You are on page 1of 6

Viral-induced encephalitis initiates distinct and

functional CD103+ CD11b+ brain dendritic cell


populations within the olfactory bulb
Paul M. DAgostinoa, Changsoo Kwaka, Haley A. Vecchiarellia, Judit Gal Totha, James M. Millera, Zahrah Masheeba,
Bruce S. McEwena,1, and Karen Bullocha,b,1
a
Laboratory of Neuroendocrinology and bLaboratory of Cellular Physiology and Immunology, The Rockefeller University, New York, NY 10065

Contributed by Bruce S. McEwen, March 6, 2012 (sent for review November 21, 2011)

Dendritic cells (DC) are antigen-presenting cells found in both RNA genome, and high sensitivity to IFN-elicited immune re-
lymphoid and nonlymphoid organs, including the brain (bDC) of sponses (23). Many VSV attributes, including host range, natural
Cd11c/eyfp transgenic C57BL/6 mice. Using an intranasal vesicular reservoirs, endemic regions, and infectious cycle are well known.
stomatitis virus infection, we demonstrated that EYFP+ cells amass One characteristic in particular, the polarized nature of VSV
in areas associated with viral antigens, take on an activated mor- virion budding, is the basis for the intranasal VSV infection
phology, and project their processes into infected neuronal tissue model used hereand by many othersto study virally induced
within the olfactory bulb. These bDC separated into three EYFP+ encephalitis in the mouse (24, 25). VSV is capable of accessing
CD45+ CD11b+ populations, all but one being able to functionally
the CNS via olfactory nerve cells that span the cribriform plate;
this, and its neuro-invasive pathology, make it amenable to
promote both T lymphocyte proliferation and TH1 cytokine pro-
studying proinammatory responses within the brain (26, 27).
duction. One population was shown to emanate from the brain In this work we found that VSV infection of Cd11c/eyfp Tg mice
and a second population was peripherally derived. The third pop- results in anatomical changes in CD11c+ cell distribution and
ulation was of indeterminate origin, being both radiosensitive and morphology relative to regions associated with viral antigen ex-
not replenished by donor bone marrow. Finally, each EYFP+ pop- pression. Flow cytometric analysis reveals the presence of unique
ulation contained CD11b+ CD103+ subpopulations and could be CD45+, CD11b+, and CD11c+ cell populations within the VSV-
distinguished in terms of CD115, Gr-1, and Ly-6C expression, high- infected OB, each with its own distinct phenotype. Functional
lighting mucosal and monocyte-derived DC lineages. analysis of these populations demonstrates the ability of some to
process and present antigen to T lymphocytes on par with splenic
antigen presentation | neuroinammation conventional DC (cDC). Finally, we explore the origin and lineage
of these unique bDC populations within the context of viral in-
fection using radiation chimeras, EdU-labeling, and phenotyping.
D endritic cells (DC) are immune cells specialized in antigen
capture, processing, and presentation to T lymphocytes for
the induction of adaptive immunity (1) or tolerance (2, 3). Since
These data revealed the presence of CD103+ CD11b+ double-
positive cells similar to those found in mucosal DC subsets of the
their initial identication (4) and characterization (5, 6), DC are intestinal lamina propria and the basal lamina of the bronchial
now recognized to represent a heterogeneous group consisting of epithelium (10, 14), as well as a population of radio-sensitive, bone
many distinct subpopulations identiable by a diverse array of marrow-independent cells resembling monocyte-derived DC.
cell-surface markers and functions (7, 8). DC subpopulations are
Results
found in both lymphoid and nonlymphoid tissue, such as the
dermis, intestine, liver, lungs, kidneys, and the heart (912). Cd11c/eyfp-Expressing Cells Respond to Early Stages of Acute VSV
Some examples of this phenotypic heterogeneity include: plas- Infection Within the OB. To investigate how CD11c-expressing
macytoid DC, the primary function of which is the production of cells respond to an infection caused by a pathogens ability to
type I interferons (13); the CD8+ DEC205+ DC in the spleen, circumvent the BBB, we intranasally infected Cd11c/eyfp Tg mice
which are involved in cross-presentation of antigen to cytotoxic T with VSV. Three cohorts of 12 mice received a LD50 of VSV,
lymphocytes; and CD103+ CD11b+ DC in the intestine, which UV-inactivated VSV, or vehicle control. Mice were killed fol-
migrate to draining lymph nodes in a CCR7-dependent manner lowing 24, 48, 72, and 96 h postinfection (hpi), and 60-m OB

IMMUNOLOGY
and promote the proliferation of regulatory T cells (Treg) (1416). sections were processed for immunouorescence staining with an
DC are also found within the brain (bDC) and meninges of the anti-VSV antiserum. Over the rst 24 hpi, we observed VSV
Itgax (Cd11c)/eyfp transgenic (Tg) mouse (17, 18). In the steady antigen within the olfactory nerve layer with no apparent change
state, bDC are found within discrete regions of the CNS (17). in the EYFP+ distribution pattern, relative to naive mice (Fig.
Furthermore, bDC have been shown to respond to physical S1A). By 48 hpi, the VSV infection had progressed through the
trauma and intracranial treatment with the proinammatory olfactory nerve and penetrated the glomerular layer, again with
cytokine IFN- (19, 20). Given the ability of bDC to present little change in EYFP+ cell distribution. However, by 72 hpi,
antigen ex vivo after stimulus with IFN-, and their association EYFP+ cells were found in abundance throughout VSV-infected
with blood brain barrier (BBB) compromised or decient ana- areas and continued to increase after 96 hpi. This accumulation
tomical regions of the CNS [e.g., the olfactory bulb (OB) and of EYFP+ cells was only observed with viable VSV and was not
circumventricular organs], bDC appear to possess the CNS an- reproduced with either UV-inactivated virus or vehicle controls
tigen presenting cell function rst attributed to bone marrow- at any time point postinfection (Fig. S1B). These data indicate
derived perivascular microglia by Hickey and Kimura (21). To
further elucidate in vivo bDC function, we capitalized on a nat-
urally occurring infectious route to the brain, namely CNS access Author contributions: P.M.D. and K.B. designed research; P.M.D., C.K., H.A.V., J.G.T.,
via olfactory nerves within the nasal epithelia. J.M.M., and Z.M. performed research; P.M.D., C.K., H.A.V., B.S.M., and K.B. analyzed data;
Using vesicular stomatitis virus (VSV) to elicit an encephalitic and P.M.D., B.S.M., and K.B. wrote the paper.
state after intranasal administration (22), we sought to better The authors declare no conict of interest.
understand the bDC response to an acute infection via pheno- 1
To whom correspondence may be addressed. E-mail: bulloch@rockefeller.edu or mcewen@
typic and functional characterization. VSV is a member of the mail.rockefeller.edu.
Rhabdoviridae viral family, most notably characterized by its This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
enveloped bullet-shaped virion, negative sense single-stranded 1073/pnas.1203941109/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1203941109 PNAS | April 17, 2012 | vol. 109 | no. 16 | 61756180


that a putative bDC response was solely attributable to an active (P valueP1vsP2 < 0.001) (Fig. 2B) and P3 (P valueP1vsP3 < 0.0001),
VSV infection rather than from irritation caused by inhalation of and P3 had the least number of EYFP+ cells (P valueANOVA <
liquid or innate immune responses within infected neurons 0.0001). In summary, most EYFP+ cells within the VSV-infected
triggered by pathogen-associated molecular patterns. Further- OB at 96 hpi fall within the P1 population.
more, CD11c+ cells that accumulated within these VSV-infected Next, the ability of the three EYFP+ populations to function
regions exhibited an activated morphology signied by stout cell as antigen-presenting cells was examined using a recombinant
bodies with stunted processes (28), unlike the thin, delicately version of VSV (rVSV-OVA) able to transduce ovalbumin
branched processes associated with bDC in unchallenged mice (OVA) expression in the cells it infects (36). rVSV-OVA was
(Fig. S2). Closer examination revealed that CD11c+ cells clus- used to induce the robust increases in EYFP+ cells observed in
tered around VSV-infected neurons within glomeruli (Fig. 1A), wild-type VSV infections, while simultaneously priming those cells
and projected their processes into neuropil-expressing VSV anti- in vivo with OVA. This rVSV-OVA infection model was sub-
gens (Fig. 1B). This phenomenon was reminiscent of the hallmark stantiated by (i) confocal micrographs demonstrating colocaliza-
probing behavior attributed to both brain resident microglia (MG) tion of OVA and VSV antigens within infected glomeruli (Fig.
and peripheral DC subsets (4, 29, 30). 3A), and (ii) ow cytometry analysis that revealed an increase
To correlate the putative bDC response to MG, changes in MG in EYFP+ bDC levels in the OB, which resolved into the three
distribution were analyzed in response to the VSV infection. In populations identied during wild-type VSV infections (Fig. 3B).
previously published work, MG were discriminated from putative Using CD45, CD11b, and EYFP as markers, cells from P1, P2,
bDC in the Cd11c/eyfp Tg mouse, primarily based on EYFP ex- and P3 were then isolated by FACS at 96 hpi from rVSV-OVA
pression in conjunction with either an Iba1+ or F4/80+ phenotype intranasally infected OB, along with MG (identied as CD45int
(17). Using this denition, the vast majority of EYFPneg Iba1+ cells CD11bhi EYFPneg). As controls, splenic cDC (B220neg EYFPhi)
showed no distributive differences between areas proximal and were also isolated from both steady state and rVSV-OVA in-
distal to the infection relative to vehicle control groups (Fig. S3), traperitoneally infected Cd11c/eyfp Tg mice. Sorted P2 and P3
even though EYFPneg Iba1+ displayed an activated morphology bDC, as well as splenic cDC from infected mice, were capable of
near the site of infection. Given the MGs developmental plasticity directly stimulating CD8 OT-I, but not CD4 OT-II, T-cell pro-
(31), we subsequently sought to phenotypically and functionally liferation (Fig. 3C). The magnitude of the observed proliferation
characterize the CD11c+ cell response to verify their role as bDC. was consistent with both the small percentage of OT-I and OT-II
epitopes present within the OVA primary sequence (SIINFEKL
bDC Responding to Acute VSV Infection Separate into Three Pheno- 2% and ISQAVHAAHAEINEAGR 4% of total OVA amino
typically and Functionally Distinct Populations. Given that the BBB acids, respectively) and the low levels of OVA mRNA produced
remains uncompromised during an intranasal VSV infection by the VSV RNA-dependent RNA polymerase, given the genes
until 68 d postinfection (22), we chose to analyze bDC accu- position in the recombinant viral genome (37). P1 and MG from
mulation at 96 hpi to maximize cell numbers while minimizing rVSV-OVA infected OB, as well as splenic cDC from uninfected
peripheral immune contributions. As such, bDC were isolated mice, were unable to stimulate OT T-cell proliferation without
and phenotypically characterized by ow cytometry at 96 hpi in further antigen supplementation. In summary, these data illus-
the VSV-infected OB. After gating for live, singlet, TCRneg, trate ongoing in vivo processing of OVA antigens by P2, P3, and
and CD19neg cells (Fig. S4), EYFP+ cells resolved into three cDC controls, but not P1 and MG, as illustrated by their ability to
distinct CD45+ and CD11b+ populations only prevalent in facilitate OVA specic T-cell proliferation.
infected OB (Fig. 2A). The EYFP+ CD45int CD11bhi (P1) When the EYFP+ populations were supplemented with excess
phenotypic population had a similar CD45 and CD11b pheno- OVA or the appropriate OT peptide signicant proliferation of
type as previously described resident MG populations found in OT T cells was observed, in some cases out-performing splenic
the steady state and inamed brains of wild-type animals (32 cDC populations. However, in all antigen-presentation assays,
35). The two other populations, only found at appreciable levels MG were unable to present OVA, OT-I or OT-II peptide anti-
in the VSV-infected OB, were dened by EYFP+ CD45hi gens to their respective OT T cells (Fig. 3 D and E). Similarly, P1
CD11bhi (P2) and EYFP+ CD45hi CD11bint (P3) phenotypes. P1 cells were unable to process and present OVA or OT-II peptide;
had the greatest number of EYFP+ cells compared with P2 however, they were able to facilitate CD8 OT-I T-cell pro-
liferation with the SIINFEKL peptide, albeit at very low levels.
In stark contrast, both P2 and P3 populations were effectively
able to present supplemented peptide antigens and OVA ex vivo
to OT T cells. In most cases, the P3 population induced a more
robust proliferation than that observed with steady state or
rVSV-OVA cDC controls (Fig. 3 D and E). This higher func-
tional ability most likely arose from greater levels of phenotyp-
ically mature DC found within the intranasally infected OB than
spleens from intraperitoneally infected mice, because the spleen
primarily monitors the blood and is not likely to be the draining
lymphoid organ for infections of the peritoneal cavity.
Finally, cytokine proles were analyzed from proliferating
CD4+ OT-II T cells induced during antigen presentation assays
(Fig. S5). Both P2 and P3 populations promoted a proinam-
matory helper T-cell (TH1) biased CD4 T lymphocyte response
characterized by elevated levels of IL-2, IFN-, and TNF, similar
to both splenic cDC populations. MG and the P1 population,
however, had undetectable levels of both pro- and anti-inam-
matory cytokines, which was consistent with the lack of CD4+
T-cell proliferation elicited by those cells. In summary, P2 and
Fig. 1. bDC surround and probe VSV-infected glomeruli. Representative P3, much like their cDC counterparts, were effectively able to
confocal z-stack analysis of an OB 72 hpi after intranasal VSV infection. (A) A orchestrate both OVA-specic cytotoxic and TH1-mediated
glomerulus labeled with anti-VSV antiserum (red) showed clustering bDC responses after in vitro supplementation with OVA or its pep-
(green). (B) Confocal section visualizing a bDC extending cellular projections tides. Conversely, P1 and MG did not participate in establishing
(arrowheads) into VSV infected glomerular neuropil. Representative images such antiviral related responses, although P1 did show a mild
from three experiments with an n = 3. (Scale bars, 10 m.) ability to induce cytotoxic effector cells.

6176 | www.pnas.org/cgi/doi/10.1073/pnas.1203941109 DAgostino et al.


Fig. 2. EYFP+ bDC within the VSV-infected OB exhibit three
phenotypically distinct populations at 96 hpi. (A) Three dis-
tinct EYFP+, CD45+, and CD11b+ populations. (B) Analysis of
the differences in each CD45+ and CD11b+ population shows
that the P1 population represents the majority of EYFP+ cells;
the P2 and P3 populations were less abundant. The EYFP+ cell
numbers found within the P1 population are nearly double
those in the P2 and P3 populations combined. Data repre-
sentative of three separate experiments with n = 3; error bars
represent SEM. ***P < 0.001

Origin of VSV-Induced bDC Populations. Radiation chimeras were even though all three populations were observed in a control in-
used to ascertain the origin of EYFP+ cell populations that respond fection of Cd11c/eyfp Tg mice (Fig. 4C). Consequently, the source
to acute VSV infection. After verifying chimerism by analyzing for P3 is ambiguous at this point and its absence could be in-
CD45.1+ and CD45.2+ leukocytes in the blood, CD11b+ cells within dicative of (i) a radiosensitive population that is not replenished
the OB were assessed for surface CD45.1+ or CD45.2+ expression. by bone marrow progenitor cells within the time constraints of our
In lethally irradiated Cd45.1 genetically engineered host mice viral infection model (i.e., mice are optimally susceptible to VSV
replenished with Cd11c/eyfp (CD45.2) Tg bone marrow, radio-re- infection between 5 and 7 wk of age, allowing only 4 wk for chi-
sistant cells were identied at 96 hpi that resembled the P1 pop- merism), or (ii) a confound of the irradiation model.
ulation in that they were CD45.1int CD11bhi (Fig. 4A). Fur- Given that the chimera data supports P1 cells as part of the
thermore, a second population was identied that expressed the brain parenchyma, the question still remains as to how these
CD45.2hi CD11bhi donor phenotype and resembled P2. These EYFP+ cells go from very low numbers in the steady-state OB to
observations were veried by EYFP uorescence because the such high levels between 72 and 96 hpi of acute VSV infection.
population resembling P1 was found to be EYFPneg, but the To address this question, EdU-labeled proliferation was assessed
population similar to P2 was EYFP+ (Fig. 4D). These data were in Cd11c/eyfp Tg mice intranasally infected with VSV (LD50) or
also anatomically corroborated in the VSV-infected chimeric OB, vehicle alone. After 96 hpi, there was an 71.7% increase in
because EYFP+ cells were present in low numbers only at the site EdU+ cells found within the VSV-infected OB, relative to HBSS
of acute viral infection (Fig. S6A). These data were recapitulated controls (Fig. S7A). However, none of the three identied
in reciprocally chimeric mice: that is, lethally irradiated Cd11c/eyfp EYFP+ CD45+ CD11b+ populations had incorporated EdU (Fig.
(CD45.2) Tg host mice replenished with Cd45.1 genetically engi- S7B); this was interesting considering P1 comprised the largest
neered bone marrow. In short, the P1 population was CD45.2int proportion of EYFP+ cells responding to the VSV infection.
CD11bhi, radio-resistant and EYFP+, and a second population
resembling P2 originated from CD45.1hi CD11bhi donor bone Lineage of VSV-Induced bDC Populations. In an effort to better
marrow and was EYFPneg (Fig. 4 B and D). These observations understand the lineage associated with all three EYFP+ pop-
were anatomically represented by the high accumulation of EYFP+ ulations, these cells were separately phenotyped in nonirradiated
cells typically seen virally infected, nonirradiated Cd11c/eyfp mice by ow cytometry using cellular markers commonly asso-
(CD45.2) Tg mice (Fig. S6B). Taken together, these data suggest ciated with DC subsets, as well as other immunological cells. As
that P1 are radio-resistant and represent a resident population controls, MG isolated from VSV-infected OB were character-
within the brain, and P2 are derived from bone marrow progenitors. ized along with cDC (TCRneg, CD19neg, NK1.1neg, and EYFPhi)
Unexpectedly, no population resembling P3 was associated from the spleen of VSV-infected mice (Fig. 5A and summarized
with CD45.1+ or CD45.2+ leukocytes in either chimeric model, in Table S1). As expected, all three EYFP+ populations were

Fig. 3. Recombinant VSV-OVA induced bDC process


and present antigen to OVA-specic T lymphocytes.
(A) Representative confocal images of anti-OVA (red)
and EYFP (green) from OB sections from mice in-

IMMUNOLOGY
tranasally infected with rVSV-OVA at 96 hpi, resulting
in OVA and VSV antigen (blue) colocalization within
glomeruli. (Scale bars, 50 m.) (B) rVSV-OVA infection
elicited EYFP+ populations analogous to wild-type
VSV. (C) P2 and P3 processed in vivo antigens during
a rVSV-OVA intranasal infection and drove ex vivo
proliferation of OVA-restricted CD8+, but not CD4+ T
cells. Bar graphs represent the average number of
proliferating T cells, as determined by a shift in 5-(and
6)-carboxyuorescein diacetate succinimidyl ester
intensity. (D) All three bDC populations facilitated
OVA-restricted CD8+ T-cell proliferation after ex
vivo supplementation with OT-I peptide; however P1
was ineffective at presenting OVA. Representative
histograms from OT-I peptide treated DC populations
plated 1:30 with T cells. (E) P2 and P3 bDC effectively
presented OVA antigens to promote OVA-restricted
CD4+ T-cell proliferation, but P1 was ineffective. His-
tograms from OT-II peptide-treated DC populations
plated 1:30, and OVA treated plated 1:5. Error bars
represent SEM; 612 replicates per treatment group.
*P valuevs. T Cell Only < 0.05, **P valuevs. T Cell Only < 0.01,
***P valuevs. T Cell Only < 0.001.

DAgostino et al. PNAS | April 17, 2012 | vol. 109 | no. 16 | 6177
positive for CD11c and MHC I, although P1 cells expressed the Discussion
lowest levels of CD11c relative to all other EYFP+ populations. Using intranasal VSV infection we show that bDC, normally
All three EYFP+ populations also contained CD103+ cells, with positioned near sites associated with olfactory nerve entry into
P3 possessing the least; MG were mostly CD103low cells. Addi- the steady-state OB, amass in great numbers and surround infec-
tionally, all populations were found mostly negative for CD8 ted neuronal tissue. Furthermore, we were able to discriminate
and DEC-205, with the exception being a very small P3 sub- three phenotypically and functionally distinct EYFP+ cell pop-
population, and all populations were found negative for CD4 ulations from the phenotype commonly associated with MG, two
and Siglec H. In summary, the three EYFP+ populations were of which with potentially different lineage associations. Because
distinguishable from MG based on CD11c and CD103 expres- the pathology associated with intranasal VSV, in terms of viral
sion, and from cDC subsets in the spleen associated with cross- progression and leukocyte recruitment, is well established (25
presentation (i.e., CD8+ and DEC-205+). 27, 40), we chose to examine a time point (96 hpi) where the
Further differences between EYFP+ populations were evident greatest accumulation of bDC coincided with the ability to ana-
when examining MHC II and the expression patterns of the lyze their early contributions toward afferent immune responses
costimulatory molecules. First, P1 cells were found to be MHC before breakdown of the BBB. Our data not only depicts an in-
crease of bDC within VSV-infected areas, but closer examination
IIneg, and a majority of the P2 and P3 populations contained
revealed these bDC often projected cellular processes directly
MHC II+ cells. Second, P1 and MG expressed low levels of the into infected tissue. Conversely, EYFPneg Iba1+ MG, although
costimulatory molecules CD40 and CD80, and moderate levels morphologically activated, did not redistribute in response to the
of CD86, but these markers were found at relatively higher levels acute viral infection. These ndings substantiate our hypothesis
within P2 and P3. These results were consistent with P2 and P3 that EYFP+ bDC occupy vulnerable CNS access points (in this
representing mature DC subsets capable of antigen presentation. case they exist within tissue that have olfactory nerve projections
In addition to differences in the expression of DC maturation from the nasal epithelium) to act as rst responders capable of
markers, there were noted differences in the markers used to orchestrating immunological responses. The nature of those im-
classify DC lineage. Both P1 and P2 populations contained mune responses was the focus of our further investigation.
macrophage colony-stimulating factor receptor (CD115)-positive Phenotypic characterization demonstrated that VSV-induced
and F4/80+ cells, but P3 cells were both CD115neg and F4/80low. EYFP+ cells resolved into three distinct populations based on
In terms of Ly-6C surface expression, unlike the P1 population, CD45 and CD11b expression not observed in the steady state. One
which was Ly-6Cneg, all of P2 and a majority of P3 cells were of these populations, described as a radio-resistant resident
identied as Ly-6C+. Interestingly, the P3 population also con- CD45int CD11bhi (P1) population, was similar in phenotype to MG
tained cells positive for NK1.1, CD49b, CD122, and CD45R, (34, 35), except that it contains a CD103+ subpopulation, a phe-
phenotypes reminiscent of NK cells and a unique IFN-producing notype resembling migratory DC (10, 11). P1, however, differed
killer DC found within both lymph nodes and spleen (38, 39). from migratory DC cell types based on surface MHC II and cos-
Finally, using a clone capable of discriminating Ly-6G (Gr-1) timulatory molecule expression. Furthermore, this population
from Ly-6C, P2 was the only population found to be Gr-1+. failed to incorporate EdU during an acute VSV infection, all of
Subsequent phenotypic analysis using CD103, MHC II, Gr-1, which strongly suggests P1 may differentiate from the CD103neg
and Ly-6C, as depicted in Fig. 5 B and C, revealed the three CD11clow MG population found within the VSV-infected OB.
P1 and MG populations were also functionally distinguishable
EYFP+ cell populations could be further divided into CD103+ from one another. Unlike MG, P1 cells were able to weakly fa-
and CD103neg subpopulations. P1 was composed of 45.3% cilitate CD8+ OT-I T-cell receptor-restricted T lymphocyte
7.5% CD103+ cells that were MHC II, Gr-1, and Ly-6Cnega- proliferation. Given these observations, it would be interesting
tive. However, unlike P1 EYFP+ cells, a majority of P2 was to assess the phenotypic and functional characteristics of P1 during
composed of CD103+ cells (62.8% 2.0%) that were also MHC later stages of the infection (e.g., between 6 and 8 d postinfection).
II+ and Ly-6C+ (45.0% 9.5%) with Gr-1+ cells associated with Such an analysis might reveal either a reversion to an inactivated
31.7% 4.5% of the CD103+ MHC II+ Ly-6C+ population. Fi- microglial phenotype, or provide evidence for this populations
nally, P3 EYFP+ cells were predominantly CD103neg (85.0% further differentiation. Given that we have not identied an im-
0.1%) cells that were also MHC II+ and Ly-6C+ (45.5% 4.5%). munogenic role for P1 and that it represents the largest population

Fig. 4. The P1 population is associated


with the brain parenchyma, but P2
EYFP+ cells are part of the inltrating
peripheral response. (A) Lethally irradi-
ated Cd45.1 mice, successfully rescued
with Cd11c/eyfp (CD45.2) Tg bone mar-
row and their (B) reciprocal chimeras
were intranasally infected with VSV at
the LD50. At 96 hpi, blood and OB were
analyzed by ow cytometry using anti-
bodies able to discriminate between
CD45.1 and CD45.2 variants. In both
scenarios a CD45int CD11bhi population
(containing P1) was identied as brain
resident and radio-resistant, and a
CD45hi CD11bhi population (containing
P2) was consistently derived from donor
progenitor cells from the periphery.
The P3 (CD45hi CD11bint) population
was conspicuously absent from each
chimera set, however, as control (C) CD11c/eyfp mice infected with the same batch of VSV possessed all three populations. These ndings were corroborated
by (D) EYFP+ cell populations. Data are representative of three experiments with n = 56 per experiment; error bars represent SEM. *P valuevs. T Cell Only < 0.05,
***P valuevs. T Cell Only < 0.001.

6178 | www.pnas.org/cgi/doi/10.1073/pnas.1203941109 DAgostino et al.


Fig. 5. bDC populations contain a diverse
array of CD103+ CD11b+ and CD103neg
CD11b+ cells. (A) Phenotypic analysis of
TCRneg, CD19neg, CD45+, and CD11b+ cells
represented by the three EYFP+ populations
using a variety of markers characteristic of
DC and other immunological subsets. MG
(CD45int, CD11b+ and EYFPneg) and common
DC (Linneg EYFPhi) from the spleen of infec-
ted mice were used as a control. bDC pop-
ulations were further discriminated by (B)
CD103+ and (C) CD103neg subpopulations
possessing MHC II, Gr-1, and Ly-6c+ cells. P1 is
composed of a more-or-less equal number of
CD103+ and CD103neg cells, but all are MHC
II, Gr-1, and Ly-6cnegative. P2 contains sev-
eral populations of cells with phenotypes
analogous to mucosal DCs. Finally, P3 is
predominantly composed of CD103neg cells

IMMUNOLOGY
with phenotypes similar to monocytes-de-
rived DC. These histograms and dot plots are
representative of three separate experi-
ments, with two mice OB pooled per anti-
body per experiment.

responding early on during the acute VSV infection, we are cur- the OB of VSV intranasally infected mice, perhaps from the nasal
rently exploring other functional possibilities for these cells. epithelium.
The second EYFP+ population of radio-sensitive CD45hi The third EYFP+ population observed was classied by
CD11bhi (P2) cells was reminiscent of peripherally inltrating a CD45hi CD11bint (P3) phenotype. We believe P3 cells represent
leukocytes identied during Theilers murine encephalitis virus a unique population of DC yet to be described in CNS viral in-
infection of rats, and experimental autoimmune encephalomyelitis fection models. This population was the least abundant of the
mouse models (34, 35). P2 cells differed from the P1 and MG EYFP+ cells, but had costimulatory molecule expression on par
populations in terms of MHC II and costimulatory molecule ex- with splenic cDC and P2 bDC, was able to process antigen and
pression. P2 cells were also found to contain CD103+ CD11c+ F4/ promote T-cell proliferation (often more robustly than splenic
80+ Gr-1+ and Ly-6Chi subpopulations, a phenotype similar to cDC controls), and induced a TH1 bias in proliferating CD4+ T
mucosal DC (10), and were found to be functionally active in most lymphocytes. The P3 population, however, was different from the
T-cell proliferation assays. The P2 bDC were also able to promote other two EYFP+ populations in that, although it possessed both
a TH1 bias in proliferating CD4+ T lymphocytes. Taken as a whole, MHC II+ and Ly-6Chi populations, it was also mostly negative for
these observations have led us to formulate the hypothesis that P2 CD103, CD115, F4/80, and Gr-1. These phenotypes draw simi-
derive from a population of mucosal DC (10), which inltrates larities to inammatory monocytes (41, 42), which have the ability

DAgostino et al. PNAS | April 17, 2012 | vol. 109 | no. 16 | 6179
to form monocyte derived DC populations. Taken as a whole, the a ToxinSensor Chromogenic LAL Endotoxin Assay Kit (GenScript). See SI
exact DC-subset and source of P3 remains uncertain, as our efforts Materials and Methods for intranasal administration procedure.
to identify the origin and source of the P3 population were con-
founded by those cells being completely absent from VSV-infec- Flow Cytometry and FACS. After 96 hpi, mice were rapidly decapitated, brains
ted irradiation chimeras. We suspect this might be because of P3 extracted, and placed in ice-cold HBSS. OB were subsequently removed and
representing a radiosensitive population, the source of which is single cell suspensions obtained by digesting with collagenase D (Roche),
some other organ (e.g., spleen or liver) that is not immediately dispase II (Roche), and DNase I (Invitrogen) in conjunction with repeated
replenished by bone marrow. However, an alternate hypothesis is trituration using a GentleMACS Dissociator (Miltenyi Biotec) and serological
that the absence of P3 is because of a lingering proinammatory pipetting. See SI Materials and Methods for antibodies, ow cytometry
condition caused by the lethal irradiation (43). staining procedure, and spleen DC isolation.
In conclusion, we identied immune cells acting as sentinels at
vulnerable points of the CNS. These cells have the capacity to Antigen Presentation Assays. Because of the limited number of viable bDC
orchestrate adaptive immune responses in the face of a viral collected per OB, DC to T-cell ratios were titered, alongside antigen con-
pathogen and demonstrate characteristics of DC. Furthermore, centrations and T-cell plating number, to maximize both proliferation sen-
our data provide insight by building upon our previous obser- sitivity and sample replicates within each experiment. See SI Materials and
vations of CD11c+ cells with a dendritic morphology to show Methods for details about the antigen presentation assay.
a heterogeneous group of immune cells with complex immuno- See SI Materials and Methods for additional details on the mouse models,
logical functions. Our hope is that further dissection of the in- antibodies, immunouorescence, radiation chimeras, EdU incorporation
teraction between bDC and disease, resulting from viral stimuli assays, and the statistics used in this article.
or in other present day models of neurodegenerative diseases,
may ultimately lead to the identication of therapeutic targets ACKNOWLEDGMENTS. We thank Drs. Carol Shoshkes Reiss, Margaret
for ameliorating neuro-inammatory diseases. MacDonald, Charles Rice, and Leo Lefranois for providing useful reagents;
Drs. Juliana Idoyaga, Andres Gottfried-Blackmore, Elizabeth Waters, Ulrike
Materials and Methods W. Kaunzner, Cheolho Cheong, and Jae-Hoon Choi for their valuable rec-
ommendations; and The Rockefeller Universitys Comparative Bioscience
Viral Infection Model. VSV, Indiana Serotype (ATCC VR-158) and rVSV-OVA Center, Flow Cytometry Resource Center, and the Bio-Imaging Resource
(36) were isolated from infected BHK (ATCC CCL-10) supernatants and pu- Center for their expert technical assistance. This research is supported by a
ried on a 540% sucrose gradient. Endotoxin levels were determined using gift from the Peter Deane Trust (to K.B.).

1. Steinman RM, Idoyaga J (2010) Features of the dendritic cell lineage. Immunol Rev endogenous interferon: possible mechanism for natural resistance to virus infection.
234:517. J Virol 61:812818.
2. Steinman RM, et al. (2003) Dendritic cell function in vivo during the steady state: A 24. Rodriguez Boulan E, Sabatini DD (1978) Asymmetric budding of viruses in epithelial
role in peripheral tolerance. Ann N Y Acad Sci 987:1525. monlayers: A model system for study of epithelial polarity. Proc Natl Acad Sci USA 75:
3. Steinman RM, Hawiger D, Nussenzweig MC (2003) Tolerogenic dendritic cells. Annu 50715075.
Rev Immunol 21:685711. 25. Grodums EI (1976) Ultrastructure of mouse periventricular and choroid plexus tissues
4. Steinman RM, Cohn ZA (1973) Identication of a novel cell type in peripheral lymphoid in experimental vesicular stomatitis virus infection. Arch Virol 51:7585.
organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med 137:11421162. 26. Lundh B, Kristensson K, Norrby E (1987) Selective infections of olfactory and re-
5. Steinman RM, Witmer MD (1978) Lymphoid dendritic cells are potent stimulators of spiratory epithelium by vesicular stomatitis and Sendai viruses. Neuropathol Appl
the primary mixed leukocyte reaction in mice. Proc Natl Acad Sci USA 75: Neurobiol 13:111122.
51325136. 27. Huneycutt BS, et al. (1994) Distribution of vesicular stomatitis virus proteins in the
6. Nussenzweig MC, Steinman RM (1980) Contribution of dendritic cells to stimulation of brains of BALB/c mice following intranasal inoculation: An immunohistochemical
the murine syngeneic mixed leukocyte reaction. J Exp Med 151:11961212. analysis. Brain Res 635:8195.
7. Shortman K, Liu YJ (2002) Mouse and human dendritic cell subtypes. Nat Rev Immunol 28. Raivich G, et al. (1999) Neuroglial activation repertoire in the injured brain: Graded
2:151161. response, molecular mechanisms and cues to physiological function. Brain Res Brain
8. Dudziak D, et al. (2007) Differential antigen processing by dendritic cell subsets Res Rev 30:77105.
in vivo. Science 315:107111. 29. Lindquist RL, et al. (2004) Visualizing dendritic cell networks in vivo. Nat Immunol 5:
9. Merad M, Ginhoux F, Collin M (2008) Origin, homeostasis and function of Langerhans 12431250.
cells and other langerin-expressing dendritic cells. Nat Rev Immunol 8:935947. 30. Nimmerjahn A, Kirchhoff F, Helmchen F (2005) Resting microglial cells are highly
10. Iwasaki A (2007) Mucosal dendritic cells. Annu Rev Immunol 25:381418. dynamic surveillants of brain parenchyma in vivo. Science 308:13141318.
11. Ginhoux F, et al. (2009) The origin and development of nonlymphoid tissue CD103+ 31. Santambrogio L, et al. (2001) Developmental plasticity of CNS microglia. Proc Natl
DCs. J Exp Med 206:31153130. Acad Sci USA 98:62956300.
12. Choi JH, et al. (2009) Identication of antigen-presenting dendritic cells in mouse 32. Mildner A, et al. (2007) Microglia in the adult brain arise from Ly-6ChiCCR2+ mono-
aorta and cardiac valves. J Exp Med 206:497505. cytes only under dened host conditions. Nat Neurosci 10:15441553.
13. Liu YJ (2005) IPC: Professional type 1 interferon-producing cells and plasmacytoid 33. Getts DR, et al. (2008) Ly6c+ inammatory monocytes are microglial precursors
dendritic cell precursors. Annu Rev Immunol 23:275306. recruited in a pathogenic manner in West Nile virus encephalitis. J Exp Med 205:
14. Sung SS, et al. (2006) A major lung CD103 (alphaE)-beta7 integrin-positive epithelial 23192337.
dendritic cell population expressing Langerin and tight junction proteins. J Immunol 34. Greter M, et al. (2005) Dendritic cells permit immune invasion of the CNS in an animal
176:21612172. model of multiple sclerosis. Nat Med 11:328334.
15. Johansson-Lindbom B, et al. (2005) Functional specialization of gut CD103+ dendritic 35. Sedgwick JD, et al. (1991) Isolation and direct characterization of resident microglial
cells in the regulation of tissue-selective T cell homing. J Exp Med 202:10631073. cells from the normal and inamed central nervous system. Proc Natl Acad Sci USA 88:
16. Jaensson E, et al. (2008) Small intestinal CD103+ dendritic cells display unique func- 74387442.
tional properties that are conserved between mice and humans. J Exp Med 205: 36. Kim SK, et al. (1998) Generation of mucosal cytotoxic T cells against soluble protein by
21392149. tissue-specic environmental and costimulatory signals. Proc Natl Acad Sci USA 95:
17. Bulloch K, et al. (2008) CD11c/EYFP transgene illuminates a discrete network of 1081410819.
dendritic cells within the embryonic, neonatal, adult, and injured mouse brain. 37. Barr JN, Whelan SP, Wertz GW (2002) Transcriptional control of the RNA-dependent
J Comp Neurol 508:687710. RNA polymerase of vesicular stomatitis virus. Biochim Biophys Acta 1577:337353.
18. Anandasabapathy N, et al. (2011) Flt3L controls the development of radiosensitive 38. Chan CW, et al. (2006) Interferon-producing killer dendritic cells provide a link be-
dendritic cells in the meninges and choroid plexus of the steady-state mouse brain. tween innate and adaptive immunity. Nat Med 12:207213.
J Exp Med 208:16951705. 39. Taieb J, et al. (2006) A novel dendritic cell subset involved in tumor im-
19. Gottfried-Blackmore A, et al. (2009) Acute in vivo exposure to interferon-gamma munosurveillance. Nat Med 12:214219.
enables resident brain dendritic cells to become effective antigen presenting cells. 40. McGavern DB, Kang SS (2011) Illuminating viral infections in the nervous system. Nat
Proc Natl Acad Sci USA 106:2091820923. Rev Immunol 11:318329.
20. Felger JC, et al. (2010) Brain dendritic cells in ischemic stroke: Time course, activation 41. Geissmann F, et al. (2008) Blood monocytes: Distinct subsets, how they relate to
state, and origin. Brain Behav Immun 24:724737. dendritic cells, and their possible roles in the regulation of T-cell responses. Immunol
21. Hickey WF, Kimura H (1988) Perivascular microglial cells of the CNS are bone marrow- Cell Biol 86:398408.
derived and present antigen in vivo. Science 239:290292. 42. Geissmann F, et al. (2010) Development of monocytes, macrophages, and dendritic
22. Bi Z, Barna M, Komatsu T, Reiss CS (1995) Vesicular stomatitis virus infection of the central cells. Science 327:656661.
nervous system activates both innate and acquired immunity. J Virol 69:64666472. 43. Linard C, et al. (2004) Acute induction of inammatory cytokine expression after
23. Vogel SN, Fertsch D (1987) Macrophages from endotoxin-hyporesponsive (Lpsd) C3H/ gamma-irradiation in the rat: effect of an NF-kappaB inhibitor. Int J Radiat Oncol
HeJ mice are permissive for vesicular stomatitis virus because of reduced levels of Biol Phys 58:427434.

6180 | www.pnas.org/cgi/doi/10.1073/pnas.1203941109 DAgostino et al.

You might also like