Download Vesicular stomatitis virus infects resident cells of the central nervous

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cellular differentiation wikipedia , lookup

Tissue engineering wikipedia , lookup

Cell encapsulation wikipedia , lookup

Cell culture wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Amitosis wikipedia , lookup

List of types of proteins wikipedia , lookup

Interferon wikipedia , lookup

JADE1 wikipedia , lookup

Transcript
Virology 400 (2010) 187–196
Contents lists available at ScienceDirect
Virology
j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / y v i r o
Vesicular stomatitis virus infects resident cells of the central nervous system and
induces replication-dependent inflammatory responses
Vinita S. Chauhan, Samantha R. Furr, David G. Sterka Jr., Daniel A. Nelson, Megan Moerdyk-Schauwecker,
Ian Marriott, Valery Z. Grdzelishvili ⁎
Department of Biology, 9201 University City Boulevard, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
a r t i c l e
i n f o
Article history:
Received 9 December 2009
Returned to author for revision
11 January 2010
Accepted 20 January 2010
Available online 20 February 2010
Keywords:
Vesicular stomatitis virus
Encephalitis
Rhabdoviruses
Sendai virus
Paramyxovirus
Microglia
Astrocytes
a b s t r a c t
Vesicular stomatitis virus (VSV) infection of mice via intranasal administration results in a severe
encephalitis with rapid activation and proliferation of microglia and astrocytes. We have recently shown that
these glial cells express RIG-I and MDA5, cytosolic pattern recognition receptors for viral RNA. However, it is
unclear whether VSV can replicate in glial cells or if such replication is required for their inflammatory
responses. Here we demonstrate that primary microglia and astrocytes are permissive for VSV infection and
limited productive replication. Importantly, we show that viral replication is required for robust
inflammatory mediator production by these cells. Finally, we have confirmed that in vivo VSV administration
can result in viral infection of glial cells in situ. These results suggest that viral replication within resident
glial cells might play an important role in CNS inflammation following infection with VSV and possibly other
neurotropic nonsegmented negative-strand RNA viruses.
© 2010 Elsevier Inc. All rights reserved.
Introduction
Vesicular stomatitis virus (VSV) is a prototypic nonsegmented
negative-strand RNA virus of the family Rhabdoviridae, order Mononegavirales. Members of this order include many medically important
pathogens including the lethal rabies, Ebola, Marburg, Nipah and
Hendra viruses. VSV has a limited human pathogenicity, making it an
attractive model for the study of other viruses in this order, including
closely related rabies virus, another member of the family Rhabdoviridae. Intranasal inoculation of mice with VSV leads to infection of
the olfactory bulb via the olfactory neurons and subsequently leads to
infection throughout the central nervous system (CNS) (Bi et al.,
1995; Lundh, 1990; Lundh et al., 1987; Miyoshi et al., 1971). This
infection is associated with acute encephalitis, breakdown of the
blood–brain barrier, and a high degree of mortality similar to that
seen with rabies infections (Huneycutt et al., 1993). Interestingly,
VSV-associated encephalitis appears to be T-cell independent (Frei et
al., 1989; Huneycutt et al., 1993; Nansen et al., 2000), suggesting a role
for resident CNS cells in the initiation and/or progression of the CNS
inflammation. While previous studies have demonstrated that
neurons are readily infected by VSV (Bi et al., 1995), the susceptibility
of other glial cell types to infection with this virus remains unclear.
⁎ Correspondence address. Fax: +1 704 687 3128.
E-mail address: [email protected] (V.Z. Grdzelishvili).
0042-6822/$ – see front matter © 2010 Elsevier Inc. All rights reserved.
doi:10.1016/j.virol.2010.01.025
Microglia and astrocytes are resident glial cells of the CNS and are
ideally situated to detect and respond to viral pathogens. Upon
activation, these cells assume immune effector functions that include
inflammatory mediator production and MHC class II molecule
expression (for review, see Chauhan and Marriott, 2007). Importantly,
microglia and astrocytes appear to respond to VSV as the encephalitis
caused by this virus is associated with proliferation of these cell types
and increased MHC class II molecule expression on their surfaces (Bi
et al., 1995). Recent studies from our laboratory have shown that
microglia and astrocytes express retinoic acid-inducible gene-I (RIGI) and melanoma differentiation-associated gene-5 (MDA5) (Furr et
al., 2008), two members of the (RIG-I)-like receptor (RLR) family of
cytosolic proteins that can serve as intracellular pattern recognition
receptors for replicative viral RNA (Meylan et al., 2006; Takeuchi and
Akira, 2008; Takeuchi and Akira, 2009). Interestingly, the level of
expression of these receptors was upregulated in microglia and
astrocytes following exposure to VSV, consistent with a functional
role for RLRs in virally-induced immune responses by these resident
CNS cells (Furr et al., 2008). However, the ability of VSV to infect and
replicate within these glial cells has not been established.
In the present study, we demonstrate that primary murine
microglia and astrocytes are permissive for VSV infection both in
vitro and in vivo. Furthermore, we show that active replication of VSV
appears to be required for robust inflammatory mediator production
by these resident CNS cell types. These data suggest that viral
replication within resident glial cells may play an important role in
188
V.S. Chauhan et al. / Virology 400 (2010) 187–196
Fig. 1. The organization of negative-sense RNA genomes of the recombinant viruses
used in the present study. The viral RNA dependent RNA polymerase (consisting of L
and P proteins) sequentially synthesizes mRNAs in the order that they appear from the
3′ end of the genome. Initiation of each downstream gene occurs sequentially as the
polymerase pauses at the intergenic junction and then reinitiates synthesis of the next
gene. (A) Recombinant VSV-wt (Indiana serotype) genomic RNA. (B) Recombinant
VSV-GFP (Indiana serotype) encoding GFP as an extra gene between G and L genes. (C)
Recombinant VSV (Indiana serotype) HR1-1 mutant with a mutation resulting in an L
protein with a single substitution D to V at the L protein amino acid position 1671. (D)
Recombinant SeV (Fushimi strain) with GFP as an extra gene upstream of the NP gene.
SeV-GFP does not require trypsin in the medium as it has a wt monobasic trypsindependent cleavage site in the F protein mutated (Fm) to an oligobasic cleavage site,
allowing F activation in any cell type through a ubiquitous furin-like protease.
the development of CNS inflammation following VSV infection.
Interestingly, both primary astrocytes and microglia were susceptible
for infection by non-neurotropic Sendai virus (SeV, a paramyxovirus),
suggesting that glial cells are permissive for infection by both
neurotropic and non-neurotropic nonsegmented negative-strand
RNA viruses.
Results
VSV and SeV infect and replicate within primary murine microglia
and astrocytes
To directly examine the ability of VSV to infect and replicate in
astrocytes and microglia, we isolated primary glial cells from neonatal
C57BL/6 mice using protocols established in our laboratory (Harry et
al., 1998; Rasley et al., 2002a; Rasley et al., 2002b; Sterka et al., 2006).
The purity of the astrocyte and microglia cell preparations was
confirmed by confocal microscopy as described in Materials and
Methods. To assess the ability of VSV to infect and replicate within
these primary glial cells we have employed a recombinant VSV (VSVGFP) containing the GFP gene incorporated between the viral G and L
genes (Das et al., 2006) (Fig. 1B). Due to the negative-sense polarity of
the VSV RNA genome and the distant position of the GFP gene from the
3′-end of the genomic RNA, we could detect GFP only following
successful VSV-GFP replication within target cells (Das et al., 2006; and
data not shown).
Confluent cultures of microglia and astrocytes were infected with
VSV-GFP at an MOIBHK of 1. This MOIBHK value was calculated based on
the VSV-GFP titration on BHK-21 cells, which are exceptionally
susceptible to VSV infection and replication (Grdzelishvili et al.,
2005). It is therefore likely that the actual MOI values for microglia
and/or astrocytes were significantly lower than 1. This was confirmed
in dose–response studies with different amounts of VSV-GFP added to
glial cells and counting GFP-expressing cells at 12 h post infection (p.i.)
(data not shown). As shown in Fig. 2A, VSV-associated GFP
fluorescence was detectable in astrocytes by 12 h p.i. and was broadly
expressed at 24 h p.i. We also detected cellular accumulation of VSV N
and P proteins 24 h p.i. in both microglia and astrocytes (data not
shown). VSV replication in astrocytes was associated with only modest
cell death as determined by trypan blue staining (the percentage of
dead cells is indicated beneath each micrograph in Fig. 2A). This
finding was confirmed by the robust accumulation of GFP in these cells
at 24 h p.i. as determined by Western blot analysis (Fig. 2B).
Interestingly, VSV-associated GFP expression was detected as rapidly
as 6 h p.i. in microglial cells as determined by Western blot analysis
(Fig. 2B) and was readily detectable by confocal microscopy at 12 h p.i.
(Fig. 2A). However, in contrast to astrocytes, VSV elicited markedly
higher cytopathic effect and cell death in microglial cells as can be seen
by the morphological changes shown in Fig. 2A at 24 and 48 h p.i., the
reduction in total protein content in samples obtained at these time
points (Fig. 2B), and by the marked increase in the number of dead
cells as determined by trypan blue staining (Fig. 2A).
In addition to VSV–GFP infections, we performed similar experiments in which astrocytes and microglia were infected with wild-type
VSV (VSV-wt, Fig. 1A) and a similar accumulation pattern of VSV N
and P proteins in these cells was confirmed by Western blot analysis
using polyclonal VSV antibodies (data not shown).
To determine whether the ability of VSV to replicate within
resident CNS cells is specific for this neurotropic RNA virus, microglia
and astrocytes were also tested for their susceptibility for the
paramyxovirus, Sendai virus (SeV, family Paramyxoviridae), another
prototypic nonsegmented negative-strand RNA virus that is distantly
related to VSV but lacks the ability to cross the blood–brain barrier
(Lundh et al., 1987). Primary murine glial cells were exposed to
recombinant SeV containing the GFP gene (SeV-GFP, Fig. 1D)
(Wiegand et al., 2007) at an MOIVERO of 1 (calculated based on the
SeV-GFP titration on Vero cells) and viral infection and replication
was again assessed by fluorescence microscopy and Western blot
analysis. As shown in Fig. 2C, SeV-associated GFP fluorescence was
readily detectable in both astrocytes and microglia at 48 h p.i., and this
observation was confirmed by Western blot analysis for GFP
expression (Fig. 2D). Interestingly, such infection and viral replication
failed to elicit a significant cell death in either astrocytes or microglia
(Figs. 2C and D). Together, these data demonstrate the ability of SeV to
infect murine glial cells and suggest that astrocytes and microglia are
permissive for infection by both neurotropic and non-neurotropic
nonsegmented negative-strand RNA viruses.
To confirm that GFP accumulation shown in Fig. 2 correlated with
the production of new copies of viral genomic RNA and new infectious
virus particles, astrocytes and microglia were infected with VSV-GFP
or SeV-GFP at MOIBHK or MOIVero of 1, respectively. Unbound viruses
were removed at 1 h p.i., and cells and media were collected at several
time points after infection and analyzed for viral RNA (Figs. 3A and B)
and infectious virus particles (Fig. 3C), respectively. Both microglia
and astrocytes produced detectable amounts of newly synthesized
VSV genomes (Fig. 3A, left panel, normalized to ribosomal RNA levels)
and N mRNA (Fig. 3B, left panel, normalized to ribosomal RNA levels).
VSV RNA levels (genomic and mRNA) sharply decreased between 24
and 48 h p.i. (Figs. 3A and B). However, due to a strong cytopathic
Fig. 2. VSV and SeV infect and replicate within primary murine microglia and astrocytes. Cells (1 × 106) were untreated or infected with VSV-GFP (A and B) or SeV-GFP (C and D) at an
MOIBHK (for VSV) or MOIVERO (for SeV) of 1 PFU per glial cell and cultured for 6, 12, 24, and 48 h. (A and C) Cells were imaged for the expression of virus-driven GFP
immunofluorescence (green) and cell nucleus (blue) and cell membrane (red)-associated immunofluorescence by confocal microscopy. In parallel studies, cell death was assessed
by trypan blue exclusion and the percentage of dead cells is indicated beneath each micrograph. (B and D) Whole-cell protein lysates were prepared and GFP expression determined
by Western blot analysis. Immunoblots shown are representative of three separate experiments. Parallel Coomassie-stained gels (shown below Western blots and indicated as “total
protein”) of the separated cell lysates loaded in the same order as in Western blot show the various housekeeping protein levels between the samples and demonstrate comparable
total protein levels between different samples.
V.S. Chauhan et al. / Virology 400 (2010) 187–196
189
190
V.S. Chauhan et al. / Virology 400 (2010) 187–196
Fig. 3. Analysis of VSV and SeV replication within primary murine microglia and astrocytes. (A and B) Bar graphs represent signal ratios of viral genomic RNA/cellular ribosomal RNA
(rRNA) (A) or viral N mRNA/cellular rRNA (B) isolated from microglia or astrocytes infected at an MOIBHK (for VSV-GFP) or MOIVERO (for SeV-GFP) of 1 PFU per glial cell. Total RNA
was extracted from the collected cells using the Quick-RNA MiniPrep kit (Zymo Research), and analyzed by Northern blotting using riboprobes complementary to the VSV or SeV
genomic RNA. These VSV and SeV ribiprobes were found to nonspecifically recognize N mRNA of the corresponding viruses. Autoradiographic images of the membranes were
obtained using a GE Healthcare Typhoon Phosphorimager, and RNA bands for viral genomic RNA, viral N mRNA and cellular rRNA (nonspecifically recognized by antigenomic
riboprobe) were quantitated using ImageQuant software (Molecular Dynamics). (C) Bar graphs represent numbers of new infectious viral particles produced in microglia (grey line)
and astrocytes (black line) infected at an MOIBHK (for VSV-GFP) or MOIVERO (for SeV-GFP) of 1 PFU per glial cell (done in triplicates). Medium was collected at the indicated time
point p.i., and virus titer was determined by standard plaque assay on BHK-21 (for VSV) or Vero cells (for SeV). Viral titers are shown in logarithmic scale for SeV.
effect of VSV in microglia beginning at 24 h p.i. (Figs. 2A and B), we
were unable to retrieve sufficient amounts of total RNA to make very
reliable calculations of viral RNA accumulation (all bands including
ribosomal RNA were extremely weak). In contrast to microglia, VSV
RNA levels steadily increased in astrocytes, which is consistent with
GFP accumulation (Figs. 2A and B). Interestingly, both microglia and
astrocytes produced very modest amounts of VSV particles (Fig. 3C, left
panel) with virus production peaking at 24 h p.i. and then sharply
declining at 48 h p.i. This decline was observed even in astrocytes,
which produced increasing amounts of VSV RNA between 24 and 48 h
p.i. (Figs. 3A and B). Similarly, SeV genomic (Fig. 3A, right panel) and N
mRNA (Fig. 3B, right panel) accumulation was higher in astrocytes
than in microglia. Also, in agreement with GFP accumulation data (Figs.
2C and D), no drop in viral RNA accumulation was observed between
24 and 48 h p.i. In general, both astrocytes and microglia supported
higher levels of virion production in SeV compared to VSV (Fig. 3C,
right panel), which can be explained by no significant cell death in
either astrocytes or microglia following SeV infection (Figs. 2C and D).
Primary murine microglia and astrocytes respond to VSV and
SeV infection by secreting key inflammatory mediators
Previous studies have indicated that resident CNS cell types
respond to the presence of VSV by the expression of MHC class II
molecules and production of NO (Bi et al., 1995). However, it is not
clear whether these effects occur as a consequence of direct viral
recognition by glial cells or as a secondary response to inflammatory mediators and/or damage-associated molecules released by
V.S. Chauhan et al. / Virology 400 (2010) 187–196
infected neurons. To directly assess the ability of glial cells to
respond to VSV we have utilized specific capture ELISAs to quantify
the production of key inflammatory cytokines by isolated cultures of
primary murine microglia and astrocytes following infection with
VSV. As shown in Figs. 4A and B, in response to VSV-wt at 12 and
24 h p.i. microglia produced significant levels of both IL-6 and TNFα, cytokines known to be produced at high levels during viral
encephalitis (Nuovo et al., 2005; Rempel et al., 2005), Maximal
cytokine responses in microglia were observed at viral MOIBHK of
0.1 and 1 at 12 and 24 h p.i., respectively (Figs. 4A and B), with
higher viral titers resulting in lower production consistent with cell
death at these MOIs (data not shown). Similarly, astrocytes also
produced significant levels of IL-6 and TNF-α at 12 and 24 h
following infection with VSV (Figs. 4A and B).
To determine whether these responses were specific for the
neurotropic VSV, we have performed similar experiments to determine the cytokine responses of microglia and astrocytes to SeV
infection. As shown in Figs. 4C and D, both microglia and astrocytes
produced detectable levels of IL-6 and TNF-α in response to SeV-GFP
at both 12 and 24 h p.i. Although these responses increased according
to the numbers of infectious viral particles introduced to the glial
cultures (Figs. 4C and D), the levels of IL-6 and TNF-α produced by
191
SeV-infected glial cells were substantially lower than those observed
for comparable numbers of infectious VSV particles (Fig. 4).
Viral replication is required for robust microglial and astrocyte immune
responses to VSV
While it is apparent that glial cells can directly respond to VSV
infection, it is unclear whether these cells respond to external/
internalized viral components or if viral replication is a prerequisite
for microglial and astrocyte immune responses. To begin to address
this question, we have performed experiments employing heatinactivated VSV. While exposure of microglia or astrocytes to heatinactivated VSV-wt resulted in production of detectable levels of both
IL-6 and TNF-α (Figs. 4E and F), the inflammatory cytokine production
was approximately 10-fold lower than that observed following
infection with viable VSV (Figs. 4A and B).
To further address this question, we have performed similar
experiments utilizing a host-range VSV mutant, HR1-1 (Fig. 1C),
previously generated and characterized in our laboratory (Grdzelishvili et al., 2005). The VSV HR1-1 mutant has a single nucleotide
substitution resulting in a mutant L protein with a D to V substitution
at position 1671 (Grdzelishvili et al., 2005). We have shown that the
Fig. 4. Production of inflammatory mediators following incubation with VSV-wt (A and B), SeV-GFP (C and D), heat inactivated VSV (E, F) or VSV HR1-1 mutant (G). Microglia and
astrocytes were uninfected or exposed to varying MOIBHK (for VSV, A and B) or MOIVERO (for SeV, C and D). At 12 and 24 h p.i., culture supernatants were assayed for the presence of
IL-6 (A and C) or TNF-α (B and D) by specific capture ELISA. Data are presented as the mean of duplicate measurements of samples from at least three separate experiments ± SEM.
(E–G) Exposure of primary murine microglia and astrocytes to heat inactivated VSV or VSV HR1-1 mutant (replication-impaired in the glial cells) elicits only modest inflammatory
mediator production by primary murine microglia and astrocytes. Microglia and astrocytes were untreated or exposed to varying MOIBHK of heat-inactivated VSV-wt (E and F) or VSV
HR1-1 (G). At 12 and 24 h p.i., culture supernatants were assayed for the presence of IL-6 (E and G) or TNF-α (F) by specific capture ELISA. Data are presented as the mean of duplicate
measurements of samples from at least three separate experiments ± SEM.
192
V.S. Chauhan et al. / Virology 400 (2010) 187–196
D1671V L mutation completely eliminates viral mRNA cap methylation at both the guanine-N7 and 2′-O-adenosine positions (Grdzelishvili et al., 2005) and results in subsequent non-translatability of
primary VSV transcripts (Grdzelishvili et al., 2005; Hercyk et al., 1988;
Horikami et al., 1992; Horikami and Moyer, 1982). As a result, VSV
HR1-1 displays a host-range (hr) phenotype characterized by severely
restricted growth in most cell types but normal replication in a very
limited number of “permissive” cells including the BHK-21 cell line,
which was used to produce this mutant virus (Grdzelishvili et al.,
2005). As shown in Fig. 4G, infection of microglia or astrocytes with
VSV HR1-1 results in detectable production of inflammatory cytokines
but such production was again 10-fold lower than that observed
following exposure to comparable numbers of infectious VSV-wt
particles (Fig. 4). Taken together, these data demonstrate that active
viral replication is required for robust inflammatory responses of
infected glial cells.
Intranasal administration of VSV results in murine glial cell infection
in situ
To confirm the ability of VSV to infect glial cells in situ we have
employed an established murine model of virally-induced encephalitis (Bi et al., 1995). VSV-wt was intranasally administrated to 3- to 4week-old male C57BL/6 mice. As shown in Fig. 5A, VSV inoculated
animals rapidly developed symptoms associated with CNS infection as
assessed by disease severity scoring in contrast to mice that received
inoculation with vehicle alone. Animals were sacrificed at 3 days p.i.
and whole brains were isolated in such a way as to avoid possible
contamination with any residual viral particles that might be present
in the olfactory canal. Brain tissue was homogenized and VSVexpressed proteins and infectious viral particle content was quantified by Western blot analysis and plaque assay, respectively. The N, P,
M and G proteins of VSV were detectable in whole brain protein
isolates as determined by Western blot analysis using a polyclonal
antibody directed against VSV (data not shown). In addition, we have
performed plaque assays using BHK-21 cells to confirm the presence
of infectious particles in the CNS. At 5 days following intranasal
administration, infected murine brain tissue yielded 8.98 × 103 ±
1.77 × 103 PFU/mg total brain tissue (n = 13). In addition, we have
confirmed the development of inflammation within the CNS following
intranasal VSV administration by direct quantification of pro- and
anti-inflammatory mediators in whole brain homogenates by specific
capture ELISAs. As shown in Fig. 5A, VSV infection resulted in marked
increases levels of the proinflammatory cytokine, IL-6. Interestingly,
this elevation was associated with a concomitant decrease in levels
of the immunosuppressive cytokine, IL-10, in infected brain tissue
(Fig. 5A). Together, these data confirmed that intranasal VSV
administration lead to establishment of brain tissue infection and
the resulting CNS inflammation.
Importantly, we have performed experiments to determine
whether resident glial cells are infected in situ following intranasal
VSV administration. To accomplish this goal, at 3 days p.i. brain tissue
was perfused in situ, isolated, and prepared for confocal microscopic
analysis to determine whether VSV proteins co-localize with cell typespecific glial markers. As shown in the representative micrographs in
Fig. 5B, the number of astrocytes (GFAP+) and microglia/macrophages (F4/80+) increases in the cortex at 3 days post VSV
administration consistent with a reactive gliosis, and increases in
VSV-associated immunofluorescence are readily apparent. Importantly, individual astrocytes and microglia/macrophages containing
VSV components can be observed in these cortical fields with colocalized VSV and cell type-specific fluorescence signals (yellow in
Fig. 5B) and are indicated in this figure.
In addition, glial cells were purified from infected whole brain
tissue homogenates by Percoll gradient and the presence of VSV
structural proteins was determined by Western blot analysis. As
Fig. 5. VSV infects microglia and astrocytes in situ following intranasal administration.
C57BL/6 mice were uninfected (0) or inoculated intranasally with 4 × 107 PFUs of VSVwt (VSV). At 5 days p.i., disease severity was assessed according to appearance and
behavior, and at 3 days p.i. levels of IL-6 and IL-10 were quantified in whole brain
homogenates by specific capture ELISA (A). Severity was scored from 1 (no detectable
behavioral differences) to 5 (moribund). Cross symbol indicates a moribund animal
that was euthanized prior to day 5. Asterisk indicates statistically significant difference
from uninfected cells (0). In addition, at 3 days post-infection brain tissue was perfused
in situ, isolated, and prepared for immunofluorescent microscopic analysis (B). B shows
representative GFAP (astrocyte) or F4/80 (microglia/macrophage)-associated immunofluorescence (green), VSV-associated immunofluorescence (red) and cell nucleusassociated immunofluorescence (blue) in coronal cortical fields from one of the four
animals in each group using a 40 × objective. Arrows indicate astrocytes (top right
panel) or microglia (lower right panel) with colocalized cellular and VSV staining
(yellow). (C) Protein isolates were prepared from density gradient purified mixed glial
cells (Glia) or prepared from astrocytes (GFAP+) or microglia/macrophages (CD11b
+) isolated from infected mouse brains by flow cytometry, and were analyzed for the
presence of the VSV G protein by Western blot analysis. Plus sign represents a positive
control with protein isolates prepared from purified VSV virions. Parallel Coomassiestained gels (shown below Western blots and indicated as “total protein”) of the
separated cell lysates loaded in the same order as in Western blot show the various
housekeeping protein levels between the samples and demonstrate comparable total
protein levels between different samples.
V.S. Chauhan et al. / Virology 400 (2010) 187–196
shown in Fig. 5C, VSV G protein was readily detectable in glial cell
protein isolates. Furthermore, we have isolated astrocytes (GFAP+)
and microglia/macrophages (CD11b+) from mixed glial preparations from VSV infected mice by flow cytometry and have
confirmed the presence of VSV G protein in these cells by Western
blot analysis (Fig. 5C).
Discussion
The order Mononegavirales consists of four families (Rhabdoviridae,
Paramyxoviridae, Filoviridae and Bornaviridae), which include many
lethal human pathogens (e.g. rabies, Ebola, and Hendra viruses);
highly prevalent human pathogens, such as the respiratory syncytial
and parainfluenza viruses; potential ethological agents of some
neurobehavioral abnormalities and psychiatric disorders in humans
(Borna disease virus); as well as viruses with a major economic impact
on the poultry and cattle industries (e.g. Newcastle disease virus and
rinderpest virus) (Ascenzi et al., 2008; Lamb and Parks, 2007; Lyles and
Rupprecht, 2007; Planz et al., 2009). Some members of this order, such
as rabies and Newcastle disease virus, have the ability to circumvent
the blood–brain barrier to establish CNS infection resulting in a severe
and often fatal inflammation of brain tissue (Leung et al., 2007; Seal et
al., 2000). VSV has the ability to infect neurons and generate acute
encephalitis in mice in a manner that resembles rabies (Huneycutt et
al., 1993) and has therefore proved to be a useful model for rabies and
other nonsegmented negative-sense RNA viruses that cause CNS
infections. Like rabies virus, VSV-associated CNS inflammation is rapid,
occurring within days of intranasal inoculation (Huneycutt et al.,
1993). This rapid onset is indicative of an innate immune response and
is likely to involve the activation of resident CNS cells that have
immune functions. Such a hypothesis is supported by the observation
that VSV-associated encephalitis is unaltered in athymic mice
suggesting that this inflammatory immune response is T-cell independent (Frei et al., 1989). Furthermore, several studies have indicated
that resident CNS cells exhibit immune functions in situ following VSV
infection (Bi et al., 1995; Christian et al., 1996; Huneycutt et al., 1993).
There is growing appreciation that microglia and astrocytes play
an important role in the generation of protective immune responses
or progressive inflammatory damage following CNS infection. These
cells are activated by a variety of microbial pathogens including
viruses and assume immune effector functions such as the production
of inflammatory mediators and the expression of MHC class II
molecules (Bi et al., 1995; Rasley et al., 2004a; Taylor et al., 2003).
Previous study demonstrated that microglia and astrocytes respond to
VSV infection by proliferation, production of iNOS, and the induction
of cell surface MHC class II molecule expression (Bi et al., 1995). Such
responses are likely to set the stage for subsequent inflammatory
damage (Schwartz et al., 1999). While it is possible that these glial
cells can directly recognize viral motifs via cell surface pattern
recognition receptors, or may respond to inflammatory mediators or
other signals released from infected neurons following lytic damage,
the mechanisms underlying activation of resident CNS cells by VSV
have not been determined.
In the present study, we demonstrate that VSV can infect,
replicate and produce new virus particles in isolated cultures of
primary murine microglia and astrocytes following in vitro viral
challenge. In addition, we have shown that VSV products can be
detected in glial cells isolated from the brains of mice inoculated
with VSV intranasally. Furthermore, we have employed dual
immunofluorescence confocal microscopy of brain tissue sections
and flow cytometric analysis of acutely isolated glial cells to
demonstrate coincident expression of VSV products and glial cell
type-specific markers in situ. Finally, we have demonstrated that
VSV replication in microglia or astrocytes precipitates inflammatory
cytokine production by these resident CNS cell types. These findings
are consistent with the previously documented in situ activation of
193
glial cell following in vivo VSV infection (Bi et al., 1995; Christian et
al., 1996). The relevancy of these findings to rabies CNS infection is
supported by the ability of rabies virus to infect cultured microglia
and astrocytes (Ray et al., 1997). Interestingly, we have shown that
the susceptibility of murine microglia and astrocytes to infection is
not restricted to rhabdoviruses as these cells can be infected with,
and support the replication of SeV, a paramyxovirus (Lundh et al.,
1987). Furthermore, we show that glial cells respond to SeV
infection by inflammatory cytokine production, albeit with smaller
responses to those seen with VSV. As such, these data suggest that
glial cells have a broad susceptibility to nonsegmented negativesense RNA viruses and can respond to members of this order by
changes in the production of immune molecules that can promote
damaging CNS inflammation.
Importantly, we have also demonstrated that viral replication is
required to elicit robust immune responses by microglia and
astrocytes. We show that heat inactivated VSV-wt elicits glial
immune responses that are an order of magnitude lower than that
induced by infectious viral particles. These results suggest that
VSV-associated inflammatory responses are not predominantly
mediated by cell surface and/or endosomal pattern recognition
receptors such as TLR3 or TLR7. Such a hypothesis is further
supported by our experiments employing the host-range VSV HR11 mutant (Grdzelishvili et al., 2005), which is replication impaired
in glial cells and fails to elicit marked inflammatory cytokine
production by microglia or astrocytes. This result indicates that
active viral replication is a critical requirement for the generation
of glial immune responses to VSV. While this effect cannot be
adequately explained by the perception of viral particles by TLRs
present on glial cells (Konat et al., 2006), the recent demonstration
that microglia and astrocytes express members of the newly
described RLR family may provide a mechanism underlying the
replication-dependent nature of these responses (Furr et al., 2008).
RLRs are present in the cytosol of many cell types and have been
shown to function as intracellular sensors for replicative viral RNA
motifs (Meylan et al., 2006; Takeuchi and Akira, 2008; Takeuchi
and Akira, 2009). Interestingly, recent studies suggest that the RLR,
RIG-I, demonstrates specificity for VSV and SeV RNA (reviewed in
Yoneyama and Fujita, 2009) and we have demonstrated that both
microglia and astrocytes constitutively express this intracellular
receptor (Furr et al., 2008). Furthermore, we have shown that such
expression is rapidly elevated in murine glial cells following
infection with either VSV or SeV (Furr et al., 2008). As such, the
replication-dependent nature of microglia and astrocyte responses
to VSV described in the present study suggests the possible
importance of RIG-I in the generation of inflammatory cytokine
production by glial cells.
Taken in concert, these studies support a hypothetic scenario in
which VSV and possibly other neurotropic nonsegmented negativesense RNA viruses can invade the CNS and infect both neuronal and
non-neuronal cell types, including microglia and astrocytes. Importantly, the generation of replicative viral RNA can subsequently be
perceived by intracellular pattern recognition receptors such as RIGI, known to be expressed by both astrocytes and microglia. The
resulting activation of RIG-I-dependent signaling pathways could
then initiate glial cell immune functions, precipitating virus
mediated lethal inflammation.
Materials and methods
Viruses
Fig. 1 shows genome organization of the four recombinant (r)
viruses used in this study. Recombinant VSV-wt (Indiana serotype),
and its derivative VSV rHR1-1 with a single amino acid substitution
D1671V in the L protein, have been previously described
194
V.S. Chauhan et al. / Virology 400 (2010) 187–196
(Grdzelishvili et al., 2005). To generate heat-inactivated VSV-wt,
infectious virus was heated at 65 °C for 15 min. This treatment was
determined empirically to eliminate virus infectivity as determined by
plaque assay using BHK-21 cells. VSV-GFP is a VSV-wt (Indiana
serotype) encoding GFP as an extra gene between G and L genes (Das
et al., 2006) and was kindly provided by Dr. Asit K. Pattnaik
(University of Nebraska). Recombinant SeV-wt (Fushimi strain)
(Leyrer et al., 1998) and SeV-GFP-Fmut (SeV-GFP) with an enhanced
green fluorescent protein (GFP) upstream of the NP gene (Wiegand et
al., 2007) were kindly provided by Dr. Wolfgang J. Neubert (MaxPlanck-Institute of Biochemistry, Germany). Monolayer cultures of
Vero or BHK-21 cell lines were maintained in Dulbecco's modified
Eagle's medium (DMEM, Cellgro) supplemented with 9% fetal bovine
serum (FBS, Gibco). To produce VSV or SeV, BHK-21 or Vero cells,
respectively, were infected with wt or mutant viruses at a multiplicity
of infection (MOI) of 0.05 plaque-forming units (PFU) per cell in
MegaVir HyQSFM4 serum-free medium (Hyclone) and incubated for
24–48 h at 34 °C. SeV-GFP was grown without acetylated trypsin in
the medium as it has a wt monobasic trypsin-dependent cleavage site
in the F protein mutated to an oligobasic cleavage site allowing F
activation in any cell type through an ubiquitous furin-like protease
(Wiegand et al., 2007). The MOI values for microglia and/or astrocytes
are relative and calculated based on the VSV titer determined on BHK21 cells or SeV titer on Vero cells supporting robust virus replication.
Therefore, MOIBHK and MOIVERO abbreviations are used throughout
this paper for VSV and SeV, respectively, and it is likely that the actual
MOI values for microglia and/or astrocytes were significantly lower
than indicated.
Isolation of primary murine microglia
Murine neonatal brain microglia were isolated and cultured as
described previously (Harry et al., 1998; Rasley et al., 2002a; Rasley
et al., 2002b; Sterka et al., 2006). Briefly, six to eight neonatal
C57BL/6 mouse brains per preparation were dissected free of
meninges and large blood vessels and finely minced with sterile
surgical scissors. The minced tissue was then forced through a wire
screen and briefly incubated with 0.01% trypsin–0.005% EDTA for
5 min. The cell suspension was then washed and this mixed glial
culture was maintained in RPMI 1640 containing 10% FBS and
gentamicin for 2 weeks. A microglia culture was obtained from this
mixed glial culture by shaking flasks for 2 h at 200 rpm in an orbital
shaker and allowing the transferred dislodged cells to adhere to
new tissue culture vessels for 1 h. The media were medium was
then removed and fresh RPMI 1640 with 10% FBS and 20%
conditioned medium from LADMAC (ATCC number CRL-2420)
cells as a source of M-CSF-1 was added to maintain microglia
cultures for 1 week, as described previously (O'Keefe et al., 2001).
Cells isolated in this manner were N95% authentic microglia as
assessed by their characteristic morphology and by the expression
of CD11b and F4/80 as determined by immunofluorescent microscopy (Rasley et al., 2002a).
Isolation of primary murine astrocytes
Brain astrocytes were isolated and cultured as described
previously (Bowman et al., 2003; Rasley et al., 2004a; Rasley et
al., 2006; Sterka et al., 2006). Astrocyte cultures were obtained from
the mixed glial cultures by shaking flasks to dislodge weakly
adherent microglia, and these cells were discarded cultured
separately as described above. The remaining astrocytes were
mechanically dissociated, transferred to new tissue culture vessels,
and cultured for 1 week prior to use. Cells isolated in this manner
were demonstrated to be greater than 97% authentic astrocytes due
to their characteristic morphology and the presence of the astrocyte
marker, glial fibrillary acidic protein (GFAP), and the absence of the
microglial cell surface marker, CD11b as determined by confocal
microscopy (Bowman et al., 2003).
In vitro viral infection
Isolated primary microglia and astrocytes were infected with VSV,
VSV-GFP, SeV, or SeV-GFP as previously described (Furr et al., 2008) at
MOIBHK (for VSV) or MOIVERO (for SeV) of 1 PFU per cell and the virus
was allowed to adsorb for 1 h. Non-adherent viral particles were
removed by washing with PBS followed by the addition of RPMI 1640
media supplemented with 10% FBS and 20% LADMAC (for microglia)
or 10% FBS (for astrocytes). Cultures were maintained for 6, 12, 24, or
48 h prior to collection of culture supernatants, preparation of wholecell protein isolates, or preparation for confocal microscopic analysis.
Fluorescence microscopy
Fluorescence photomicrographs of cultured microglia and astrocytes infected in vitro with VSV-GFP or SeV-GFP were taken using a
Laser Scanning Microscope 710 (Carl Zeiss, Thornwood, NY) with a
10 × objective and Zen2008 software. Cell nuclei were visualized with
Hoechst 33342 and cell membranes were visualized with AlexaFluor594-labeled wheat germ agglutinin (Invitrogen, Carlsbad, CA).
Western blot analysis
Western blot analyses for the presence of GFP or viral proteins in
microglia, astrocytes, and whole brain samples were performed as
described previously by our laboratory (Bowman et al., 2003; Rasley
et al., 2006). Protein samples were obtained from microglia,
astrocytes, and brain tissue samples in a buffer containing 125 mM
Tris base, 20% glycerol, 2% SDS, 1% bromophenol blue and 2% 2mercaptoethanol. Samples were separated using 10% SDS PAGE and
transferred to Immobilon-P membranes (Millipore, Bedford, MA).
Membranes were blocked for 18 h with 5% skimmed milk at 4 °C. After
incubation with either a rabbit polyclonal antibody against VSV
(Grdzelishvili et al., 2005), a purified goat polyclonal antibody
directed against VSV G protein (Bethyl Laboratories Inc., Montgomery,
TX), or a mouse monoclonal antibody against GFP (Thermo Scientific
Affinity BioReagents, Freemont, CA), for 24 h at 4 °C, blots were
washed and incubated in the presence of an HRP-conjugated donkey
anti-rabbit antibody (Cell Signaling, Danvers, MA), an HRP-conjugated goat anti-mouse IgG antibody (Sigma-Aldrich, St. Louis, MO), or a
HRP-conjugated donkey anti-goat antibody (R&D Systems, Minneapolis, MN). Bound enzyme was detected with the Super Signal system
(Thermo Scientific, Rockford, IL).
Northern blot analysis
Probes were synthesized by in vitro run off transcription in the
presence of [32P]CTP using the MAXIscript T7 kit (Ambion).
pBluSKP-VSV-N (Stillman et al., 1995) digested with HincII and
pGEM3-SeV-NP (Leyrer et al., 1998) digested with PflMI were used
as the in vitro transcription templates to generate riboprobes
complementary to the VSV and SeV genomes, respectively. These
VSV and SeV ribiprobes were found to nonspecifically recognize N
mRNA of the corresponding viruses. Total RNA was extracted from
the collected cell pellets using the Quick-RNA MiniPrep kit (Zymo
Research). For each sample, 1 μg of total RNA was separated on a
1.2% agarose-formaldehyde gel, transferred to a nylon membrane
and incubated with the appropriate probe overnight at 60 °C.
Autoradiographic images of the membranes were obtained using a
GE Healthcare Typhoon Phosphorimager, and RNA bands for viral
genomic RNA, viral N mRNA and cellular rRNA (nonspecifically
recognized by antigenomic riboprobe) were quantitated using
ImageQuant software (Molecular Dynamics).
V.S. Chauhan et al. / Virology 400 (2010) 187–196
Quantification of IL-6, TNF-α, and IL-10 secretion
Specific capture ELISAs were performed to quantify IL-6, TNF-α,
and IL-10 secretion by glial cells using commercially available ELISA
kits (R&D Systems, Minneapolis, MN) as described previously by our
laboratory (Anguita et al., 2001; Chauhan et al., 2008; Rasley et al.,
2004b; Rasley et al., 2006). Commercially available ELISA kits were
used to measure TNF-α (R&D Systems, Minneapolis, MN) and IL-10
(BD Pharmingen, San Diego, CA) secretion. IL-6 secretion was
measured using a rat anti-mouse IL-6 capture antibody (Clone MP520F3) and a biotinylated rat anti-mouse IL-6 detection antibody
(Clone MP5-C2311) (BD Pharmingen, San Diego, CA). Briefly, 96well ELISA plates (ICN Pharmaceuticals, Costa Mesa, CA) were
coated with the capture antibody (2 mg/ml) overnight at 4 °C. After
blocking with PBS plus 10% FBS for 2 h at room temperature,
samples were applied and incubated for 1 h at 37 °C. The
biotinylated detection antibody (1 mg/ml) was added after washing
the plates with PBS plus 0.5% v/v Tween 20 (PBS/Tween 20). HRPconjugated streptavidin was then added prior to addition of
tetramethylbenzidine substrate (Kirkegaard and Perry Laboratories,
Gaithersburg, MD) and stop solution (25% v/v sulfuric acid in
distilled water). Quantification of cytokine levels was achieved by
comparison with standard concentrations of recombinant mouse IL6, IL-10, and TNF-α (R&D Systems, Minneapolis, MN).
In vivo VSV infection
VSV-wt was administered to 3- to 4-week-old male wt C57BL/6
mice (Jackson Laboratories) via intranasal (i.n.) infection essentially
as previously described by our laboratory (Furr et al., 2008; GasperSmith et al., 2006). Anesthetized animals were untreated or received i.
n. administration of VSV (4 × 107 PFU) in PBS (final volume of 20 μl).
Prior to sacrifice, the severity of VSV-associated disease was scored
5 days p.i. for disease severity as previously described by our
laboratory (Rettew et al., 2008). A score of 1 was given to mice with
percolated fur but no detectable behavioral differences compared to
untreated mice, a score of 2 was given to mice with percolated fur and
a huddle reflex but were still active, a score of 3 was given to mice that
were less active and were relatively passive when handled, a score 4
was assigned to inactive mice that exhibited only limited response
when handled, and a score of 5 was applied to moribund mice. In
parallel experiment, animals were euthanized at 3 days p.i. and
protein isolates were prepared from whole brain tissue homogenates
or mixed glial cells were acutely isolated from infected and uninfected
animals as described below (Chauhan et al., 2008). All studies were
performed in accordance with relevant federal guidelines and
institutional policies regarding the use of animals for research
purposes.
195
using an R-phycoerythrin conjugated monoclonal anti-mouse CD11b
antibody (BD Biosciences, clone M1-70) and an AlexaFluor488conjugated monoclonal anti-mouse GFAP antibody (Invitrogen,
Eugene, OR, clone 131-17719). Protein isolates were prepared from
these purified cells and analyzed for the presence of VSV products by
Western blotting.
Immunofluorescent analyses of murine brain sections
Infected and non-infected mice were euthanized 3 days p.i. and the
brains were perfused with Prefer fixative (Anatech, Battle Creek, MI)
via intra-atrial injection. Samples were embedded in paraffin and
sectioned serially (5–7 μm). Briefly, paraffin embedded sections were
heated and rehydrated with decreasing concentrations of ethyl
alcohol prior to rinsing in distilled water. Astrocytes or microglia/
macrophages were identified in deparaffinized coronal sections
blocked with goat serum (Zymed Laboratories, San Francisco, CA)
according to GFAP and F4/80 expression, respectively. GFAP was
detected using a monoclonal mouse antibody directed against murine
GFAP (Invitrogen) and an AlexaFluor488 conjugated chicken antimouse secondary antibody (Invitrogen). F4/80 was detected using a
monoclonal rat antibody directed against murine F4/80 (Biolegend,
San Diego, CA) and a FITC-conjugated goat anti-rat secondary
antibody (Invitrogen). Sections were co-stained with a rabbit
polyclonal antibody against VSV and an AlexaFluor594-conjugated
goat anti-rabbit secondary antibody (Invitrogen), and cell nuclei were
visualized with DAPI (Invitrogen). GFAP, F4/80, VSV and DAPI
staining was assessed in multiple microscopy fields (40 × objective)
of the same cortical locations in animals from each experimental
group using the LSM710 confocal microscopy system (Zeiss).
Statistical analysis
All results are presented as the mean ± SEM and were tested
statistically by Student's t test or one-way analysis of variance
(ANOVA) with Tukey's post hoc test, as appropriate, using commercially available software (Sigma Stat; Systat Software, San Jose, CA).
Results were considered to be statistically significant at a probability
of b0.05.
Acknowledgments
We are grateful to Sue Moyer (University of Florida College of
Medicine) for providing VSV and SeV reagents for this project. We
thank Dr. Asit K. Pattnaik (University of Nebraska) and Dr. Wolfgang J.
Neubert (Max-Planck-Institute of Biochemistry, Germany) for providing VSV-GFP and SeV-GFP recombinant viruses, respectively.
Acute isolation and cytometric analysis of CNS cells
References
Mixed CNS cells were acutely isolated from infected and
uninfected animals using a protocol modified from (Campanella et
al., 2002) as previously described (Chauhan et al., 2008). Briefly,
brains were rapidly removed and mechanically disrupted in a glass
homogenizer, washed, and resuspended in PBS/30% Percoll (Fluka,
Sigma Aldrich, St. Louis, MO) solution. This was overlaid on a gradient
containing 37% and 70% Percoll solutions and centrifuged at 600 × g
for 20 min at room temperature. Glial cells were then collected from
the interface and washed with PBS. For VSV-wt infected animals,
protein isolates were prepared from the purified cells and analyzed
for the presence of VSV products by Western blotting using a
polyclonal antibody directed against this virus or a purified goat
polyclonal antibody directed against VSV G protein (Bethyl Laboratories Inc.). In addition, in some experiments astrocytes and microglia
were isolated from the mixed glial preparation by flow cytometry
Anguita, J., Thomas, V., Samanta, S., Persinski, R., Hernanz, C., Barthold, S.W., Fikrig, E.,
2001. Borrelia burgdorferi-induced inflammation facilitates spirochete adaptation
and variable major protein-like sequence locus recombination. J. Immunol. 167,
3383–3390.
Ascenzi, P., Bocedi, A., Heptonstall, J., Capobianchi, M.R., Di Caro, A., Mastrangelo, E.,
Bolognesi, M., Ippolito, G., 2008. Ebolavirus and Marburgvirus: insight the
Filoviridae family. Mol. Aspects Med. 29, 151–185.
Bi, Z., Barna, M., Komatsu, T., Reiss, C.S., 1995. Vesicular stomatitis virus infection of the
central nervous system activates both innate and acquired immunity. J. Virol. 69,
6466–6472.
Bowman, C.C., Rasley, A., Tranguch, S.L., Marriott, I., 2003. Cultured astrocytes express
toll-like receptors for bacterial products. Glia 43, 281–291.
Campanella, M., Sciorati, C., Tarozzo, G., Beltramo, M., 2002. Flow cytometric analysis of
inflammatory cells in ischemic rat brain. Stroke 33, 586–592.
Chauhan, V.S., Marriott, I., 2007. Bacterial infections of the central nervous system: a
critical role for resident glial cells in the initiation and progression of inflammation.
Curr. Immunol. Rev. 3, 133–143.
Chauhan, V.S., Sterka Jr., D.G., Furr, S.R., Young, A.B., Marriott, I., 2008. NOD2 plays an
important role in the inflammatory responses of microglia and astrocytes to
bacterial CNS pathogens. Glia 57, 414–423.
196
V.S. Chauhan et al. / Virology 400 (2010) 187–196
Christian, A.Y., Barna, M., Bi, Z., Reiss, C.S., 1996. Host immune response to vesicular
stomatitis virus infection of the central nervous system in C57BL/6 mice. Viral
Immunol. 9, 195–205.
Das, S.C., Nayak, D., Zhou, Y., Pattnaik, A.K., 2006. Visualization of intracellular transport
of vesicular stomatitis virus nucleocapsids in living cells. J. Virol. 80, 6368–6377.
Frei, K., Malipiero, U.V., Leist, T.P., Zinkernagel, R.M., Schwab, M.E., Fontana, A., 1989. On
the cellular source and function of interleukin 6 produced in the central nervous
system in viral diseases. Eur. J. Immunol. 19, 689–694.
Furr, S.R., Chauhan, V.S., Sterka Jr., D., Grdzelishvili, V., Marriott, I., 2008. Characterization of retinoic acid-inducible gene-I expression in primary murine glia following
exposure to vesicular stomatitis virus. J. Neurovirol. 7, 1–11.
Gasper-Smith, N., Marriott, I., Bost, K.L., 2006. Murine gamma-herpesvirus 68 limits
naturally occurring CD4+CD25+T regulatory cell activity following infection.
J. Immunol. 177, 4670–4678.
Grdzelishvili, V.Z., Smallwood, S., Tower, D., Hall, R.L., Hunt, D.M., Moyer, S.A., 2005. A
single amino acid change in the L-polymerase protein of vesicular stomatitis virus
completely abolishes viral mRNA cap methylation. J. Virol. 79, 7327–7337.
Harry, G.J., Billingsley, M., Bruinink, A., Campbell, I.L., Classen, W., Dorman, D.C., Galli, C.,
Ray, D., Smith, R.A., Tilson, H.A., 1998. In vitro techniques for the assessment of
neurotoxicity. Environ. Health Perspect. 106 (Suppl. 1), 131–158.
Hercyk, N., Horikami, S.M., Moyer, S.A., 1988. The vesicular stomatitis virus L protein
possesses the mRNA methyltransferase activities. Virology 163, 222–225.
Horikami, S.M., Moyer, S.A., 1982. Host range mutants of vesicular stomatitis virus
defective in in vitro RNA methylation. Proc. Natl. Acad. Sci. U.S.A. 79, 7694–7698.
Horikami, S.M., Curran, J., Kolakofsky, D., Moyer, S.A., 1992. Complexes of Sendai virus
NP-P and P-L proteins are required for defective interfering particle genome
replication in vitro. J. Virol. 66, 4901–4908.
Huneycutt, B.S., Bi, Z., Aoki, C.J., Reiss, C.S., 1993. Central neuropathogenesis of vesicular
stomatitis virus infection of immunodeficient mice. J. Virol. 67, 6698–6706.
Konat, G.W., Kielian, T., Marriott, I., 2006. The role of Toll-like receptors in CNS response
to microbial challenge. J. Neurochem. 99, 1–12.
Lamb, R.A., Parks, G.D., 2007. Paramyxoviridae: the viruses and their replication, In:
Knipe, D.M., Howley, P.M. (Eds.), Fields Virology, 5th ed. Lippincott Williams and
Wilkins, Philadelphia, pp. 1449–1496.
Leung, A.K., Davies, H.D., Hon, K.L., 2007. Rabies: epidemiology, pathogenesis, and
prophylaxis. Adv. Ther. 24, 1340–1347.
Leyrer, S., Neubert, W.J., Sedlmeier, R., 1998. Rapid and efficient recovery of Sendai virus
from cDNA: factors influencing recombinant virus rescue. J. Virol. Methods 75,
47–58.
Lundh, B., 1990. Spread of vesicular stomatitis virus along the visual pathways after
retinal infection in the mouse. Acta Neuropathol. 79, 395–401.
Lundh, B., Kristensson, K., Norrby, E., 1987. Selective infections of olfactory and
respiratory epithelium by vesicular stomatitis and Sendai viruses. Neuropathol.
Appl. Neurobiol. 13, 111–122.
Lyles, D.S., Rupprecht, C.E., 2007. Rhabdoviridae, In: Knipe, D.M., Howley, P.M. (Eds.),
Fields Virology, 5th ed. Lippincott Williams and Wilkins, Philadelphia, pp.
1363–1408.
Meylan, E., Tschopp, J., Karin, M., 2006. Intracellular pattern recognition receptors in the
host response. Nature 442, 39–44.
Miyoshi, K., Harter, D.H., Hsu, K.C., 1971. Neuropathological and immunofluorescence
studies of experimental vesicular stomatitis virus encephalitis in mice. J. Neuropathol.
Exp. Neurol. 30, 266–277.
Nansen, A., Marker, O., Bartholdy, C., Thomsen, A.R., 2000. CCR2+and CCR5+CD8+T
cells increase during viral infection and migrate to sites of infection. Eur. J.
Immunol. 30, 1797–1806.
Nuovo, G.J., Defaria, D.L., Chanona-Vilchi, J.G., Zhang, Y., 2005. Molecular detection of rabies
encephalitis and correlation with cytokine expression. Mod. Pathol. 18, 62–67.
O'Keefe, G.M., Nguyen, V.T., Ping Tang, L.L., Benveniste, E.N., 2001. IFN-gamma
regulation of class II transactivator promoter IV in macrophages and microglia:
involvement of the suppressors of cytokine signaling-1 protein. J. Immunol. 166,
2260–2269.
Planz, O., Pleschka, S., Wolff, T., 2009. Borna disease virus: a unique pathogen and its
interaction with intracellular signalling pathways. Cell. Microbiol. 11, 872–879.
Rasley, A., Anguita, J., Marriott, I., 2002a. Borrelia burgdorferi induces inflammatory
mediator production by murine microglia. J. Neuroimmunol. 130, 22–31.
Rasley, A., Bost, K.L., Olson, J.K., Miller, S.D., Marriott, I., 2002b. Expression of functional
NK-1 receptors in murine microglia. Glia 37, 258–267.
Rasley, A., Bost, K.L., Marriott, I., 2004a. Murine gammaherpesvirus-68 elicits robust
levels of interleukin-12 p40, but not interleukin-12 p70 production, by murine
microglia and astrocytes. J. Neurovirol. 10, 171–180.
Rasley, A., Marriott, I., Halberstadt, C.R., Bost, K.L., Anguita, J., 2004b. Substance P
augments Borrelia burgdorferi-induced prostaglandin E2 production by murine
microglia. J. Immunol. 172, 5707–5713.
Rasley, A., Tranguch, S.L., Rati, D.M., Marriott, I., 2006. Murine glia express the
immunosuppressive cytokine, interleukin-10, following exposure to Borrelia
burgdorferi or Neisseria meningitidis. Glia 53, 583–592.
Ray, N.B., Power, C., Lynch, W.P., Ewalt, L.C., Lodmell, D.L., 1997. Rabies viruses infect
primary cultures of murine, feline, and human microglia and astrocytes. Arch. Virol.
142, 1011–1019.
Rempel, J.D., Quina, L.A., Blakely-Gonzales, P.K., Buchmeier, M.J., Gruol, D.L., 2005. Viral
induction of central nervous system innate immune responses. J. Virol. 79, 4369–4381.
Rettew, J.A., Huet-Hudson, Y.M., Marriott, I., 2008. Testosterone reduces macrophage
expression in the mouse of toll-like receptor 4, a trigger for inflammation and
innate immunity. Biol. Reprod. 78, 432–437.
Schwartz, M., Moalem, G., Leibowitz-Amit, R., Cohen, I.R., 1999. Innate and adaptive
immune responses can be beneficial for CNS repair. Trends Neurosci. 22, 295–299.
Seal, B.S., King, D.J., Sellers, H.S., 2000. The avian response to Newcastle disease virus.
Dev. Comp. Immunol. 24, 257–268.
Sterka Jr., D., Rati, D.M., Marriott, I., 2006. Functional expression of NOD2, a novel
pattern recognition receptor for bacterial motifs, in primary murine astrocytes. Glia
53, 322–330.
Stillman, E.A., Rose, J.K., Whitt, M.A., 1995. Replication and amplification of novel
vesicular stomatitis virus minigenomes encoding viral structural proteins. J. Virol.
69, 2946–2953.
Takeuchi, O., Akira, S., 2008. MDA5/RIG-I and virus recognition. Curr. Opin. Immunol.
20, 17–22.
Takeuchi, O., Akira, S., 2009. Innate immunity to virus infection. Immunol. Rev. 227, 75–86.
Taylor, W.R., Rasley, A., Bost, K.L., Marriott, I., 2003. Murine gammaherpesvirus-68
infects microglia and induces high levels of pro-inflammatory cytokine production.
J. Neuroimmunol. 136, 75–83.
Wiegand, M.A., Bossow, S., Schlecht, S., Neubert, W.J., 2007. De novo synthesis of N and
P proteins as a key step in Sendai virus gene expression. J. Virol. 81, 13835–13844.
Yoneyama, M., Fujita, T., 2009. RNA recognition and signal transduction by RIG-I-like
receptors. Immunol. Rev. 227, 54–65.