ArticlePDF Available

A critical role for the sphingosine analog AAL-R in dampening the cytokine response during influenza virus infection

Authors:

Abstract and Figures

Pulmonary tissue damage resulting from influenza virus infection is caused by both the cytolytic activity of the virus and the host immune response. Immune-mediated injury results from T cell-mediated destruction of virus-infected cells and by release of cytokines and chemokines that attract polymorphonuclear leukocytes (PML) and macrophages to the infected site. The cytokines/chemokines potentiate dendritic cell (DC) activation and T cell expansion, which further enhances local damage. Here we report that immune modulation by local administration to the respiratory tract of sphingosine analog AAL-R significantly dampens the release of cytokines and chemokines while maintaining protective neutralizing antibody and cytotoxic T cell responses. As a result there was a marked reduction of infiltrating PML and macrophages into the lung and resultant pulmonary tissue injury. DC maturation was suppressed, which limited proliferation of specific antiviral T cells in the lung and draining lymph nodes. Further, AAL-R was effective in controlling CD8(+) T cell accumulation in the lungs even when given 4 days after initiation of influenza virus infection. These data indicate that sphingosine analogs display useful potential for controlling the immunopathology caused by influenza virus.
Sphingosine analog AAL-R inhibits both induction of influenza virus-specific T cells in MLN and activation of DC in the lung. (A) Number (left) and percentage (right) of Thy1.1 TCR transgenic GP33/CD8 T cells in MLN 5 dpi with mock or FLU-LCMV and i.t. administration of VEH or AAL-R (0.3 mg/kg) 1 hpi. Representative of two different experiments, both showed similar results, n 3-4 mice per group, mean SEM; * P 0.05. (B) Number (left) and percentage (right) of Thy1.1 TCR transgenic GP65/CD4 T cells in MLN 6 dpi with mock or FLU-LCMV and i.t. instillation with VEH or AAL-R (0.1 mg/kg) 1 hpi. n 4 mice per group, mean SEM; * P 0.07. Number of (C) CD11c CD11b CD103cells and (D) CD11c CD11b-CD103 cells in the lungs at 2 dpi with mock or FLU-LCMV and i.t. treatment with VEH or 0.1 mg/kg AAL-R 1 hpi. (E) Number of annexin-V CD11c cells in lungs 12 h after infection with mock or FLULCMV, and i.t. treatment with VEH or 0.1 mg/kg AAL-R 1 hpi. Representative of two different experiments, all showed similar results, n 4 mice per group; * P 0.05. (F and G) Mean fluorescence intensity (MFI) of MHC-I (H-2K b ), MHC-II (I-A b ) and B7-2 on (F) CD11b DC and (G) CD103 DC was analyzed. Cells were isolated from lungs 2 dpi with FLU-LCMV and i.t. treatment with VEH or AAL-R (0.1 mg/kg) 1 hpi. n 4 mice per group; mean SEM. * , P 0.05. (H) Proliferation of T cells cultured alone (top); with DC from FLU-LCMV VEH treated mice (center); with DC from FLU-LCMV AAL-R-treated mice (bottom). See SI Materials and Methods for details. Representative of 3 different experiments, all showed similar results.
… 
Content may be subject to copyright.
A critical role for the sphingosine analog AAL-R in
dampening the cytokine response during influenza
virus infection
David Marsolais
a,1
, Bumsuk Hahm
b,c,1
, Kevin B. Walsh
b,1
, Kurt H. Edelmann
b
, Dorian McGavern
b
, Yasuko Hatta
d
,
Yoshihiro Kawaoka
d,e
, Hugh Rosen
a,2
, and Michael B. A. Oldstone
b,2
aDepartments of Chemical Physiology and Immunology, The Scripps Research Institute, La Jolla, CA 92037; bDepartment of Immunology and Microbial
Science, The Scripps Research Institute, La Jolla, CA 92037; cDepartments of Surgery, and Molecular Microbiology and Immunology, Center for Cellular
and Molecular Immunology, University of Missouri, Columbia, MO 65212; dDepartment of Pathobiological Sciences, University of Wisconsin-Madison,
Madison, WI 53706; and eDivision of Virology, Department of Microbiology and Immunology, and International Research Center for Infectious
Diseases, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
Contributed by Michael B. A. Oldstone, December 12, 2008 (sent for review November 6, 2008)
Pulmonary tissue damage resulting from influenza virus infection
is caused by both the cytolytic activity of the virus and the host
immune response. Immune-mediated injury results from T cell-
mediated destruction of virus-infected cells and by release of
cytokines and chemokines that attract polymorphonuclear leuko-
cytes (PML) and macrophages to the infected site. The cytokines/
chemokines potentiate dendritic cell (DC) activation and T cell
expansion, which further enhances local damage. Here we report
that immune modulation by local administration to the respiratory
tract of sphingosine analog AAL-R significantly dampens the re-
lease of cytokines and chemokines while maintaining protective
neutralizing antibody and cytotoxic T cell responses. As a result
there was a marked reduction of infiltrating PML and macrophages
into the lung and resultant pulmonary tissue injury. DC maturation
was suppressed, which limited proliferation of specific antiviral T
cells in the lung and draining lymph nodes. Further, AAL-R was
effective in controlling CD8
T cell accumulation in the lungs even
when given 4 days after initiation of influenza virus infection.
These data indicate that sphingosine analogs display useful potential
for controlling the immunopathology caused by influenza virus.
immunopathology dendritic cells cytokine storm T cells
The antiviral host response evolved to limit the spread of
infection at the cost of causing tissue injury. There is a balance
between the protective and injurious responses that leads either to
the purging of infectious virus and host rec overy or to severe disease
and even death. Thus, strategies to balance the antiviral immune
response in favor of host outcome need to be developed.
T cell response elicited early in the course of infection recognizes,
attacks and lyses virus-infected cells to eliminate potential factories
of progeny viruses (1–5). The protective inf luenza antibody re-
sponse is elicited later and plays a role in controlling re-infection (6,
7). The innate and adaptive immune systems release wide varieties
of cytokines and chemokines that activate and attract inflammator y
cells to the site of infection (2, 8, 9). However, these molecules can
also cause the host harm by a phenomenon known as cytokine
storm. Cytokine storm has been convincingly documented both in
experimental animals infected with the 1918/1919 and H5N1 in-
fluenza viruses (2, 10 –13) as well as in humans (14–18) succumbing
to H5N1 infection. Although antiviral drugs can be used to treat the
virus, a strategy to balance the resultant cytokine release and lung
injury while maintaining benefits of the antiviral protective immune
response is needed (19). Influenza virus replication is most often
limited primarily to the respiratory tract but the systemic signs and
symptoms of disease, e.g., fever, muscle pain and shakes, intestinal
tract involvement, are related to cytokine effects (20, 21).
We reported that intratracheal (i.t.) delivery of the sphingosine
analog AAL-R or its phosphate ester inhibited virus-specific T cell
responses to influenza-virus infection whereas, in contrast, neither
intraperitoneal (i.p.) delivery of AAL-R nor i.t. administration of
the non-phosphorylable stereoisomer AAL-S were effective (22).
Here, we extend those findings by determining the mechanism of
AAL-R down-modulation of the antiviral T cell response. Using a
mouse model of influenza virus infection, three findings are made.
First, local administration of sphingosine analog AAL-R to the
respiratory tract down-modulates the anti-influenza virus T cell
response and inhibits cytokine/chemokine release without altering
the formation, kinetics or titer of anti-inf luenza-neutralizing anti-
bodies. Limiting the navigation signals of cytok ines and chemokines
reduces the infiltration of PML and macrophages into the lung.
Second, AAL-R acts on pulmonary and draining lymph node
dendritic cells (DC). AAL-R significantly down-regulates major
histocompatibility (MHC) class I and II molecules, as well as
co-stimulatory B7–2 molecule from the surfaces of DC in the lung
and draining lymph nodes, leading to a significant decrease in
accumulation of virus-specific T cells in the lung. Third, AA L-R
therapy is effective when administered 4 days after initiation of
influenza virus infection. Thus, short-term modulation of S1P
receptors in lungs interferes with antigen presentation by DC and
provides an additional therapeutic approach in treating aberrant
cytokine production in influenza and likely other infectious respi-
ratory diseases.
Results
An In Vivo Model for Quantitating and Monitoring Migration of
Influenza Virus-Specific T Cells into the Lung. We established an in
vivo model system to study inf luenza virus-mediated host immune
responses by generating a recombinant influenza virus. A recom-
binant A/WSN/33 (WSN; H1N1) virus (FLU-LCMV) was engi-
neered to bear the immunodominant H-2D
b
CD8 (GP 33–41) and
I-A
b
CD4 (GP 65–77) T cell receptor epitopes of lymphocytic
choriomeningitis virus (LCMV) in its neuraminidase (NA) stalk
using reverse genetics (23) (Fig. 1A). Availability of transgenic (tg)
mice in which the T cell receptor (TCR) is specific for these LCMV
immunodominant epitopes allowed in vivo quantification of T cell
trafficking (24) and visualization of T cell immunobiology (25).
Author contributions: D. Marsolais, B.H., K.B.W., K.H.E., H.R., and M.B.A.O. designed
research; D. Marsolais, B.H., K.B.W., K.H.E., and D. McGavern performed research; Y.H.
and Y.K. contributed new reagents/analytic tools; D. Marsolais, B.H., K.B.W., K.H.E., D.
McGavern, H.R., and M.B.A.O. analyzed data; and D. Marsolais, B.H., K.B.W., H.R., and
M.B.A.O. wrote the paper.
The authors declare no conflict of interest.
1D. Marsolais, B.H., and K.B.W. contributed equally to this work.
2To whom correspondence may be addressed. E-mail: hrosen@scripps.edu or
mbaobo@scripps.edu.
This article contains supporting information online at www.pnas.org/cgi/content/full/
0812689106/DCSupplemental.
© 2009 by The National Academy of Sciences of the USA
1560–1565
PNAS
February 3, 2009
vol. 106
no. 5 www.pnas.orgcgidoi10.1073pnas.0812689106
Intranasal (i.n.) or i.t. inoculation of 1 10
5
PFU of FLU-LCMV
resulted in expression of influenza viral antigens throughout the
lungs. As shown in Fig. 1C, at 6 days post-infection (dpi), viral
antigen is observed throughout the lung parenchyma. High power
examination showed viral antigen in bronchioles and epithelial cells
surrounding the airways (data not shown). Mice adoptively trans-
ferred with 2.5 10
4
Thy1.1
GP 33–41-restricted CD8
(GP33/
CD8
) T cells and 2.5 10
4
GFP-expressing GP 65–77-restricted
CD4
(GP65/CD4
) T cells and subsequently inoculated i.n. with
FLU-LCMV displayed pulmonary infiltration of virus-specific
CD8
and CD4
T cells (Fig. 1D) at six dpi. As shown in Fig. 1E
and F, pulmonary accumulation of GFP
GP33/CD8
T cells
occurred after 4 dpi, reached a plateau between 6 and 8 dpi, then
dramatically decreased at 10 dpi, as revealed by both fluorescence
imaging on whole lung sections (Fig. 1E) and on single-cell sus-
pension from lung tissue analyzed by f low cytometry (Fig. 1F). This
model system provided an opportunity for quantification of virus-
specific T cells and a mechanistic study of the interplay between
host immunity, inf luenza virus, and their modulation by chemical
probes.
Sphingosine Analog AAL-R Inhibits the Accumulation of Influenza
Virus-Specific T Cells in the Lung but Does Not Interfere with the
Production of Protective Neutralizing Antiiinfluenza Antibodies. Ear-
lier we reported that local administration of a single 0.1-mg/kg dose
of AAL-R by an i.t., but not i.p. route, specifically down-modulated
virus-specific T cell accumulation in the lung, whereas both routes
of delivery induced T cell recirculation from the blood to secondary
lymphoid tissues (22). This inhibition of T cell accumulation in the
lung depended on phosphorylation of AAL-R as the chiral enan-
tiomer molecule AAL-S that cannot be phosphorylated efficiently
in vivo was impotent in restricting T cell infiltration in the lung.
We now assessed the effect of AA L-R treatment on the virus-
specific immune responses. Treatment with 0.1 mg/kg AAL-R i.t.
following FLU-LCMV inoculation inhibited the accumulation of
total CD4
and CD8
T cells (Fig. 2A), as well as virus-specific T
cells in the lung (Fig. 2B). However, AAL-R i.t. administration had
no effect on the production of influenza virus-specific total immu-
noglobulins (Ig) (Fig. 2C) and IgM (data not shown) at 6, 10, 15, and
30 days following virus infection. Assays for influenza virus neu-
tralizing antibodies revealed that the amount of immunoglobulins
was not impaired by AAL-R administration when compared to
AAL-S and vehicle-treated groups (Fig. 2D). Administration of
AAL-R did not significantly increase viral burden within the lung,
but slightly delayed viral clearance (Fig. 2 E). By day 10 post-
infection, both vehicle and AAL-R-treated mice were negative for
infectious virus. These results indicate that AAL-R therapy given
locally into the respiratory tract interferes with neither influenza
virus replication nor the generation of protective antibodies but
significantly down-regulated the numbers of virus-specific T cells in
the lung. Although numbers of virus-specific CD8
T cells were
reduced, influenza virus infection was still controlled.
AAL-R Does Not Decrease FLU-LCMV Titers or Cytotoxic Activity of
Remaining Virus-Specific CD8
T Cells. We next determined how
AAL-R treatment would influence viral kinetics and ex vivo
cytotoxic T lymphocyte (CTL) activity following FLU-LCMV
infection. As expected, AAL-R reduced the pulmonary content of
CD8
T cells having the ability to produce IFN-
in response to
GP33 peptide stimulation in vitro 7 dpi, and this effect was
maintained until 8 dpi (Table 1). Also, the kinetics of viral clearance
in mice treated with AAL-R was not significantly altered compared
to viral clearance observed in mice administered with VEH or
AAL-S (Table 1), suggesting the remaining anti-viral T cell activity
was sufficient to clear virus from the lungs. We further tested this
idea by measuring the cell-based cytotoxic activity of GP33-specific
CD8
T cells extracted from lung and spleen after treatment with
AAL-R, AAL-S or VEH (Fig. S1). CTL activity of virus-specific
CD8
T cells in the lung (Fig. S1A) or spleen (Fig. S1B) was not
adversely affected by AAL-R when compared with VEH and
AAL-S treatments. Intracellular cytokine staining revealed a sig-
nificant reduction in the number of virus-specific CD8
T cells in
AAL-R treated mice (Fig. S1C). Calculation of cytolytic activity on
a per cell basis revealed a significant increase in the CTL activity of
GP33-specific splenocytes in AAL-R-treated mice when compared
with AAL-S or vehicle treatments.
Decreased Accumulation of Virus-Specific T Lymphocytes in the Lung
Following AAL-R Treatment Is Associated with Suppression of Cyto-
kine Synthesis and Alleviation of Immunopathology. Next we assessed
the ability of AAL-R to regulate inf luenza virus-induced cytokines/
Fig. 1. Generation of recombinant influenza virus and pulmonary accumula-
tion of viral antigens and virus-specific T cells in vivo. (A) Reverse genetics strategy
to insert CD4 and CD8 immunodominant epitopes of LCMV glycoprotein (GP),
amino acids (aa) 33–41 (GP33) for CD8T cells, and GP amino acid 65–77 (GP65)
for CD4T cells, into the neuraminidase (NA) stalk of A/WSN/33 virus. (B) Cartoon
to display the administration of fluorochrome-labeled or phenotypically identi-
fiable congenic LCMV-specific GP33/CD8T cells and GP65/CD4T cells from T cell
receptor transgenic mice and instillation of influenza virus. (C) Distribution of
influenza viral antigens (green) in the whole lung at day 6 postinfluenza virus i.n.
administration. Nuclei appear in blue. (D) Simultaneous detection of adoptively
transferred CD4(green) and CD8T cells (red) specific for FLU-LCMV at day 6
post-influenza i.n. inoculation. Nuclei appear in blue. (E) Analysis of virus-specific
GFPTCR transgenic GP33/CD8T cells on day 4 (d4), d6, d8, and d10 after i.n.
FLU-LCMV infection. GFPTCR transgenic GP33/CD8T cells appear in green and
nuclei were stained with DAPI (blue). (F) By comparison to (E), flow cytometric
analysis of GFPTCR transgenic GP33/CD8T cells after i.n. FLU-LCMV infection.
The percentages of GFPTCR transgenic GP33/CD8T cells are depicted on
scattergrams. Eand Fshow representative samples of one mouse from a group of
three.
Marsolais et al. PNAS
February 3, 2009
vol. 106
no. 5
1561
MICROBIOLOGY
chemokines synthesis in vivo. We noted that upon FLU-LCMV
infection there was a significant up-regulation of multiple cytokines/
chemokines, most notably marked increase of interleukin (IL)-1
,
IL-1
, IL-6, IL-10, IL-12, monocyte chemoattractant protein
(MCP-1, CCL2), TNF-
, macrophage inflammatory protein (MIP-
1
, CCL3), granulocyte macrophage colony stimulating factor
(GM-CSF), and RANTES (CCL5) (Fig. 3A). Of interest IL-6 and
MCP-1 levels were strikingly increased in our model, an event also
observed in both H5N1-infected humans (16), as well as in ma-
caques (11) and mice (10) infected with the 1918/1919 recon-
structed influenza virus. Treatment with AAL-R significantly
inhibited the production of IL-1
, IL-1
, IL-6, IL-10, MCP-1,
TNF-
, and GM-CSF 2 dpi, when compared with mice treated with
AAL-S (Fig. 3B). Histological examination of lungs revealed that
AAL-R, but not AAL-S, dramatically decreased the accumulation
of lymphocytes, leukocytes, monocytes and macrophages and in-
jury in the distal airways (Fig. 3C) while modestly enhancing
survival time (Fig. 3D). Thus, delivery of AAL-R i.t. diminished
cytokine-mediated histopathologic injury of the lungs during inf lu-
enza virus infection. Cytokine/chemokine suppression was associ-
ated with a decrease of GR-1
PML and F4/80
macrophages (Fig.
3E) into the lung which directly correlated with the diminished
alveolar cytopathology and infiltration in the lung. However, de-
spite AAL-R successfully down-modulating T cell-mediated injury
and cytokine/chemokine release there was no change in lung
A/WSN/33 viral titers even at 2 mg/kg of AAL-R compared with
treatment with AAL-S or vehicle (Fig. 3F). This indicated that
AAL-R neither disrupted viral replication nor the resultant pro-
tective antiviral T cell immune responses but did successfully
downmodulate immunopathology.
AAL-R Protects by Impairing the Antigen-Presenting Capacity of
Pulmonary DC. Upon pulmonary infection, inf luenza virus-specific
T cells are rapidly induced and proliferate in mediastinal lymph
node (MLN), then migrate to the infected sites, including the lungs
(26, 27). Given i.t., AAL-R significantly reduced the number of
virus-specific CD8
(Fig. 4A) and CD4
(Fig. 4B) T cells in the
MLNs 5 and 6 dpi with FLU-LCMV, indicating that AAL-R
inhibited clonal expansion of T cells upon influenza virus infection.
The number of Annexin V-positive virus-specific T cells in both
MLNs and lungs was not significantly altered by local treatment
with AAL-R (data not shown), suggesting that AAL-R altered T
cell stimulation by influencing antigen-presenting DC (28) rather
than deleting T cells.
We then focused on pulmonary DC and found that i.t. delivery
of AAL-R neither reduced the numbers of total DC (data not
shown) nor of specific DC subsets (Fig. 4Cand D). Moreover,
AAL-R did not interfere with the viability of CD11c
cells (Fig.
4E) in the lungs upon influenza virus infection. However, AAL-R
strongly suppressed influenza virus-induced DC activation in the
lungs as measured by down-regulation of MHC-I, MHC-II and
co-stimulatory molecule B7–2 on the surfaces of CD11c
cells.
AAL-R impaired maturation of CD11c
cell subsets
(CD11b
CD103
-
; CD11b
CD103
, CD11b
-
CD103
, and CD11b
-
CD103
-
) in the lung by reducing surface expression of MHC-I,
MHC-II and B7–2, with suppression of the CD11b
CD103
-
subset
being affected the most (Fig. 4Fand G) (data not shown). Stimu-
latory capacity of CD11c
cells was impaired by in vivo AAL-R
treatment, as confirmed by inefficient induction of virus-specific
CD8
T cell proliferation in vitro (Fig. 4H).
We found that impaired activation of DC in the lungs resulted in
Fig. 2. Effect of sphingosine analog on influenza virus-specific T cells and
antibodies as well as virus titers in the lung. (Aand B) Mice (n4 mice per group)
were infected with 1 105PFU of FLU-LCMV and administered i.t. with vehicle
(VEH) or 0.1 mg/kg AAL-R, or in B, with VEH, 0.1 mg/kg of AAL-R, or isomer control
AAL-S. At 6 dpi, lungs were processed for flow cytometric analysis to obtain
numbers of CD4and CD8T cells in lungs (A) or percentage of virus-specific TCR
transgenic GP33/CD8T cells among total CD8T cells in lungs (B). (Cand D) Four
to six mice per group were infected with mock or FLU-LCMV and treated with
VEH, AAL-S or AAL-R (0.1 mg/kg) 1 hpi. (C) Influenza virus-specific total immu-
noglobulins in serum were assessed by ELISA at 6, 10, 15, and 30 dpi, mean SD.
(D) Titers of virus neutralizing antibodies in serum were evaluated 30 dpi, mean
SEM. (E) PFU/gram of lung tissue in normal, unaltered C57BL/6 mice following i.t.
inoculation of 1 105PFUs of FLU-LCMV and i.t. delivery of 0.1 mg/kg of AAL-R
or vehicle 1hpi. At day 8 following AAL-R treatment, 2 of 4 mice had detectable
levels of virus at 1 102PFU/gram of lung. The level of detection is 1 102PFU/g
tissue. Four mice were used per group per timepoint. P0.23 for days 2, 4, and
6 after infection. ND: not detectable.
Table 1. Effect of AAL-R on antigen-specific CD8
T cells accumulation and viral titers
Treatment n
Day 5 after infection Day 7 after infection Day 8 after infection
GP33-specific cells Titer GP33-specific cells Titer GP33-specific cells Titer
VEH 4 4.1 10
2
1.1* 14.3 10
4
5.0
7.6 10
3
2.0 3.9 10
2
0.8 3.0 10
4
1.3 1.3 10
2
(1/4)
AAL-S 4 3.3 10
2
0.9 7.9 10
4
2.0 4.7 10
3
1.8 3.6 10
2
0.9 3.8 10
4
0.8 0.2 10
2
0.03 (3/4)
AAL-R 4 1.5 10
2
0.5 6.9 10
4
1.6 1.8 10
3
0.2* 26.0 10
2
12.0 2.3 10
4
0.8 10.1 10
2
6.8 (2/4)
Mice were infected with 1 105PFU of FLU-LCMV and administered i.t. with VEH and or 0.1 mg/kg AAL-S or AAL-R 1 hpi. *,P0.05 when compared to
vehicle-treated mice.
*Values are presented as average number SEM.
Values are presented as average PFU/g of tissue SEM.
Virus-positive mice/n.
1562
www.pnas.orgcgidoi10.1073pnas.0812689106 Marsolais et al.
subsequent inhibition of influenza virus-induced elevation of
CD11c
cell numbers (both CD11b
and CD8
) in the MLNs
(Fig. S2A), as well as impaired activation of CD8
DCs present
in the lymph nodes, which are efficient at priming CD8
T cells
(29). Intratracheal delivery of A AL-R also down-regulated MHC-I,
MHC-II and B7–2 co-stimulatory molecules on DC in MLN (Fig.
S2B). Thus, these results indicate that upon influenza virus infec-
tion, AAL-R locally administered to the respiratory tract disrupts
antigen-presenting DC network by blocking DC-mediated signal
transmission from the infection site to the MLNs, leading to a
dramatic decrease in T cell expansion.
AAL-R Therapy Administered at a Later Time Point During Influenza
Virus Infection Is Still Effective in Modulating the CD8
T Cell Response
to the Virus. The last experiment determined whether administering
0.1 mg/kg of AAL-R locally into the respiratory tract at 4 dpi was
effective in inhibiting T cell accumulation in the lung. As shown in
Fig. 5, six and eight days after giving 1 10
5
PFU of FLU-LCMV
the number of virus-specific CD8
T cells was significantly reduced
in the lung. In contrast, providing vehicle had no therapeutic effect.
Discussion
Here we make three main points. First, a single low-dose admin-
istration of the sphingosine analog AAL-R locally into the respi-
ratory airway, when provided at the time of inf luenza virus infec-
tion or after 4 days of infection, is able to effectively alter one of the
two major arms of the antiviral immune response. Virus-specific T
cell expansion in MLNs and accumulation in the lungs are signif-
icantly down-regulated whereas the production, kinetics and
amounts of protective neutralizing antibodies are not altered. In
addition, CD8
T cell cytotoxic activity is not impaired. Second,
AAL-R delivery in the respiratory tract can significantly decrease
the release of a variety of cytokines and chemokines known to
contribute to the cytokine storm effect, especially inhibition of IL-6
and MCP-1. The diminished synthesis of cytokines/chemokines
following AAL-R treatment correlates directly with a decreased
infiltration of PML and macrophages in the lung, resulting in less
cytopathology of alveolar cells and inflammatory driven congestion
of air spaces. Third, mechanistically, AA L-R acts on DC from both
draining lymph nodes and the lung. The drug inhibits DC accu-
mulation in the draining lymph nodes and down-regulates MHC
class I and II molecules as well as co-stimulatory molecule B7–2.
The decreased accumulation of DCs in the draining lymph nodes
combined with the decreased expression of stimulatory and co-
stimulatory molecules likely accounts for the diminished prolifer-
ation and expansion of T cells in draining lymph node and subse-
quent accumulation in the lung.
Injury to the lung accompanying influenza virus infection de-
pends on both the virus and action of the host’s immune response.
The virus being cytopathic lyses cells. In addition, infected cells may
apoptose thus removing the nidus of infection. Similarly, the
cytotoxic function of antiviral T cells is to kill virus-infected cells,
especially early in the infectious process, removing cells that make
progeny virus. However, by virus or immune-induced lysis of cells,
factors are released that attract scavenger PMLs and macrophages
to the infected site. These cells, while removing cellular debris, also
contribute to airway disease. Most important, the antiviral T cells
release cytokines/chemokines at the site of infection via their
specific recognition of infected targets. These factors then further
act to provide navigation signals to attract the PML and macro-
phages thereby potentiating the pulmonary injury. In addition, the
released cytokines/chemokines play the dominant role in causing
the generalized systemic manifestations of inf luenza virus infection.
Administration of sphingosine analog AAL-R strikingly im-
paired accumulation of DC in the draining lymph nodes, as well as
maturation of DC in the lung and MLN. There was a down-
regulation of MHC and co-stimulatory molecules required to
engage specific antiviral T cells. This led to diminished proliferation
and clonal expansion of T cells in the MLN. These effects could also
be shown with the phosphorylated S1P receptor agonist AFD-R
(22), suggesting that targeting S1P receptors locally in the lung and
on DC was sufficient to inhibit proinflammatory cytokine release
and virus-specific T cells expansion. Others have reported that
systemic delivery of sphingosine analog FTY720 can sequester
normal expanding lymphocytes in lymphoid organs in various
models (30–32) including during virus infection (33). However, this
was unlikely a mechanism in our studies for the following reasons.
First, elsewhere we documented that delivery of a single dose of the
sphingosine analog AA L-R (0.1 mg/kg) in the airways did not cause
sequestration of lymphocytes in draining lymphoid organs at 6 days
Fig. 3. AAL-R impairs cytokine release and leukocyte infiltration in the lungs
resulting in diminished alveolar inflammation and congestion but without alter-
ing infectious influenza virus titer. (Aand B) Multiplex ELISA was performed on
lung homogenates obtained at 2 dpi from uninfected mice (n4) or mice
infected with 1 105PFU FLU-LCMV i.t. (n12). Virus-infected mice were treated
with VEH or AAL-R (0.1 mg/kg) (n6). Cytokines detected at levels of more than
40 pg/ml in supernatant of lung homogenates and significanlty increased by at
least two-fold by viral infection over uninfected mice are shown. (A) Fold increase
of cytokines/chemokines from FLU-LCMV-infected mice over that of uninfected
mice. (B) Percentage of inhibition caused by AAL-R (black bars) and AAL-S
treatment (white bars) relatively to vehicle-treated mice. Mean SEM. *,P
0.05. (C) Naive C57BL/6 mice were (i) left untreated or (ii,iii,iv) infected i.t. with
1105PFU of FLU-LCMV. One hpi mice were either administered i.t. with VEH (ii),
AAL-S (iii: 0.1 mg/kg) or AAL-R (iv: 0.1 mg/kg), and euthanized 6 days later. Lungs
were processed for hematoxylin and eosin staining. Representative fields are
shown. (Scale bars, 50
m.) Results were equivalent in more than 20 fields
analyzed per group (5 fields/section, 2 sections/mouse and 2 mice/group). (D)
Percent survival was observed with 13 mice per group from two combined
experiments. Mice were infected i.t. with 5 103PFU of A/WSN/33 virus. One hour
later, mice were administered i.t. with vehicle or AAL-R (0.1 mg/kg) and moni-
tored daily for mortality. (E) Decrease in PML (GR-1) and macrophages (F4/80)
infiltrates in the lung. Mice were infected with 1 105PFU of FLU-LCMV and then
received i.t. either vehicle or AAL-R (0.1 mg/kg). Two dpi, Macrophages (F4/80)
and PML (GR1) numbers were evaluated by flow cytometry. Pool of 2 experi-
ments, n7–8, mean SEM; *significantly different from vehicle, P0.05. (F)
Titers of A/WSN/33 in lung three days after i.n. infection with 1 105 PFU of
A/WSN/33 and i.t. administration of VEH, or 2 mg/kg of AAL-S and AAL-R. Virus
plaques were quantitated on MDCK cells. Mean SEM; *P0.05.
Marsolais et al. PNAS
February 3, 2009
vol. 106
no. 5
1563
MICROBIOLOGY
after AAL-R treatment, but a decreased accumulation of virus-
specific T cells does occur in the lung (22). The latter was not
observed with systemic delivery of A AL-R. Further, i.p. delivery of
AAL-R, the route used by others to show sequestering of lympho-
cytes in lymphoid organs, was not associated with inhibition of DC
accumulation in draining lymph nodes (not shown). The fact that
MLN DC numbers were significantly altered by i.t. but not i.p.
treatment with AAL-R further indicates the likelihood of local
effects of the AAL-R analog on DC maturation and function
rather than changes of DC or lymphocyte migration from the
blood to MLN (34, 35). Thus, the effects of local administration
coupled with the failure of systemic dosing to induce suppression
of DC molecules indicate that lymphopenia alone could not
explain the immunomodulation we observed in the influenza
model. Further, we noted a dramatic decrease in numbers of
both CD11b
and CD8
DC in MLN. Others have shown that
CD8
DC are potent in stimulating T cells and do not originate
from the lung after inf luenza virus infection (26). Our results
thus suggest that the immunosuppressive effect of AAL-R is
caused by modulation of DC function rather than by altering T
cell trafficking.
Sphingosine analog did not inhibit the production of protective
antibody (Fig. 2) even in the occurrence of significant T cell
suppression. Generation of antibodies upon viral infection depends
on multiple factors including infection route, duration of viral
components’ presence in specific locations, contribution of various
anatomically distributed germinal centers (36), and dependency on
Fig. 4. Sphingosine analog AAL-R inhibits
both induction of influenza virus-specific T
cells in MLN and activation of DC in the lung.
(A) Number (left) and percentage (right) of
Thy1.1TCR transgenic GP33/CD8T cells in
MLN 5 dpi with mock or FLU-LCMV and i.t.
administration of VEH or AAL-R (0.3 mg/kg) 1
hpi. Representative of two different experi-
ments, both showed similar results, n3–4
mice per group, mean SEM; *P0.05. (B)
Number (left) and percentage (right) of
Thy1.1TCR transgenic GP65/CD4T cells in
MLN 6 dpi with mock or FLU-LCMV and i.t.
instillation with VEH or AAL-R (0.1 mg/kg) 1
hpi. n4 mice per group, mean SEM; *P
0.07. Number of (C) CD11cCD11bCD103-
cells and (D) CD11cCD11b-CD103cells in
the lungs at 2 dpi with mock or FLU-LCMV and
i.t. treatment with VEH or 0.1 mg/kg AAL-R 1
hpi. (E) Number of annexin-VCD11ccells in
lungs 12 h after infection with mock or FLU-
LCMV, and i.t. treatment with VEH or 0.1
mg/kg AAL-R 1 hpi. Representative of two
different experiments, all showed similar re-
sults, n4 mice per group; *P0.05. (Fand
G) Mean fluorescence intensity (MFI) of
MHC-I (H-2Kb), MHC-II (I-Ab) and B7–2 on (F)
CD11bDC and (G) CD103DC was analyzed.
Cells were isolated from lungs 2 dpi with
FLU-LCMV and i.t. treatment with VEH or
AAL-R (0.1 mg/kg) 1 hpi. n4 mice per group;
mean SEM. *,P0.05. (H) Proliferation of
T cells cultured alone (top); with DC from
FLU-LCMV VEH treated mice (center); with
DC from FLU-LCMV AAL-R-treated mice
(bottom). See SI Materials and Methods for
details. Representative of 3 different experi-
ments, all showed similar results.
Fig. 5. Sphingosine analog AAL-R administered i.t. 4 days after FLU-LCMV
infection is effective in limiting the accumulation of virus-specific CD8T cells
in the lungs. (A) Cartoon of the experimental protocol. Mice, 4 per group, were
infected with mock or 1 105PFU of FLU-LCMV i.t. followed 4 days later by i.t.
instillation of VEH or AAL-R (0.1 mg/kg). (B) Number of GP33/CD8T cells in
lungs is shown. Mean SEM; *,P0.09; **,P0.07.
1564
www.pnas.orgcgidoi10.1073pnas.0812689106 Marsolais et al.
T helper cells. Currently, the effect of systemic delivery of sphin-
gosine analog on B cell response is controversial (33, 37), although
no data are available for antibody response affected by local
treatment of the analog. It is possible that local AAL-R-mediated
split of B and T cell responses was caused by partial blockade of host
immunity to inf luenza virus as: 1) the sphingosine analog was
shown not to completely inhibit the release of cytokines upon
influenza virus infection (Fig. 3B) (22); 2) remaining virus-specific
CD4
T cells (Fig. 4B) might be sufficient to reach the threshold for
the generation of antibodies dependent on T helper cells; 3) there
was sufficient effector T cell activity to balance inf luenza virus
replication in the lung as infectious virus titers in the lung were
equivalent with AAL-R, A AL-S or vehicle treatment (Fig. 2E,Fig.
3F, and Table 1). Recently, other investigators showed that CD11c
hi
DC depletion upon influenza virus infection caused inhibition of T
cell expansion but did not affect antibody production (38), which is
consistent with our data. We are currently investigating local
modulation of S1P receptors and the mechanism for separation of
B cell response from T cell immunity.
Lastly, the observation that with AAL-R therapy virus titers in
the lung were not significantly increased despite the down-
modulation of numbers of antiviral-specific T cells, along with
diminished cytokine/chemokine release and decreased injur y to the
lung suggest that combinatory therapy using antiviral drugs in
concert with S1P analogs would be a preferred therapy. We are
currently evaluating this possibility in mice models of vir ulent H5N1
influenza virus infection. Further, DC are known to contain at least
four of the five S1P receptors. Dissection of the pulmonary DC S1P
receptor(s) involved could suggest the design of more effective
sphingosine analogs for treatment of lung inflammatory disorders.
Materials and Methods
Mice. Mice were bred and maintained in a closed breeding facility at The Scripps
Research Institute. C57BL/6, C57BL/6 Thy1.1DbGP33–41 TCR-tg, C57BL/6
GFPDbGP33–41 TCR-tg, C57BL/6 GFPI-AbGP61– 80 TCR-tg, and C57BL/6
Thy1.1I-AbGP61–80 TCR-tg were used in this study. The handling of all mice
conformed to the requirements of the National Institutes of Health and The
Scripps Research Institute’s Institutional Animal Care and Use Committee.
Cell Transfer. Virus-specific CD8or CD4T cells were enriched by negative
selection using Stemsep kit (Stemcell technologies), from the spleen of transgenic
mice. Cells (5 104) were then injected in the tail vein of congenic mice 1 day
before infection. When both CD8and CD4cells were transferred, 2.5 104of
each was used. To exclude any effect of transferred cells on the immunopatho-
logical response triggered by influenza virus infection, Figs. 2 CE,3AF, and 4
CHwere conducted without prior adoptive transfer of lymphocytes.
Viruses. To generate a recombinant WSN mutant virus (FLU-LCMV), we inserted
the LCMV immunodominant T cell-specific GP33 and GP65 tandem sequence
AAGGCTGTCTACAATTTTGCCACCTGTGGGGGACGCACAAUGGGTCTTAAGGG-
ACCCGACATTTACAAAGGAGTTTACCAATTTAAGTCAGTGGAGTTTGAT between
nucleotides 145 and 146 of WSN NA gene. Insertion of up to 28 aa into the NA
stalk does not impair NA function but insertion of more than 12 aa attenuates
the virus. A/WSN/33 (WSN; H1N1) and FLU-LCMV were generated by using
plasmid-based reverse genetics (23). Viruses were amplified and plaqued on
Madin-Darby Canine Kidney (MDCK) cells.
Statistical Analysis. Unless otherwise stated, bars represent means SEM and
averages were compared using a bidirectional unpaired Student’s ttest with a 5%
significance level. Star (*) was used to mark significant differences between 2
groups unless otherwise stated.
For specific information regarding the analytic assays performed in this study,
please consult SI Text.
ACKNOWLEDGMENTS. This is Publication Number 19725 from the Department
of Immunology and Microbial Science, Infectology and Chemical Physiology and
Immunology; and The Scripps Research Institute Molecular Screening Center, The
Scripps Research Institute (TSRI). This work was supported in part by USPHS grants
AI074564 (MBAO, HR, YK, BH, DM, KW), AI009484 (MBAO), AI05509 (HR),
AI069274 (YK), and NIMH-074404 (HR). YK and YH are also supported by Grants-
in-Aid from the Ministries of Education, Culture, Sports, Science and Technology
and of Health, Labor, and Welfare of Japan; HR is supported in part by a grant
from Kyorin Pharmaceutical Company. DM is supported by Le Fonds de la Re-
cherche en Sante du Quebec, Canada. The authors would like to acknowledge the
technical assistance of Megan Welch and Nora Leaf. The authors thank the TSRI
Flow Cytometry Core Facility; and Dusko Trajkovic for histological analyses.
1. Whitton JL, Oldstone MB (2001) in Fields virology, 4th ed., eds Knipe DM, Howley PM,
Griffin DE, Lamb RE, Martin MA, Rolzman B, Straus SE (Lippincott Williams & Wilkins,
Philadelphia), pp 285–320.
2. La Gruta NL, Kedzierska K, Stambas J, Doherty PC (2007) A question of self-preservation:
immunopathology in influenza virus infection. Immunol Cell Biol 85:85–92.
3. Graham MB, Braciale TJ (1997) Resistance to and recovery from lethal influenza virus
infection in B lymphocyte-deficient mice. J Exp Med 186:2063–2068.
4. Zinkernagel RM, Althage A (1977) Antiviral protection by virus-immune cytotoxic T
cells: infected target cells are lysed before infectious virus progeny is assembled. J Exp
Med 145:644– 651.
5. Moskophidis D, Kioussis D (1998) Contribution of virus-specific CD8cytotoxic T cells
to virus clearance or pathologic manifestations of influenza virus infection in a T cell
receptor transgenic mouse model. J Exp Med 188:223–232.
6. Goulding J, et al. (2007) Respiratory infections: do we ever recover? Proc Am Thorac Soc
4:618– 625.
7. Gerhard W (2001) The role of the antibody response in influenza virus infection. Curr
Top Microbiol Immunol 260:171–190.
8. Biron CA, Sen GC (2007) in Fields Virology, 5th ed., eds Knipe DM, Howley PM, Griffin
DE, Lamb RE, Martin MA, Rolzman B, Straus SE (Lippincott Williams & Wilkins, Phila-
delphia), pp 249–278.
9. Braciale TJ, Hahn YS, Burton DR (2007) in Fields Virology, 5th ed., eds Knipe DM,
Howley PM, Griffin DE, Lamb RE, Martin MA, Rolzman B, Straus SE (Lippincott Williams
& Wilkins, Philadelphia), pp 279–326.
10. Kash JC, et al. (2006) Genomic analysis of increased host immune and cell death
responses induced by 1918 influenza virus. Nature 443:578–581.
11. Kobasa D, et al. (2007) Aberrant innate immune response in lethal infection of
macaques with the 1918 influenza virus. Nature 445:319–323.
12. Cheung CY, et al. (2002) Induction of proinflammatory cytokines in human macro-
phages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human
disease? Lancet 360:1831–1837.
13. Tumpey TM, et al. (2005) Characterization of the reconstructed 1918 Spanish influenza
pandemic virus. Science 310:77–80.
14. Beigel JH, et al. (2005) Avian influenza A (H5N1) infection in humans. N Engl J Med
353:1374–1385.
15. Uiprasertkul M, et al. (2007) Apoptosis and Pathogenesis of Avian Influenza A (H5N1)
Virus in Humans. Emerg Infect Dis 13:708–712.
16. de Jong MD, et al. (2006) Fatal outcome of human influenza A (H5N1) is associated with
high viral load and hypercytokinemia. Nat Med 12:1203–1207.
17. To KF, et al. (2001) Pathology of fatal human infection associated with avian influenza
A H5N1 virus. J Med Virol 63:242–246.
18. Peiris JS, et al. (2004) Re-emergence of fatal human influenza A subtype H5N1 disease.
Lancet 363:617–619.
19. Pearson H (2006) Controlling immune response may cut bird flu death rate. Nat Med
12:1105.
20. Hilleman MR (2002) Realities and enigmas of human viral influenza: pathogenesis,
epidemiology and control. Vaccine 20:3068–3087.
21. Eccles R (2005) Understanding the symptoms of the common cold and influenza. Lancet
Infect Dis 5:718–725.
22. Marsolais D, et al. (2008) Local not systemic modulation of dendritic cell S1P receptors in
lung blunts virus-specific immune responses to influenza. Mol Pharmacol 74:896–903.
23. Neumann G, et al. (1999) Generation of influenza A viruses entirely from cloned cDNAs.
Proc Natl Acad Sci USA 96:9345–9350.
24. Christen U, et al. (2004) A viral epitope that mimics a self antigen can accelerate but not
initiate autoimmune diabetes. J Clin Invest 114:1290–1298.
25. McGavern DB, Christen U, Oldstone MB (2002) Molecular anatomy of antigen-specific
CD8() T cell engagement and synapse formation in vivo. Nat Immunol 3:918–925.
26. Belz GT, et al. (2004) Distinct migrating and nonmigrating dendritic cell populations
are involved in MHC class I-restricted antigen presentation after lung infection with
virus. Proc Natl Acad Sci USA 101:8670– 8675.
27. Baumgarth N, Kelso A (1996) Functionally distinct T cells in three compartments of the
respiratory tract after influenza virus infection. Eur J Immunol 26:2189–2197.
28. Steinman RM, Hemmi H (2006) Dendritic cells: translating innate to adaptive immunity.
Curr Top Microbiol Immunol 311:17–58.
29. Allan RS, et al. (2006) Migratory dendritic cells transfer antigen to a lymph node-
resident dendritic cell population for efficient CTL priming. Immunity 25:153–162.
30. Budde K, et al. (2006) FTY720 (fingolimod) in renal transplantation. Clin Transplant 20
Suppl 17:17–24.
31. Mandala S, et al. (2002) Alteration of lymphocyte trafficking by sphingosine-1-
phosphate receptor agonists. Science 296:346–349.
32. Sanna MG, et al. (2004) Sphingosine 1-phosphate (S1P) receptor subtypes S1P1 and
S1P3, respectively, regulate lymphocyte recirculation and heart rate. J Biol Chem
279:13839–13848.
33. Pinschewer DD, et al. (2000) FTY720 immunosuppression impairs effector T cell pe-
ripheral homing without affecting induction, expansion, and memory. J Immunol
164:5761–5770.
34. Lan YY, et al. (2005) The sphingosine-1-phosphate receptor agonist FTY720 modulates
dendritic cell trafficking in vivo. Am J Transplant 5:2649–2659.
35. Maeda Y, et al. (2007) Migration of CD4 T cells and dendritic cells toward sphingosine
1-phosphate (S1P) is mediated by different receptor subtypes: S1P regulates the functions
of murine mature dendritic cells via S1P receptor type 3. J Immunol 178:3437–3446.
36. Moyron-Quiroz JE, et al. (2006) Persistence and responsiveness of immunologic mem-
ory in the absence of secondary lymphoid organs. Immunity 25:643–654.
37. Han S, et al. (2004) FTY720 suppresses humoral immunity by inhibiting germinal center
reaction. Blood 104:4129– 4133.
38. GeurtsvanKessel CH, et al. (2008) Clearance of influenza virus from the lung depends
on migratory langerinCD11b- but not plasmacytoid dendritic cells. J Exp Med
205:1621–1634.
Marsolais et al. PNAS
February 3, 2009
vol. 106
no. 5
1565
MICROBIOLOGY
... Of these, S1P has been implicated in the modulation of cellular processes, including those important for inflammation and immune responses (Singer et al. 2005;Maeda et al. 2007;Hammad et al. 2008;Keul et al. 2011). S1P binding to its receptor (S1PR1) on the endothelial cell surface is a regulator of influenza virusinduced cytokine storm (Marsolais et al. 2009;Teijaro et al. 2011;Walsh et al. 2011) and can limit the migration of effector lymphocytes to immunologic injury areas, down-modulate the number of virus-specific T cells, blunt cytokine/chemokine expression and reduce the supply of innate inflammatory cells (Marsolais et al. 2009;Walsh et al. 2011). Activation of S1PR1 signaling by endogenous S1P inhibits type-I IFN responses. ...
... Of these, S1P has been implicated in the modulation of cellular processes, including those important for inflammation and immune responses (Singer et al. 2005;Maeda et al. 2007;Hammad et al. 2008;Keul et al. 2011). S1P binding to its receptor (S1PR1) on the endothelial cell surface is a regulator of influenza virusinduced cytokine storm (Marsolais et al. 2009;Teijaro et al. 2011;Walsh et al. 2011) and can limit the migration of effector lymphocytes to immunologic injury areas, down-modulate the number of virus-specific T cells, blunt cytokine/chemokine expression and reduce the supply of innate inflammatory cells (Marsolais et al. 2009;Walsh et al. 2011). Activation of S1PR1 signaling by endogenous S1P inhibits type-I IFN responses. ...
Article
Full-text available
Lipid Droplets (LDs) contribute to key pathways important for the physiology and pathophysiology of cells. In a homeostatic view, LDs regulate the storage of neutral lipids, protein sequestration, removal of toxic lipids and cellular communication, however, recent advancements in the field show these organelles as essential for various cellular stress response mechanisms, including inflammation and immunity, with LDs acting as hubs that integrate metabolic and inflammatory processes. The accumulation of LDs has become a hallmark of infection, and is often thought to be virally driven, however recent evidence is pointing to a role for the upregulation of LDs in the production of a successful immune response to viral infection. The fatty acids housed in LDs are also gaining interest due to the role that these lipid species play during viral infection, and their link to the synthesis of bioactive lipid mediators which have been found to have a very complex role in viral infection. This review explores the role of LDs and their subsequent lipid mediators during viral infections and poses a paradigm shift in thinking in the field; whereby LDs may play pivotal roles in protecting the host against viral infection.
... The proportion of lung vs blood-derived antigen-specific producing B cells was also increased upon multiple airway antigen challenges [43], supporting the possibility that systemic responses may yield quantitatively normal antibody responses in the lung, given enough time. Our finding that mLNs of mice with CD19-driven S1P 1 deletion feature MSS-specific antibody-producing B cells supports this notion and aligns with the fact that pharmacologically-induced sequestration of lymphocytes in lymphoid organs does not compromise airway pathogen-specific antibody responses [44,45]. We propose that, in mice with CD19driven S1P 1 deletion, circulating MSS-specific antibodies combined with unaltered T cell responses are likely sufficient for HP pathogenesis. ...
Article
Full-text available
Background Hypersensitivity pneumonitis (HP) is an interstitial lung disease characterized by antigen-triggered neutrophilic exacerbations. Although CD4 ⁺ T cells are sufficient for HP pathogenesis, this never translated into efficient T cell-specific therapies. Increasing evidence shows that B cells also play decisive roles in HP. Here, we aimed to further define the respective contributions of B and T cells in subacute experimental HP. Methods Mice were subjected to a protocol of subacute exposure to the archaeon Methanosphaera stadmanae to induce experimental HP. Using models of adoptive transfers of B cells and T cells in Rag1-deficient mice and of B cell-specific S1P 1 deletion, we assessed the importance of B cells in the development of HP by evaluating inflammation in bronchoalveolar lavage fluid. We also aimed to determine if injected antibodies targeting B and/or T cells could alleviate HP exacerbations using a therapeutic course of intervention. Results Even though B cells are not sufficient to induce HP, they strongly potentiate CD4 ⁺ T cell-induced HP‑associated neutrophilic inflammation in the airways. However, the reduction of 85% of lung B cells in mice with a CD19-driven S1P 1 deletion does not dampen HP inflammation, suggesting that lung B cells are not necessary in large numbers to sustain local inflammation. Finally, we found that injecting antibodies targeting B cells after experimental HP was induced does not dampen neutrophilic exacerbation. Yet, injection of antibodies directed against B cells and T cells yielded a potent 76% inhibition of neutrophilic accumulation in the lungs. This inhibition occurred despite partial, sometimes mild, depletion of B cells and T cells subsets. Conclusions Although B cells are required for maximal inflammation in subacute experimental HP, partial reduction of B cells fails to reduce HP-associated inflammation by itself. However, co-modulation of T cells and B cells yields enhanced inhibition of HP exacerbation caused by an antigenic rechallenge.
... In these mice, increased levels of monocyte chemoattractant protein-1 (MCP-1) and TNF-α in the lungs enabled a reduced viral burden [73,76]. In other studies, both MCP-1 and TNF-α helped in the protection against IV, but were associated with lung immunopathology if over produced [59,[79][80][81][82]. Interestingly, the induction of pulmonary TNF-α prior to IV infection actually reduces lung inflammation and damage [75]. ...
Article
GM-CSF acts as a pro-inflammatory cytokine and a key growth factor produced by several immune cells such as macrophages and activated T cells. In this review, we discuss recent studies that point to the crucial role of GM-CSF in the immune response against infections. Upon induction, GM-CSF activates four main signalling networks including the JAK/STAT, PI3K, MAPK, and NFκB pathways. Many of these transduction pathways such as JAK/STAT signal via proteins commonly activated with other antiviral signalling cascades, such as those induced by IFNs. GM-CSF also helps defend against respiratory infections by regulating alveolar macrophage differentiation and enhancing innate immunity in the lungs. Here, we also summarize the numerous clinical trials that have taken advantage of GM-CSF’s mechanistic attributes in immunotherapy. Moreover, we discuss how GM-CSF is used as an adjuvant in vaccines and how its activity is interfered with to reduce inflammation such as in the case of COVID-19. This review brings forth the current knowledge on the antiviral actions of GM-CSF, the associated signalling cascades, and its application in immunotherapy.
... In this study, leukocyte accumulation in the lung was dependent on cytokine and chemokine production and mortality was mitigated following treatment with S1P analogs. [81][82][83][84] Recent experimentation using influenza has implicated interferon regulatory factor-5 (IRF-5) as essential for the generation of the cytokine storm. 85 It is notable that TLR7 can directly activate MyD88 and that IRF5 signaling is a downstream target of MyD88. ...
Article
Full-text available
The single stranded RNA virus SARS-CoV-2 has caused a massive addition to the already leading global cause of mortality, viral respiratory tract infections. Characterized by and associated with early and deleteriously enhanced production of pro-inflammatory cytokines by respiratory epithelial cells, severe COVID-19 illness has the potential to inflict acute respiratory distress syndrome and even death. Due to the fast spreading nature of COVID-19 and the current lack of a vaccine or specific pharmaceutical treatments, understanding of viral pathogenesis, behavioral prophylaxis, and mitigation tactics are of great public health concern. This review article outlines the immune response to viral pathogens, and due to the novelty of COVID-19 and the large body of evidence suggesting the respiratory and immune benefits from regular moderate intensity exercise, provides observational and mechanistic evidence from research on other viral infections that suggests strategically planned exercise regimens may help reduce susceptibility to infection, while also mitigating severe immune responses to infection commonly associated with poor COVID-19 prognosis. We propose that regular moderate intensity exercise should be considered as part of a combinatorial approach including widespread hygiene initiatives, properly planned and well-executed social distancing policies, and use of efficacious facial coverings like N95 respirators. Studies discerning COVID-19 pathogenesis mechanisms, transfer dynamics, and individual responses to pharmaceutical and adjunct treatments are needed to reduce viral transmission and bring an end to the COVID-19 pandemic.
... SP1 analogs have the potential to inhibit virus multiplication as well as to decrease the intensity of the cytokine storm. 21,36 ...
Article
Full-text available
Despite the recent announcement of promising drug candidates to treat COVID-19, there is currently no effective antiviral drug or vaccine. There is strong evidence that acute lung injury/acute respiratory distress syndrome (ALI/ARDS), likely triggered by a cytokine storm, is responsible for the severity of disease seen in COVID-19 patients. In support of this hypothesis, pilot studies using IL-6 receptor inhibitors such as tocilizumab have shown promising results. Therefore, the use of drugs or cocktails of drugs with broader ability to inhibit these cytokine receptors is likely to be effective. In this article, we propose the use of sphingosine analogues, which have been shown to mitigate acute lung damage in animal models of ALI/ARDS, as early adjuvant therapies to prevent and/or mitigate the cytokine response in COVID-19 patients. This proposal is based on the ability of these drugs to decrease the production of IL-6 and other cytokines. The potential application of fingolimod (FTY720), the oldest sphingosine analogue approved for the treatment of multiple sclerosis, in the early stages of COVID-19 is discussed in more detail as a prototype drug.
Article
Extensive vascular leakage and shock is a major cause of dengue‐associated mortality. At present, there are no specific treatments available. Sphingolipid pathway is a key player in the endothelial barrier integrity; and is mediated through the five sphingosine‐1‐phosphate receptors (S1PR1‐S1PR5). Signaling through S1PR2 promotes barrier disruption; and in Dengue virus (DENV)‐infection, there is overexpression of this receptor. Fingolimod (FTY720) is a specific agonist that targets the remaining barrier‐protective S1P receptors, without targeting S1PR2. In the present study, we explored whether FTY720 treatment can alleviate DENV‐induced endothelial hyperpermeability. In functional assays, in both in vitro systems and in AG129 animal models, FTY720 treatment was found effective. Upon treatment, there was complete restoration of the monolayer integrity in DENV serotype 2‐infected human microvascular endothelial cells (HMEC‐1). At the molecular level, the treatment reversed activation of the S1P pathway. It significantly reduced the phosphorylation of the key molecules such as PTEN, RhoA, and VE‐Cadherin; and also, the expression levels of S1PR2. In DENV2‐infected AG129 mice treated with FTY720, there was significant improvement in weight gain, in overall clinical symptoms, and in survival. Whereas 100% of the DENV2‐infected, untreated animals died by day‐10 post‐infection, 70% of the FTY720‐treated animals were alive; and at the end of the 15‐day post‐infection observation period, 30% of them were still surviving. There was a significant reduction in the Evan's‐blue dye permeability in the organs of FTY720‐treated, DENV‐2 infected animals; and also improvement in the hemogram, with complete restoration of thrombocytopenia and hepatic function. Our results show that the FDA‐approved molecule Fingolimod (FTY720) is a promising therapeutic intervention in severe dengue.
Conference Paper
Full-text available
Introduction: Recent evidence suggests that in patients infected with COVID-19, lung severity should be quantifed by segments prior to bedside evaluations of the patient’s respiratory mechanics and alveolar recruitment capacity, knowing what their characteristics are and relationship after an alveolar recruitment maneuver with PEEP titration in a world population is insufcient. Objectives: To compare the characteristics of the alveolar recruitment potential based on ventilatory mechanics and segmental pulmonary severity by chest tomography after a pulmonary recruitment maneuver with PEEP titration, in patients with SARS Cov-2 on admission to the ICU. Methods: The study corresponds to a descriptive cross-sectional cohort design. A total of 42 patients (69% men, with 38% presenting more than 2 chronic diseases, age 51.4 [42.3 -63.0] years), were assigned into two groups prior to evaluating the inspiratory recruitability ratio ( R/I ratio), high recruitment potential group (HRPG) (n=21) and low recruitment potential group (LRPG) (n=21), was compared with the assessment of severity index by chest tomography (CT- SS) and ventilatory mechanics after a pulmonary recruitment maneuver with PEEP titration. The sample was expressed as median /interquartile range/ or frequency (percentage) as appropriate. For continuous variables with normal and non-normal distribution, we used the Student’s t-test and the Mann–Whitney test. For comparisons of categorical variables we used a chi-square test or Fisher’s exact test. respectively. Spearman’s correlation coefcient (rho) was calculated to compare the relationship of the variables. All data analyzes were performed with R software. Statistical signifcance was considered when p<0.05. Results: Patients with HRP have lower airway opening pressure (p=0.029), higher PEEP (p=0.027), lower driving pressure (p=0.037) and lower mean pressure (p=0.016), when evaluating CT – SS show signifcantly less severity in anterior (p=0.003) and apical (p= <0.001) segments of the right lung and anterior (p=0.029), apical (p=0.043), superior (p=0.009) and anterior basal segments (p=0.006) left lung compared with LRP. When correlating ventilatory mechanics (LRP (rho=-0.116) vs HRP (rho=0.431)) and pulmonary severity LRP (rho=0.093) vs LRP (rho=0.113)) as a function of recruitment potential, we found no signifcant relationship. Conclusion: Patients who have HPRG present better ventilatory mechanics and less pulmonary severity in relation to patients with LPRG, after a pulmonary recruitment maneuver with PEEP titration, allowing to know the characteristics and ranges of the Chilean population for the R/I ratio, with safe method that can be used at the patient’s bedside.
Chapter
Full-text available
The resilience of our immune system is remarkable. It is always on guard against various pathogens that we encounter whether we eat, work, or sleep. A healthy body needs a balanced immune system to combat a variety of disease-causing agents. More importantly, the harmonious cross-talk between the immune system and other systems of our body is the key to good health. When this multi-level system fails us, we become prone not only to germs and diseases but also to many autoimmune disorders and cancers. Immunomodulators are substances that assist in the proper functioning of the immune system directly or indirectly by turning down some proteins (immune suppressors) or turning up others (immune enhancers). Among different classes of immunomodulators, cytokines play an important role as messenger molecules that effectively connect different processes and regulate immune cell growth and their maturation and responsiveness. They are the group of small glycosylated polypeptides with molecular mass less than 30 kDa and are key mediators in innate and adaptive immunity, hematopoiesis, inflammation, tumorigenesis, viral pathogenesis, etc. The chapter here deals with cytokines and their action as immunomodulators at different levels of the immune system of our body and their role in immunotherapies.KeywordsInterleukinsInterferonLymphokinesChemokines
Article
Full-text available
This study systematically review knowledge about the mechanisms of formation of an inflammatory reaction under the influence of biological, physical, and chemical factors, their similarities and differences, and possible methods of pharmacological correction of pathological conditions associated with excessive activation. The effect of adverse environmental factors, such as biological, physical, and chemical factors, causes a systemic response, which is aimed at maintaining homeostasis and is caused, among other things, by a coordinated reaction of the immune system. Phlogogenic agents result in the activation and regulation of the inflammatory response, which is formed by cellular and humoral components of innate immunity. The activation of innate immunity is characterized by a rapid host response, which diminishes following the elimination of foreign invaders, endogenous killer cells, and neogenesis. Depending on the nature of the active factors (biopathogens, allergens, toxins, ionizing radiation, etc.), the mechanisms of immune response arousal have unique features mainly originating from the differences in the recognition of specific molecular patterns and danger signals by different receptors. However, inflammatory mediators and inflammatory response patterns at the systemic level are largely similar even under widely different triggers. Inflammation, having evolved as an adaptive reaction directed at the immune response, can lead to the development of chronic inflammation and autoimmune diseases due to a mismatch in mechanisms of its control. A failure in the regulation of the inflammatory process is the excessive activation of the immune system, which leads to the cytokine release syndrome (hypercytokinemia, or cytokine storm) and can cause self-damage (destruction) of tissues, multiple-organ failure, sepsis, and even death. Modern advances in the study of the pathogenetic bases of the inflammatory response are suggested, such as pharmacological correction using pattern recognition receptor antagonists, pro-inflammatory cytokine inhibitors, or blocking of key control genes or signaling pathways.
Article
Full-text available
Sphingosine-1-phosphate receptor 1 (S1PR1) plays a crucial role in infectious diseases. Targeting S1PR1 provides protection against pathogens, such as influenza viruses. This study is aimed at investigating S1PR1 in response to bacterial infection by assessing S1PR1 expression in S. aureus-infected mice. A rodent local muscle bacterial infection model was developed by injecting S. aureus to the lower hind limb of Balb/c mice. The changes of S1PR1 expression in response to bacterial infection and blocking treatment were assessed using ex vivo biodistribution and in vivo positron emission tomography (PET) after intravenous injection of an S1PR1-specific radiotracer [¹⁸F]TZ4877. The specificity of [¹⁸F]TZ4877 was assessed using S1PR1-specific antagonist, NIBR-0213, and S1PR1-specific DsiRNA pretreated the animals. Immunohistochemical studies were performed to confirm the increase of S1PR1 expression in response to infection. Ex vivo biodistribution data showed that the uptake of [¹⁸F]TZ4877 was increased 30.6%, 54.3%, 74.3%, and 115.3% in the liver, kidney, pancreas, and thymus of the infected mice, respectively, compared to that in normal control mice, indicating that S1PR1 is involved in the early immune response to bacterial infection. NIBR-0213 or S1PR1-specific DsiRNA pretreatment reduced the tissue uptake of [¹⁸F]TZ4877, suggesting that uptake of [¹⁸F]TZ4877 is specific. Our PET/CT study data also confirmed that infected mice have increased [¹⁸F]TZ4877 uptake in several organs comparing to that in normal control mice. Particularly, compared to control mice, a 39% increase of [¹⁸F]TZ4877 uptake was observed in the infected muscle of S. aureus mice, indicating that S1PR1 expression was directly involved in the inflammatory response to infection. Overall, our study suggested that S1PR1 plays an important role in the early immune response to bacterial infection. The uptake of [¹⁸F]TZ4877 is tightly correlated with the S1R1 expression in response to S. aureus infection. PET with S1PR1-specific radiotracer [¹⁸F]TZ4877 could provide a noninvasive tool for detecting the early S1PR1 immune response to infectious diseases. 1. Introduction Staphylococcus aureus (S. aureus) is a commensal human microbiota with approximately 30% of the human population which is colonized with S. aureus [1]. S. aureus often causes skin and soft tissue infections but can also cause pneumonia, bloodstream infections, endocarditis, and osteomyelitis [2–4]. To date, with the majority of studies focused on virus infection, particularly influenza, only little is known about the role of sphingosine-1-phosphate (S1P) and S1P receptors (S1PRs) in response to bacterial infection. Igawa and colleagues demonstrated that the expression of S1PR2 was elevated in S. aureus-treated human epidermal keratinocytes; they also showed that both S1PR1 and S1PR2 controlled the proinflammatory cytokine expression and secretion during the S. aureus invasion [5]. S1P binds to specific G protein-coupled S1P receptors 1-5 and triggers the S1P/S1PR pathway which plays a regulatory role in many pathophysiological processes, including angiogenesis [6], neurodegeneration [7], and immune response [8]. Intriguingly, it has been reported that S1P signaling via S1PRs is related to various aspects of inflammatory cell function. Accumulated evidence suggests that the S1P/S1PR pathway is impaired during infectious diseases. Furthermore, the S1P/S1PR pathway also plays an important role in infection-induced sepsis [9–11]. As a consequence, the S1P/S1PR pathway has been widely accepted as a therapeutic target in inflammatory and infection diseases [12]. Of note, several multiple sclerosis (MS) disease-modifying therapies targeting S1P/S1PRs are under investigation for treating coronavirus disease-2019 (COVID-19) [13, 14]. Out of the five S1PRs, S1PR1 is the most abundant S1P receptor and is widely studied in different diseases. It is ubiquitously expressed among tissues and is heavily involved in immune cell regulation. S1PR1 has been found to mediate functions in most immune cells and therefore plays a curial role in both innate and adaptive immune responses [15]. Previous studies suggest that S1PR1 is essential for lymphocyte recirculation by regulating lymphocyte egress from both the thymus and peripheral lymphoid organs [16, 17]; activation of S1PR1 inhibits the migration of T lymphocytes into different lymphatics and results in retention of T lymphocytes in nonlymphoid peripheral tissues [18]. S1PR1 also regulates the migration of B lymphocytes and osteoclasts [19, 20]. It was reported that S1PR1 plays an important role in the immune response to infectious diseases by regulating recruitment and trafficking of innate immune cells, macrophage polarization [21], and dendritic cell functions [22]. In addition, S1PR1 is considered to play a critical role in the development of sepsis [11]; for example, S1PR1-specific agonist SEW2871 can successfully protect against renal injury in a sepsis model [23]. Numerous studies show that S1PR1 expression or activation is tightly correlated with the inflammatory response to infectious diseases such as Newcastle disease virus infection [24], influenza A virus H1N1 [25], H9N2 infection [26], herpes simplex virus type 1 infection [27], Pseudomonas aeruginosa lung infection [28], and human immunodeficiency viruses HIV-1 infection [29]. It is also believed that host-directive therapy using modulators of S1PR1 and other S1PRs may be an effective treatment against severe infectious diseases, such as severe acute respiratory syndrome (SARS) and COVID-19 [30]. Despite the current understanding of S1PR1 in normal lymphocyte trafficking, the precise mechanisms of S1PR1 activation in response to pathogen-derived pathological infectious consequences remain not clear. Our previous positron emission tomography (PET) studies with S1PR1-specific tracer demonstrated an increased expression of S1PR1 in rodent models of vascular inflammation and neuroinflammation [31, 32]. To evaluate the ability of S1PR1-specific tracer for the assessment of other inflammatory diseases, as well as to further understand the role of S1PR1 in response to pathogen infection, we performed ex vivo biodistribution, microPET, and immunohistochemistry staining studies to investigate S1PR1 expression on S. aureus-infected mice using our recently reported S1PR1-specific radiotracer [ [18]F]TZ4877 [33–35]. In our studies, we observed an increased uptake of [¹⁸F]TZ4877 in several organs in response to the infection; a 39% increase of [¹⁸F]TZ4877 uptake was also observed at the local hind limb muscle infected site. Our results indicated that the infection-induced increase of S1PR1 expression is tightly correlated with pathogen-derived inflammation. Our findings are consistent with other studies that S1PR1 plays a critical role in response to pathogen infection. 2. Materials and Methods 2.1. Animals All animal experiments were conducted by following the Guidelines for the Care and Use of Research Animals under a research protocol approved by Washington University Institutional Animal Care and Use Committee (IACUC). Biodistribution and microPET/CT studies were conducted in Preclinical Imaging Facility at Washington University School of Medicine in St. Louis. For infection model, mice were injected with bacteria or PBS under 2–3% isoflurane in O2. For biodistribution study, mice were euthanized by cervical dislocation under anesthesia with 2–3% isoflurane in O2 and then, tissues of interest were collected. For microPET/CT studies, mice were euthanized by CO2 inhalation after use. 2.2. Radiosynthesis S1PR1-specific radiotracer [¹⁸F]TZ4877 was used in this study in order to detect changes of S1PR1 in response to S. aureus infection. The synthesis and quality control of [¹⁸F]TZ4877 were achieved as previously reported (Figure 1(a)) [33]. For each batch, the synthesis of [¹⁸F]TZ4877 was accomplished with a radiochemical purity of >99%, chemical purity of >95%, and molar activity of >48 GBq/μmol (decay corrected to the end of synthesis, EOB). (a)
Article
Full-text available
Avian influenza A (H5N1) viruses cause severe disease in humans, but the basis for their virulence remains unclear. In vitro and animal studies indicate that high and disseminated viral replication is important for disease pathogenesis. Laboratory experiments suggest that virus-induced cytokine dysregulation may contribute to disease severity. To assess the relevance of these findings for human disease, we performed virological and immunological studies in 18 individuals with H5N1 and 8 individuals infected with human influenza virus subtypes. Influenza H5N1 infection in humans is characterized by high pharyngeal virus loads and frequent detection of viral RNA in rectum and blood. Viral RNA in blood was present only in fatal H5N1 cases and was associated with higher pharyngeal viral loads. We observed low peripheral blood T-lymphocyte counts and high chemokine and cytokine levels in H5N1-infected individuals, particularly in those who died, and these correlated with pharyngeal viral loads. Genetic characterization of H5N1 viruses revealed mutations in the viral polymerase complex associated with mammalian adaptation and virulence. Our observations indicate that high viral load, and the resulting intense inflammatory responses, are central to influenza H5N1 pathogenesis. The focus of clinical management should be on preventing this intense cytokine response, by early diagnosis and effective antiviral treatment.
Article
Full-text available
Virus-immune cytotoxic T cells can inhibit effectively growth of vaccinia virus in acutely infected target cells in vitro by destroying infected target cells before infectious virus progeny is assembled. Together with the fact that virus-specific T cells are demonstrable after 3 days, very early during infection, and with strong circumstantial evidence from adoptive transfer models in vivo, these data suggest that in some virus infections T cells may in fact act cytolytically in vivo to prevent virus growth and spread and be an important early antiviral effector mechanism.
Article
Full-text available
In the adaptive immune response to most viruses, both the cellular and humoral arms of the immune system play complementary roles in eliminating virus and virus-infected cells and in promoting recovery. To evaluate the relative contribution of CD4+ and CD8+ effector T lymphocytes in virus clearance and recovery, we have examined the host response to lethal type A influenza virus infection in B lymphocyte-deficient mice with a targeted disruption in the immunoglobulin mu heavy chain. Our results indicate that naive B cell-deficient mice have a 50- 100-fold greater susceptibility to lethal type A influenza virus infection than do wild type mice. However, after priming with sublethal doses of influenza, immune B cell-deficient animals show an enhanced resistance to lethal virus infection. This finding indicates that an antibody-independent immune-mediated antiviral mechanism accounts for the increased resistance to lethal virus challenge. To assess the contribution of influenza-specific CD4+ and CD8+ effector T cells in this process, defined clonal populations of influenza-specific CD4+ and CD8+ effector T cells were adoptively transferred into lethally infected B cell-deficient mice. Cloned CD8+ effectors efficiently promoted recovery from lethal infection, whereas cloned CD4+ T cells conferred only partial protection. These results suggest that memory T lymphocytes can act independently of a humoral immune response in order to confer resistance to influenza infection in immune individuals. The potential implications of these results for vaccination against human influenza infection are discussed.
Article
Full-text available
The ability of influenza virus to evade immune surveillance by neutralizing antibodies (Abs) directed against its variable surface antigens provides a challenge to the development of effective vaccines. CD8+ cytotoxic T lymphocytes (CTLs) restricted by class I major histocompatibility complex molecules are important in establishing immunity to influenza virus because they recognize internal viral proteins which are conserved between multiple viral strains. In contrast, protective Abs are strain-specific. However, the precise role of effector CD8+ CTLs in protection from influenza virus infection, critical for understanding disease pathogenesis, has not been well defined. In transgenic mice with a very high frequency of antiinfluenza CTL precursors, but without protective Abs, CD8+ CTLs conferred protection against low dose viral challenge, but exacerbated viral pathology and caused mortality at high viral dose. The data suggest a dual role for CD8+ CTLs against influenza, which may present a challenge to the development of effective CTL vaccines. Effector mechanisms used by CD8+ CTLs in orchestrating clearance of virus and recovery from experimental influenza infection, or potentiation of lethal pathology, are discussed.
Article
Full-text available
We describe a new reverse-genetics system that allows one to efficiently generate influenza A viruses entirely from cloned cDNAs. Human embryonic kidney cells (293T) were transfected with eight plasmids, each encoding a viral RNA of the A/WSN/33 (H1N1) or A/PR/8/34 (H1N1) virus, flanked by the human RNA polymerase I promoter and the mouse RNA polymerase I terminator-together with plasmids encoding viral nucleoprotein and the PB2, PB1, and PA viral polymerases. This strategy yielded >1 x 10(3) plaque-forming units (pfu) of virus per ml of supernatant at 48 hr posttransfection. The addition of plasmids expressing all of the remaining viral structural proteins led to a substantial increase in virus production, 3 x 10(4)-5 x 10(7) pfu/ml. We also used reverse genetics to generate a reassortant virus containing the PB1 gene of the A/PR/8/34 virus, with all other genes representing A/WSN/33. Additional viruses produced by this method had mutations in the PA gene or possessed a foreign epitope in the head of the neuraminidase protein. This efficient system, which does not require helper virus infection, should be useful in viral mutagenesis studies and in the production of vaccines and gene therapy vectors.
Article
Full-text available
FTY720 (2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol hydrochloride) prolongs survival of solid organ allografts in animal models. Mechanisms of FTY720 immunomodulation were studied in mice infected with lymphocytic choriomeningitis virus (LCMV) to assess T cell responses or with vesicular stomatitis virus to evaluate Ab responses. Oral FTY720 (0.3 mg/kg/day) did not affect LCMV replication and specific CTL and B cells were induced and expanded normally. Moreover, the anti-viral humoral immune responses were normal. However, FTY720 treatment showed first a shift of overall distribution of CTL from the spleen to peripheral lymph nodes and lymphocytopenia was observed. This effect was reversible within 7-21 days. Together with unimpaired T and B cell memory after FTY720 treatment, this finding rendered enhancement of lymphocyte apoptosis by FTY720 in vivo unlikely. Secondly, the delayed-type hypersensitivity reaction to a viral MHC class I-presented peptide was markedly reduced by FTY720. These results were supported by impaired circulation of LCMV specific TCR transgenic effector lymphocytes in the peripheral blood and reduced numbers of tissue infiltrating CTL in response to delayed-type hypersensitivity reaction. Thirdly, in a CD8+ T cell-mediated diabetes model in a transgenic mouse expressing the LCMV glycoprotein in the islets of the pancreas, FTY720 delayed or prevented disease by reducing islet-infiltrating CTL. Thus, FTY720 effectively reduced recirculation of CD8+ effector T cells and their recruitment to peripheral lesions without affecting the induction and expansion of immune responses in secondary lymphoid organs. These properties may offer the potential to treat ongoing organ-specific T cell-mediated immunopathologic disease.
Article
Diss. med. Zürich (kein Austausch). Literaturverz. Sonderdr. aus: The journal of immunology ; 164 (11), (2000), S. 5761-5770.
Article
This study aimed to resolve, firstly, whether T cell responses induced in one tissue site are similar to those induced by the same antigen in another site and, secondly, whether influenza virus infection induces one predominant type of T cell response locally in the respiratory tract. To address these questions, T cell responses in three compartments of the respiratory tract were compared after infection of mice with a sublethal dose of influenza virus: the draining mediastinal lymph nodes (MLN), the lung parenchyma and the airways. Each compartment harbored a T cell response substantially different from that found at the other sites. A preferential accumulation of ex vivo-cytolytic CD8+ T cells was found in the airways (CD4/CD8 ratio 1:2) and to a lesser extent in the lung parenchyma (CD4/CD8 ratio 1:1). T cells from both compartments expressed high levels of various cytokine mRNA, but showed differences in their respective expression pattern, with those from lung tissue showing particularly high levels of IFN-gamma mRNA. The response in the draining lymph nodes, on the other hand, was dominated by CD4+ T cells (CD4/CD8 ratio 2:1) with a higher proliferative capacity (after TCR/CD3 cross-linking) and which provided better B cell help in vitro than CD4+ T cells isolated from lung tissue. T cells from MLN expressed mRNA for a variety of cytokines with only low levels of IFN-gamma mRNA and they showed no CTL activity ex vivo. These functional differences were not due to differences in the kinetics of the response, or to the higher frequencies of activated T cells in lung tissue and airways compared to MLN, since the differences remained when cell-sorter-purified activated (CD18hi, CD44hi) T cells from MLN and lung tissue were compared in a time-course study. Taken together, these findings indicate that pathogens such as influenza virus induce a heterogenous set of T cell responses in different tissue sites affected by the infection.
Article
Eighteen cases of human influenza A H5N1 infection were identified in Hong Kong from May to December 1997. Two of the six fatal cases had undergone a full post-mortem which showed reactive hemophagocytic syndrome as the most prominent feature. Other findings included organizing diffuse alveolar damage with interstitial fibrosis, extensive hepatic central lobular necrosis, acute renal tubular necrosis and lymphoid depletion. Elevation of soluble interleukin-2 receptor, interleukin-6 and interferon-gamma was demonstrated in both patients, whereas secondary bacterial pneumonia was not observed. Virus detection using isolation, reverse transcription-polymerase chain reaction and immunostaining were all negative. It is postulated that in fatal human infections with this avian subtype, initial virus replication in the respiratory tract triggers hypercytokinemia complicated by the reactive hemophagocytic syndrome. These findings suggest that the pathogenesis of influenza A H5N1 infection might be different from that of the usual human subtypes H1-H3.