:
1918–1921.
Oliver KR, Fazakerley JK (1998). Transneuronal spread
of Semliki Forest virus in the developing mouse ol-
factory system is determined by neuronal maturity.
Neuroscience 82: 867–877.
Oliver KR, Scallan MF, Dyson H, Fazakerley JK (1997). Sus-
ceptibility to a neurotropic virus and its changing dis-
tribution in the developing brain is a function of CNS
maturity. J NeuroVirol 3: 38–48.
Omar A, Koblet H (1988). Semliki Forest virus-particles
containing only the E1 envelope glycoprotein are in-
fectious and can induce cell cell-fusion. Virology 166:
17–23.
Parsons LM, Webb HE (1982a). Blood brain barrier distur-
bance and immunoglobulin G levels in the cerebrospinal
fluid of the mouse following peripheral infection
with the demyelinating strain of Semliki Forest virus.
J Neurol Sci 57: 307–318.
Parsons LM, Webb HE (1982b). Virus titers and persis-
tently raised white cell counts in cerebrospinal-fluid in
mice after peripheral infection with demyelinating Sem-
liki Forest virus. Neuropathol Appl Neurobiol 8: 395–
401.
Parsons LM, Webb HE (1984). Specific immunoglobulin G
in serum and cerebrospinal fluid of mice infected with
the demyelinating strain of Semliki Forest virus. Micro-
bios Lett 25: 135–140.
Pathak S, Webb HE (1974). Possible mechanisms for the
transport of Semliki Forest virus into and within mouse
brain: an electron microscopic study. J Neurol Sci 23:
175–184.
Pathak S, Webb HE (1978). An electron-microscopic study
of avirulent and virulent Semliki Forest virus in the
brains of different ages of mice. J Neurol Sci 39: 199–
211.
Pathak S, Webb HE (1983a). Effect of myocrisin (sodium
auro-thio-malate) on the morphogenesis of avirulent
Semliki Forest virus in mouse brain: an electron mi-
croscopical study. Neuropathol Appl Neurobiol 9: 313–
327.
Pathak S, Webb HE (1983b). Semliki Forest virus multipli-
cation in oligodendrocytes in mouse-brain with refer-
ence to demyelination. J Physiol (London) 339: 17.
Pathak S, Webb HE (1988). An electron microscopical study
of the replication of avirulent Semliki Forest virus in the
retina of mice. J Neurol Sci 85: 87–96.
Pathak S, Webb HE, Oaten SW, Bateman S (1976).
An electron-microscopic study of the development of
virulent and avirulent strains of Semliki Forest virus in
mouse brain. J Neurol Sci 28: 289–300.
SFV encephalitis
74 JK Fazakerley
Peranen J, Kaariainen L (1991). Biogenesis of type-I cyto-
pathic vacuoles in Semliki Forest virus-infected BHK
cells. J Virol 65: 1623–1627.
Peranen J, Rikkonen M, Liljestrom P, Kaariainen L (1990).
Nuclear-localization of Semliki Forest virus-specific
nonstructural protein-nsp2. J Virol 64: 1888–1896.
Peranen J, Takkinen K, Kalkkinen N, Kaariainen L (1988).
Semliki Forest virus-specific nonstructural protein nsp3
is a phosphoprotein. J Gen Virol 69: 2165–2178.
Purves D, Riddle DR, LaManita A-S (1992). Iterated pat-
terns of brain circuitry (or how the cortex gets its spots).
Trends Neurol Sci 15: 632–638.
Pusztai R, Gould E, Smith H (1971). Infection pattern in
mice of an avirulent and virulent strain of Semliki Forest
virus. Br J Exp Pathol 52: 669–677.
Rikkonen M, Peranen J, Kaariainen L (1992). Nuclear and
nucleolar targeting signals of Semliki Forest virus non-
structural protein-nsp2. Virology 189: 462–473.
Rikkonen M, Peranen J, Kaariainen L (1994). ATPase and
GTPase activities associated with Semliki Forest virus
nonstructural protein nsP2. J Virol 68: 5804–5810.
Robin Y, Bourdin P, Le Gonidec G, Heme G (1974). Virus
de la foret de Semliki et encephalomyelites equines au
Senegal. Ann Microbiol (Inst Pasteur) 125: 235–241.
Santagati MG, Maatta JA, Itaranta PV, Salmi AA, Hinkkanen
AE (1995). The Semliki Forest virus E2 gene as a viru-
lence determinant. J Gen Virol 75: 47–52.
Santagati MG, Maatta JA, Roytta M, Salmi AA, Hinkkanen
AE (1998). The significance of the 30-nontranslated re-
gion and E2 amino acid mutations in the virulence of
Semliki Forest virus in mice. Virology 243: 66–77.
Scallan MF, Allsopp TE, Fazakerley JK (1997). bcl-2 acts
early to restrict Semliki Forest virus replication and de-
lays virus-induced programmed cell death. J Virol 71:
1583–1590.
Scallan MF, Fazakerley JK (1999). Aurothiolates enhance
the replication of Semliki Forest virus in the CNS and
the exocrine pancreas. J NeuroVirol 5: 392–400.
Seamer J, Fitzgeorge R, Smith CE (1967a). Resistance of the
short-tailed vole Microtus agrestis (L.) to infection with
two arboviruses. Br J Exp Pathol 48: 463–467.
Seamer J, Randles WJ, Fitzgeorge R (1967b). The course of
Semliki Forest virus infection in mice. Br J Exp Pathol
48: 395–402.
Smithburn KC, Haddow WJ (1944). Semliki Forest virus. I.
Isolation and pathogenic properties. J Immunol 49: 141–
145.
Snijders A, Benaissa-Trouw BJ, Oosterlaken TA, Puijk WC,
Posthumus WP, Meloen RH, Boere WA, Oosting JD,
Kraaijeveld CA, Snippe H (1991). Identification of linear
epitopes on Semliki Forest virus E2 membrane protein
and their effectiveness as a synthetic peptide vaccine.
J Gen Virol 72: 557–565.
Snijders A, Benaissatrouw BJ, Snippe H, Kraaijeveld CA
(1992a). Immunogenicity and vaccine efficacy of syn-
thetic peptides containing Semliki Forest virus B-cell
and T-cell epitopes. J Gen Virol 73: 2267–2272.
Snijders A, Benaissatrouw BJ, Visservernooy HJ, Fernandez
I, Snippe H, Kraaijeveld CA (1992b). A delayed-type
hypersensitivity-inducing T-cell epitope of Semliki
Forest virus mediates effective T-helper activity for
antibody-production. Immunology 77: 322–329.
SoiluHanninen M, Eralinna JP, Hukkanen V, Roytta M,
Salmi AA, Salonen R (1994). Semliki-Forest virus in-
fects mouse-brain endothelial-cells and causes blood-
brain-barrier damage. J Virol 68: 6291–6298.
SoiluHanninen M, Roytta M, Salmi AA, Salonen R (1997).
Semliki Forest virus infection leads to increased expres-
sion of adhesion molecules on splenic T-cells and on
brain vascular endothelium. J NeuroVirol 3: 350–360.
Subak-Sharpe I, Dyson H, Fazakerley JK (1993). In vivo de-
pletion of CD8+T cells prevents lesions of demyelina-
tion in Semliki Forest virus infection. J Virol 67: 7629–
7633.
Suckling AJ, Jagelman S, Webb HE (1982). Immunoglobu-
lin synthesis in nude (nu/nu), nu/+and reconstituted
nu/nu mice infected with a demyelinating strain of
Semliki Forest virus. Clin Exp Immunol 47: 283–288.
Suckling AJ, Pathak S, Jagelman S, Webb HE (1978).
Virus associated demyelination: a model using aviru-
lent Semliki Forest virus infection of mice. J Neurol Sci
36: 147–154.
Suomalainen M, Liljestrom P, Garoff H (1992). Spike
protein-nucleocapsid interactions drive the budding of
alphaviruses. J Virol 66: 4737–4747.
Suopanki J, Sawicki DL, Sawicki SG, Kaariainen L (1998).
Regulation of alphavirus 26S mRNA transcription by
replicase component nsP2. J Gen Virol 79: 309–319.
Takkinen K (1986). Complete nucleotide-sequence of the
nonstructural protein genes of Semliki Forest virus.
Nucl Acid Res 14: 5667–5682.
Tan KB, Sambrook JF, Bellett AJ (1969). Semliki Forest virus
temperature-sensitive mutants: isolation and character-
ization. Virology 38: 427–439.
Tarbatt CJ, Glasgow GM, Mooney DA, Sheahan BJ, Atkins
GJ (1997). Sequence analysis of the avirulent, demyeli-
nating A7 strain of Semliki Forest virus. J Gen Virol 78:
1551–1557.
Tubulekas I, Berglund P, Fleeton M, Liljestrom P (1997).
Alphavirus expression vectors and their use as recom-
binant vaccines: a minireview. Gene 190: 191–195.
Tuittila MT, Santagati MG, Roytta M, Maatta JA, Hinkkanen
AE (2000). Replicase complex genes of Semliki Forest
virus confer lethal neurovirulence. J Virol 74: 4579–
4589.
Vasiljeva L, Merits A, Auvinen P, Kaariainen L (2000). Iden-
tification of a novel function of the alphavirus capping
apparatus. J Biol Chem 275: 17281–17287.
Wahlberg JM, Garoff H (1992). Membrane-fusion process
of Semliki Forest virus 1. Low pH-induced rearrange-
ment in spike protein quaternary structure precedes
virus penetration into cells. J Cell Biol 116: 339–348.
Wang YF, Sawicki SG, Sawicki DL (1991). Sindbis virus
nsP1 functions in negative-strand RNA synthesis. J Virol
65: 985–988.
Willems WR, Kaluza G, Boschek GB, Bauer H (1979).
Semliki Forest virus: cause of a fatal case of human en-
cephalitis. Science 203: 1128–1129.
... The age dependence of neuroinvasiveness is well known from other flavi-and alphaviruses. The age until which neuroinvasion by peripheral inoculations can be achieved ranges from 12 days (Semliki forest virus, SFV) (Fazakerley 2002), over 6 weeks (WNV) (Eldadah et al. 1967), and 4 months (SLEV) (Andersen andHanson 1974, Monath et al. 1974) to even adult age (Banzi virus) Jacoby and Bhatt 1976). Thus it seems that the age-dependent neuroinvasiveness of the Austrian USUV strain is the lowest ever been tested among several mosquito-borne viruses. ...
... SFV infection causes self-limited febrile diseases in humans including fever, headache, and arthralgia, and most infections are asymptomatic or very mild. However, in mice, SFV is able to enter the central nervous system (CNS) and infect neurons and oligodendrocytes, leading fatal diseases to the animals (Fazakerley, 2002). The features of relatively low level of virulence in humans, high neuroinvasion in mice, and ability to be handled in BSL-2 laboratory, make SFV a good model to investigate the mechanisms of viral replication and pathogenesis, and to conduct antiviral screening for alphaviruses. ...
Article
Full-text available
Alphaviruses, which contain a variety of mosquito-borne pathogens, are important pathogens of emerging/re-emerging infectious diseases and potential biological weapons. Currently, no specific antiviral drugs are available for the treatment of alphaviruses infection. For most highly pathogenic alphaviruses are classified as risk group-3 agents, the requirement of biosafety level 3 (BSL-3) facilities limits the live virus-based antiviral study. To facilitate the antiviral development of alphaviruses, we developed a high throughput screening (HTS) platform based on a recombinant Semliki Forest virus (SFV) which can be manipulated in BSL-2 laboratory. Using the reverse genetics approach, the recombinant SFV and SFV reporter virus expressing eGFP (SFV-eGFP) were successfully rescued. The SFV-eGFP reporter virus exhibited robust eGFP expression and remained relatively stable after four passages in BHK-21 cells. Using a broad-spectrum alphavirus inhibitor ribavirin, we demonstrated that the SFV-eGFP can be used as an effective tool for antiviral study. The SFV-eGFP reporter virus-based HTS assay in a 96-well format was then established and optimized with a robust Z' score. A section of reference compounds that inhibit highly pathogenic alphaviruses were used to validate that the SFV-eGFP reporter virus-based HTS assay enabled rapid screening of potent broad-spectrum inhibitors of alphaviruses. This assay provides a safe and convenient platform for antiviral study of alphaviruses.
... 6 Members of the Old World alphaviruses rarely cause neurological disease in humans, although SFV and SINV can infect neuron cells and induce encephalitis in rodents. 7,8 The virions of alphaviruses are spherical and approximately 70 nm in diameter. The outermost layer of the virion is a T = 4 icosahedral protein shell that is assembled by 240 copies of the envelope protein E1 and E2 heterodimer. ...
Article
Semliki Forest virus (SFV) is an alphavirus that uses the very-low-density lipoprotein receptor (VLDLR) as a receptor during infection of its vertebrate hosts and insect vectors. Herein, we used cryoelectron microscopy to study the structure of SFV in complex with VLDLR. We found that VLDLR binds multiple E1-DIII sites of SFV through its membrane-distal LDLR class A (LA) repeats. Among the LA repeats of the VLDLR, LA3 has the best binding affinity to SFV. The high-resolution structure shows that LA3 binds SFV E1-DIII through a small surface area of 378 Å2, with the main interactions at the interface involving salt bridges. Compared with the binding of single LA3s, consecutive LA repeats around LA3 promote synergistic binding to SFV, during which the LAs undergo a rotation, allowing simultaneous key interactions at multiple E1-DIII sites on the virion and enabling the binding of VLDLRs from divergent host species to SFV.
... 13,103,104 Demyelination is the hallmark of CHIKV neonatal encephalopathy, which is caused by autoreactive CD8 + T lymphocytes to clear infected cells. 105,106 CD8 + T cells are frequently seen in the CSF of cynomolgus macaques, the only nonhuman primate model challenged by CHIKV. 107 ...
Article
Full-text available
Structure: Chikungunya virus (CHIKV) is an arthropod-borne ribonucleic acid (RNA) virus, classified in the genus alphavirus in the family Togaviridae. Clinical presentation: Perinatal/neonatal infections are rare, but some infants can develop fever, thrombocytopenia, lymphopenia, pigmentary changes, and a maculopapular rash. The neurocognitive outcome of some infants with vertically transmitted mother-to-child perinatal infections and CHIKV neonatal encephalopathy can be poor. Diagnosis: The diagnosis of CHIKV infections can be confirmed by the detection of chikungunya viral RNA via real-time reverse-transcription polymerase chain reaction (RT-PCR) and/or specific immunoglobulin (Ig)M and IgG serology. Treatment: Currently, no specific antiviral treatment(s) are available for CHIKV, and management is limited to supportive care by maintaining adequate intravascular volume by intravenous fluids and oral rehydration. Infants exposed in utero or during the perinatal period need to be monitored for adverse neurocognitive outcomes.
... The Semliki Forest virus (SFV), the positive-stranded RNA virus, contains approximately 13,000 base pair genomes which encode nine proteins. Uganda Virus Research Institute first identified SFV from mosquitoes of the Semliki Forest in 1942 and it spreads by mosquito bites (Atkins et al. 1999;Contu et al. 2021;Fazakerley 2002). ...
Article
Full-text available
Triterpenes are naturally occurring derivatives biosynthesized following the isoprene rule of Ruzicka. The triterpenes have been reported to possess a wide range of therapeutic applications including anti-viral properties. In this review, the recent studies (2010–2020) concerning the anti-viral activities of triterpenes have been summarized. The structure activity relationship studies have been described as well as brief biosynthesis of these triterpenes is discussed. © 2022, The Author(s), under exclusive licence to Springer Nature B.V.
... For example, the neuroinvasive potential of WNV is strain-dependent, but mainly due to differential strain ability to reach the CNS [43]. In some cases, this difference could also be due to different replication capacities in BBB cells or directly in neurons, as already demonstrated for WNV [59] and Semliki Forest Virus [87]. It is documented that there is a close relationship between European USUV strains (nucleotide identity 99%) while there is a greater divergence between African USUV isolates (nucleotide identity 96%). ...
Article
Full-text available
Background Usutu virus (USUV) is an emerging neurotropic arthropod-borne virus recently involved in massive die offs of wild birds predominantly reported in Europe. Although primarily asymptomatic or presenting mild clinical signs, humans infected by USUV can develop neuroinvasive pathologies (including encephalitis and meningoencephalitis). Similar to other flaviviruses, such as West Nile virus, USUV is capable of reaching the central nervous system. However, the neuropathogenesis of USUV is still poorly understood, and the virulence of the specific USUV lineages is currently unknown. One of the major complexities of the study of USUV pathogenesis is the presence of a great diversity of lineages circulating at the same time and in the same location. Methods The aim of this work was to determine the neurovirulence of isolates from the six main lineages circulating in Europe using mouse model and several neuronal cell lines (neurons, microglia, pericytes, brain endothelial cells, astrocytes, and in vitro Blood-Brain Barrier model). Results Our results indicate that all strains are neurotropic but have different virulence profiles. The Europe 2 strain, previously described as being involved in several clinical cases, induced the shortest survival time and highest mortality in vivo and appeared to be more virulent and persistent in microglial, astrocytes, and brain endothelial cells, while also inducing an atypical cytopathic effect. Moreover, an amino acid substitution (D3425E) was specifically identified in the RNA-dependent RNA polymerase domain of the NS5 protein of this lineage. Conclusions Altogether, these data show a broad neurotropism for USUV in the central nervous system with lineage-dependent virulence. Our results will help to better understand the biological and epidemiological diversity of USUV infection
Article
Full-text available
Tick-borne encephalitis (TBE) is a viral arthropod infection, endemic to large parts of Europe and Asia, and is characterised by neurological involvement, which can range from mild to severe, and in 33–60% of cases, it leads to a post-encephalitis syndrome and long-term morbidity. While TBE virus, now identified as Orthoflavivirus encephalitidis, was originally isolated in 1937, the pathogenesis of TBE is not fully appreciated with the mode of transmission (blood, tick, alimentary), viral strain, host immune response, and age, likely helping to shape the disease phenotype that we explore in this review. Importantly, the incidence of TBE is increasing, and due to global warming, its epidemiology is evolving, with new foci of transmission reported across Europe and in the UK. As such, a better understanding of the symptomatology, diagnostics, treatment, and prevention of TBE is required to inform healthcare professionals going forward, which this review addresses in detail. To this end, the need for robust national surveillance data and randomised control trial data regarding the use of various antivirals (e.g., Galidesivir and 7-deaza-2′-CMA), monoclonal antibodies, and glucocorticoids is required to improve the management and outcomes of TBE.
Article
Cholesterol-24-hydroxylase (CH24H or Cyp46a1) is a reticulum-associated membrane protein that plays an irreplaceable role in cholesterol metabolism in the brain and has been well-studied in several neuro-associated diseases in recent years. In the present study, we found that CH24H expression can be induced by several neuroinvasive viruses, including vesicular stomatitis virus (VSV), rabies virus (RABV), Semliki Forest virus (SFV) and murine hepatitis virus (MHV). The CH24H metabolite, 24-hydroxycholesterol (24HC), also shows competence in inhibiting the replication of multiple viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). 24HC can increase the cholesterol concentration in multivesicular body (MVB)/late endosome (LE) by disrupting the interaction between OSBP and VAPA, resulting in viral particles being trapped in MVB/LE, ultimately compromising VSV and RABV entry into host cells. These findings provide the first evidence that brain cholesterol oxidation products may play a critical role in viral infection.
Thesis
Les virus émergents sont définis comme étant des virus gagnant de nouveaux territoires ou comme des virus connus mais ayant muté ou recombiné au point de devenir beaucoup plus transmissibles et contagieux dans la population. Ces virus posent des problèmes majeurs pour la santé publique car ils peuvent être à l’origine de maladies parfois inconnues et potentiellement graves notamment neuroinvasives. C’est le cas, par exemple, avec l’émergence récente de deux virus neurotropes : Zika (ZIKV) et Usutu (USUV). ZIKV a été responsable d’épidémies principalement en Polynésie Française en 2013 et en Amérique Latine en 2015-2016 où il a été la cause chez certains patients d’atteintes neurologiques (encéphalite, méningo-encéphalite, syndrome de Guillain-Barré, microcéphalie, etc.). USUV, quant à lui, a émergé récemment en Europe, où il a été impliqué dans une importante mortalité aviaire (principalement chez le merle noir) mais aussi dans des infections neuroinvasives chez l’Homme (encéphalite, méningo-encéphalite, etc.). Malgré cela, la pathogenèse de ces deux virus reste peu explorée. L'objectif de cette thèse était de caractériser l’effet de l’infection du ZIKV sur la structure et l’homéostasie de la barrière hémato-encéphalique (BHE) ainsi que d'évaluer et de comparer la neuropathogénicité de différentes lignées d’USUV en combinant des approches in vivo et in vitro. Nos résultats suggèrent premièrement une interaction complexe entre le ZIKV et la BHE qui pourrait déclencher une inflammation locale, le recrutement de leucocytes et une éventuelle perturbation vasculaire cérébrale sur du long terme. Deuxièmement, pour USUV, nos résultats corrèlent avec les pathologies inflammatoires cérébrales engendrées chez l’Homme comme l’encéphalite. De plus, suite à l’analyse des lignées circulantes d’USUV, nos résultats suggèrent que certaines lignées d’USUV peuvent avoir une neurovirulence plus importante, notamment la lignée Europe 2. L'émergence et la propagation rapide des nouveaux virus illustrent le défi complexe et constant pour la santé humaine. Aujourd’hui, de nombreux facteurs tels que les modifications écologiques, l’augmentation des déplacements animaliers et de la densité de la population mondiale ainsi que l’amélioration des moyens de transport favorisent l’apparition des virus émergents et facilitent leur transmission. Pour ces raisons, il est nécessaire d’étudier plus en détails les mécanismes impliqués dans l’infection virale (entrée du virus dans les cellules, sa transmission, sa multiplication et etc.) pour développer des traitements antiviraux adaptés ou des vaccins spécifiques.Mots clés : Virus Zika, Virus Usutu, flavivirus, système nerveux central, neurovirulence et neurotropisme
Article
Full-text available
Multiple sclerosis (MS) is a common inflammatory demyelinating disease of the central nervous system. Although the etiology of MS is unknown, genetics and environmental factors, such as infections, play a role. Viral infections of mice have been used as model systems to study this demyelinating disease of humans. Three viruses that have long been studied in this capacity are Theiler’s murine encephalomyelitis virus, mouse hepatitis virus, and Semliki Forest virus. This review describes the viruses themselves, the infection process, the disease caused by infection and its accompanying pathology, and the model systems and their usefulness in studying MS.
Article
Full-text available
Both genomic and subgenomic RNAs of theAlphavirus have m7G(5′)ppp(5′)N (cap0 structure) at their 5′ end. Previously it has been shown thatAlphavirus-specific nonstructural protein Nsp1 has guanine-7N-methyltransferase and guanylyltransferase activities needed in the synthesis of the cap structure. During normal cap synthesis the 5′ γ-phosphate of the nascent viral RNA chain is removed by a specific RNA 5′-triphosphatase before condensation with GMP, delivered by the guanylyltransferase. Using a novel RNA triphosphatase assay, we show here that nonstructural protein Nsp2 (799 amino acids) of Semliki Forest virus specifically cleaves the γ,β-triphosphate bond at the 5′ end of RNA. The same activity was demonstrated for Nsp2 of Sindbis virus, as well as for the amino-terminal fragment of Semliki Forest virus Nsp2-N (residues 1–470). The carboxyl-terminal part of Semliki Forest virus Nsp2-C (residues 471–799) had no RNA triphosphatase activity. Replacement of Lys-192 by Asn in the nucleotide-binding site completely abolished RNA triphosphatase and nucleoside triphosphatase activities of Semliki Forest virus Nsp2 and Nsp2-N. Here we provide biochemical characterization of the newly found function of Nsp2 and discuss the unique properties of the entire Alphavirus-capping apparatus.
Article
Summary Eleven monoclonal antibodies (MAs) reacted strongly in an enzyme immunoassay with virulent Semliki Forest virus (SFV) replicating in L cell monolayers. Three MAs showed a considerably diminished reaction with an avirulent strain of SFV both in enzyme immunoassays and plaque reduction tests.
Article
Summary After subcutaneous infection of mice with Semliki Forest virus, a delayed-type hypersensitivity (DH) could be demonstrated by footpad swelling. Pretreatment with cyclophosphamide resulted in enhanced DH if neutralizing antibodies were undetectable in serum.
Article
1. A filterable virus has been isolated from Aëdes abnormalis group mosquitoes caught in the Semliki Forest in western Uganda. It has been called the Semliki Forest virus. 2. The agent exerts its principal pathogenic properties on the central nervous system of susceptible hosts. It is virulent for, and lethal to, mice by various routes of inoculation, and for guinea-pigs, rabbits, and rhesus and red-tail monkeys by intracerebral inoculation. It is effective in very great dilution. When introduced extraneurally into resistant hosts, or even some susceptible hosts, it induces the formation of protective antibody without causing marked illness. 3. In mice inoculated intracerebrally the virus is present in the blood and various other tissues in considerable concentration before the onset of objective illness. It has been demonstrated in the circulation of certain other inoculated animals, namely the red-tail and blue monkeys. 4. The virus can be preserved by drying while frozen. It remains viable in saline at low temperatures for a number of days but gradually loses potency. It retains potency for several weeks when stored in serum in the refrigerator. It is not excessively sensitive to room or incubator temperatures. It survives heating at 60 C for ½ hour but not for 1 hour. It is inactivated by heating at 62 C for 30 minutes. 5. Lesions caused by the virus in various animals have been described.
Article
Parsons L.M. & Webb H.E.1982Neuropathology and Applied Neurobiology 8, 395–401 Virus titres and persistently raised white cell counts in cerebrospinal fluid in mice after peripheral infection with demyelinating Semliki Forest virus Virus titres and cell counts in the cerebrospinal fluid of mice have been examined following intraperitoneal inoculation of demyelinating Semliki Forest virus (SFV). Virus titres in cerebrospinal fluid were highest 2–4 days after infection and fell again after day 4, 48 h before reduction in brain titres. White cell counts were very low in the first 48 h and showed a steep rise from 4 days after infection to a peak on day 6. After day 8, cell numbers fell to a plateau level which was sustained up to day 61. The significance of these findings in relation to the pathogenesis of virus-induced disease of the central nervous system is discussed.
Article
In neonatal mice the A7(74) and L10 strains of Semliki Forest virus (SFV) are virulent. In 3- to 4-week-old mice the L10 strain is virulent, the A7(74) strain is avirulent. Following intraperitoneal inoculation of 3- to 4-week-old mice both strains produce a transient plasma viremia. This is cleared by IgM antibodies. IgG antibodies of all subclasses are produced. The distribution of viral RNA in the brain as determined by autoradiographic analysis of in situ hybridizations shows that in all cases virus is first apparent as small foci of infected cells around cerebral capillaries. In both neonatal and 3- to 4-week-old mice infected with L10 or neonatal mice infected with A7(74), infection spreads rapidly from the original foci to infect large areas throughout the brain. Both neurons and glial cells are infected resulting in pycnosis and death of the animals. In the brains of 3- to 4-week-old mice infected with A7(74) virus there is little spread from the original perivascular foci. Again neurons and oligodendrocytes are infected but cellular destruction is minimal. The same pattern of A7(74) infection is observed in 3- to 4-week-old athymic nu/nu mice and mice with severe combined immunodeficiency, indicating that failure to spread is not related to specific immune responses. Furthermore, in nu/nu and SCID mice the small restricted foci of A7(74) infection persist. Comparison of the replication of these two viruses by electronmicroscopy shows that although A7(74) virus replicates completely in the neurons of neonatal mice, the virus is unable to bud from the neurons of 3- to 4-week-old mice and aggregates of viral RNA and capsid accumulate. We conclude that there is an age-related restriction of A7(74) replication in mouse neurons and that this restriction is not associated with the maturity of virus-specific immune responses but probably reflects age-related changes in neurons.
Article
We have recently shown that the 3′-nontranslated region (3′-NTR) of the avirulent Semliki Forest virus A7(74) [SFVA7(74)] contains a unique sequence of 101 nucleotides and five repetitive nucleotide units whereas the 3′-NTR of the neurovirulent SFV4 has only two repeats. A chimeric virus was constructed by replacing the entire 3′-NTR of the SFV4 clone with the A7(74) 3′-NTR. The hybrid replicated efficiently in the central nervous system (CNS) of adult Balb/c mice and, similarly to SFV4, led to high mortality after intraperitoneal inoculation. In contrast, another chimeric virus, CME2, containing the E2 gene of the avirulent SFVA7(74) virus in the SFV4 clone was recently shown to be avirulent for mice. Several derivatives with single-site or a constellation of amino acid mutations were constructed. Two single-site E2 mutants, Val37Ile and Asn212Ser, displayed an attenuated phenotype in mice with mortality reduced from 90 to 48 and 43%, respectively. None of the multiple site mutants were significantly attenuated. Adult female mice showed a greater resistance to SFV infection than male mice. The SFV hybrid viruses, CM3NTR and CME2, reached the CNS similarly to the parental viruses, but the single-site E2 mutants were only sporadically found in the CNS. We conclude that in mice the 3′-NTR does not play a significant role in the pathogenesis of Semliki Forest virus and that specific E2 amino acid mutations reduce the virulence, especially in female mice. The results additionally suggest that individual amino acid mutations in the E2 glycoprotein affect the efficiency of migration into the CNS.
Article
Alphavirus vectors have become widely used in basic research to study the structure and function of proteins and for protein production purposes. Development of a variety of vectors has made it possible to deliver foreign sequences as naked RNA or DNA, or as suicide virus particles produced using helper vector strategies. Preliminary reports also suggest that these vectors may be useful for in vivo applications where transient, high-level protein expression is desired, such as recombinant vaccines. The initial studies have already shown that alphavirus vaccines can induce strong humoral and cellular immune responses with good immunological memory and protective effects.