You are on page 1of 23

Report

Autophagy Is Required for Neutrophil-Mediated


Inflammation
Graphical Abstract Authors
Abhisek Bhattacharya, Qin Wei, Jin Na
Shin, ..., Diana L. Bonilla, Qian Xiang, N.
Tony Eissa

Correspondence
teissa@bcm.edu

In Brief
Using several mouse models,
Bhattacharya et al. identify autophagy as
an important regulator of neutrophil
degranulation and ROS production by
NADPH oxidase. The authors find that
autophagy is required for inflammatory
activity of neutrophils.

Highlights
d Autophagy regulates neutrophil degranulation

d Autophagy regulates NADPH-oxidase-mediated reactive


oxygen species production

d Autophagy deficiency reduces inflammatory activity of


neutrophils

Bhattacharya et al., 2015, Cell Reports 12, 1731–1739


September 22, 2015 ª2015 The Authors
http://dx.doi.org/10.1016/j.celrep.2015.08.019
Cell Reports

Report

Autophagy Is Required
for Neutrophil-Mediated Inflammation
Abhisek Bhattacharya,1,2 Qin Wei,1 Jin Na Shin,1 Elmoataz Abdel Fattah,1 Diana L. Bonilla,1 Qian Xiang,1
and N. Tony Eissa1,2,*
1Department of Medicine
2Department of Pathology and Immunology
Baylor College of Medicine, Houston, TX 77030, USA
*Correspondence: teissa@bcm.edu
http://dx.doi.org/10.1016/j.celrep.2015.08.019
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).

SUMMARY secretory vesicles contain several important molecules, in-


cluding b2-integrins (CD11b/CD18) and complement receptors,
Autophagy, an intracellular degradation and en- which play an essential role in neutrophil migration and activation
ergy recycling mechanism, is emerging as an im- (Amulic et al., 2012). Upon activation, these granules are mobi-
portant regulator of immune responses. However, lized, fuse with phagosomes or plasma membrane, and release
the role of autophagy in regulating neutrophil func- their contents in the respective environments. Secondary gran-
tions is not known. We investigated neutrophil ules are of interest, as the membranes of these granules contain
flavocytochrome b558, a component of the NADPH oxidase ma-
biology using myeloid-specific autophagy-deficient
chinery (Amulic et al., 2012). Thus, fusion of secondary granules
mice and found that autophagy deficiency re-
with phagosomes or plasma membrane results in formation of a
duced neutrophil degranulation in vitro and in vivo. functional NADPH oxidase complex, which then produces reac-
Mice with autophagy deficiency showed reduced tive oxygen species (ROS). Degranulation, a process of regu-
severity of several neutrophil-mediated inflammatory lated exocytosis of granules, is one of the major mechanisms
and autoimmune disease models, including PMA- of inflammatory response by neutrophils. However, the underly-
induced ear inflammation, LPS-induced breakdown ing regulatory pathways and their respective roles remain incom-
of blood-brain barrier, and experimental autoimmune pletely understood.
encephalomyelitis. NADPH oxidase-mediated reac- Autophagy plays important roles in the immune responses and
tive oxygen species generation was also reduced abnormalities in this pathway have been linked to several dis-
in autophagy-deficient neutrophils, and inhibition of eases (Levine et al., 2011). More recently, autophagy has been
implicated in regulating secretion (Cadwell et al., 2008; Patel
NADPH oxidase reduced neutrophil degranulation,
et al., 2013; Ushio et al., 2011; Bhattacharya et al., 2014b),
suggesting NADPH oxidase to be a player at the
though the underlying mechanisms and in vivo relevance are
intersection of autophagy and degranulation. Over- not fully understood. The role of autophagy in neutrophil func-
all, this study establishes autophagy as an important tions remains largely unexplored.
regulator of neutrophil functions and neutrophil- Here, we show that deficiency of autophagy reduced degran-
mediated inflammation in vivo. ulation of neutrophils. Autophagy-deficient neutrophils had re-
duced NADPH oxidase-mediated ROS generation. Moreover,
inhibition of NADPH oxidase reduced neutrophil degranulation,
INTRODUCTION
implicating NADPH oxidase in mediating effects of autophagy
on neutrophil degranulation. The in vivo relevance of these find-
Neutrophils play a critical role as one of the first lines of innate
ings was shown in the context of autoimmune and inflammatory
immune defense. Activated neutrophils migrate to the site of
processes. These findings establish autophagy as an important
inflammation and control microbes by phagocytosis, formation
regulator of neutrophil functions and suggest that targeting auto-
of neutrophil extracellular traps, and secretion of antimicro-
phagy pathway could have therapeutic value during infection,
bials, stored in specialized granular compartments. Neutro-
inflammation, or neutrophil-mediated autoimmune diseases.
phils possess at least four types of granules. Among these,
primary, secondary, and tertiary granules develop from the
endoplasmic reticulum (ER)-Golgi network, whereas secretory RESULTS
vesicles have an endocytic origin (Borregaard, 1997). Primary
(azurophilic) granules contain myeloperoxidase (MPO), b-glucu- Deficiency of Autophagy Reduced Neutrophil
ronidase, elastase, and other antimicrobial factors, whereas Degranulation
lactoferrin and matrix metalloprotease-9 (MMP-9) are stored To evaluate the role of autophagy in neutrophils, we generated
in secondary (specific) granules. Tertiary (gelatinase) granules mice with myeloid-specific deletion of Atg7 (called Atg7MD mice
also contain MMP-9 but lack lactoferrin. The membranes of hereafter) by breeding mice bearing floxed Atg7 alleles (Atg7f/f

Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors 1731
Figure 1. ATG7 Deficiency Reduced Neutrophil Degranulation
(A) Representative immunoblot of ATG7 using lysates from purified peritoneal macrophages (PM), bone marrow-derived macrophages (BMDM), spleen-derived
macrophages (SpDM), and bone marrow neutrophils (BMN). Lanes marked f/f and MD denote lysates from control and Atg7MD mice, respectively.
(B) BMN from control or Atg7MD mice were primed with cytochalasin B and then stimulated with 1 mM fMLF for 15 or 30 min. Gelatinase zymography was
performed on supernatants and on lysates from unstimulated BMN; n = 6/group. Densitometry analysis and a representative image, in which each lane or symbol
represents one mouse, are shown.
(C) Lysates from unstimulated BMN, and supernatant collected following cytochalasin B and 1 mM fMLF treatment of Atg7f/f and Atg7MD neutrophils were used for
immunoblotting by lactoferrin antibody. n = 6–9/group. Each lane or symbol represents one mouse.
(D) BMN from control or Atg7MD mice were stimulated as above. Supernatant was collected and incubated overnight with p-nitrophenyl-b-D-glucuronide (for b-D-
glucuronidase). OD values were measured at 405 nm.
(E and F) BMN were stimulated with cytochalasin B and 1 mM fMLF, 1 mg/ml LPS, or 1 mg/ml PMA for 15 min and evaluated by flow cytometry using antibodies
against Ly6G and CD11b (E) or LAMP-1 (F). n = 6/group. Each symbol represents one mouse.
Data are mean ± SEM; *p < 0.05, **p < 0.01, ***p < 0.001. MFI, mean fluorescence intensity.

mice) with transgenic LysM-cre mice (Abdel Fattah et al., 2015; ATG7, and the efficiency of genetic deletion was comparable to
Bonilla et al., 2013; Shin et al., 2013). Bone marrow neutrophils that observed in peritoneal macrophages or macrophages gener-
(BMNs) from Atg7MD mice showed substantial reduction of ated from bone marrow or spleen from Atg7MD mice (Figure 1A).

1732 Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors
Figure 2. ATG5 Deficiency Reduced Neutro-
phil Degranulation
BMN from control or Atg5MD mice were primed
with cytochalasin B and then stimulated with 1 mM
fMLF for 15 or 30 min.
(A and B) Supernatant was collected, incubated for
15 min with tetramethyl-benzidine to determine
levels of MPO (A), or incubated overnight with
p-nitrophenyl-b-D-glucuronide to determine b-D-
glucuronidase (B).
(C) In addition, supernatant was used for immu-
noblotting by lactoferrin antibody. One represen-
tative blot in which each lane corresponds to one
mouse and densitometry analysis is shown.
(D) Gelatinase zymography was performed on
supernatant, and densitometry analysis is shown.
For all panels, each symbol represents one mouse.
n = 6/group. Data are mean ± SEM; *p < 0.05, **p <
0.01.

secondary, and tertiary granules from


BMNs (Figure 2). These data suggested
that autophagy was required for neutro-
phil degranulation.
We then wanted to determine whether
there was a global dysfunction in auto-
phagy-deficient neutrophils. We have
previously shown that zymosan phago-
As autophagy has been recently implicated in the secretory cytosis was not altered in autophagy-deficient macrophages
pathway, we hypothesized that autophagy deficiency would (Bonilla et al., 2013). To test whether autophagy deficiency
affect neutrophil degranulation. Among the granules origi- altered zymosan uptake by neutrophils, we incubated BMN
nating from ER-Golgi apparatus, tertiary granules are the first from control or Atg7MD mice with FITC-labeled zymosan A
to release their contents upon activation. To test the effect of and checked phagocytosis by flow cytometry. There was no
autophagy on degranulation of tertiary granules, we stimu- significant difference in zymosan A uptake, or surface levels
lated BMN with cytochalasin B and bacterial tripeptide of CD11b, between control and Atg7MD neutrophils (Figures
fMLF, and used the supernatant for gelatinase zymography. S1A and S1B). However, consistent with our previous findings,
Loss of ATG7 reduced the levels of MMP-9 secreted after we observed substantial reduction of MMP-9 in the super-
fMLF treatment (Figure 1B). Gelatinase zymography, per- natant collected after incubation of zymosan A with Atg7MD
formed on the lysates of unstimulated BMN, showed that neutrophils (Figure S1C). Thus, the defect in neutrophil degran-
baseline levels of MMP-9 in Atg7MD neutrophils were para- ulation in Atg7MD mice was not part of a global dysfunction in
doxically elevated (Figure 1B), suggesting that the reduction these cells.
in MMP-9 secretion was due to retention, rather than a defect
in its synthesis. Reduced In Vivo Neutrophil-Mediated Inflammation in
To determine whether autophagy was involved in the secre- Atg7MD Mice
tion of other types of neutrophil granules, we stimulated We hypothesized that autophagy deficiency would reduce
BMN with cytochalasin B and fMLF and evaluated the secre- severity of neutrophil-mediated inflammatory diseases in vivo.
tion of b-D-glucuronidase (stored in primary granules) and To test this hypothesis, we utilized a model of neutrophil-medi-
lactoferrin (stored in secondary granules). Deficiency of ated PMA-induced ear inflammation (Gross et al., 2009). Atg7MD
ATG7 reduced degranulation from both primary and second- mice showed substantial reduction in ear swelling (Figure 3A).
ary granules (Figures 1C and 1D). In contrast, upregulation After PMA application, degranulation of MPO, detectable by
of CD11b in response to various stimuli remained unaltered in vivo bioluminescence imaging, requires fusion of primary
in Atg7MD neutrophils (Figure 1E). Secretory lysosomes were granules with phagosomes (Gross et al., 2009). Neutrophil
also unaffected, as evidenced by similar LAMP-1 levels in MPO in Atg7MD mice was substantially reduced (Figure 3B).
ATG7-deficient and control neutrophils following PMA stimula- These data suggested that autophagy deficiency reduced
tion (Figure 1F). degranulation of primary granules by reducing their fusion with
To rule out autophagy-independent roles of Atg7 in neutrophil phagosomes.
degranulation, we generated mice with myeloid cell-specific We then extended our findings to another in vivo model of
deletion of Atg5 (Atg5MD mice) (Abdel Fattah et al., 2015). We inflammation. Intraperitoneal LPS injection increases the perme-
found that deletion of Atg5 reduced degranulation of primary, ability of blood-brain barrier, an effect dependent on MMP-9 (Aid

Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors 1733
Figure 3. Reduced Neutrophil-Mediated
Inflammation in Atg7MD Mice In Vivo
(A and B) Ear inflammation was induced by topical
application of PMA for 24 hr, and ear thickness
was then measured; n = 7–8/group (A). Biolumi-
nescence imaging was performed. A representa-
tive image and quantitative analysis are shown (B).
Similar symbols denote matched pairs. n = 7/
group.
(C) LPS was injected intra-peritoneally (5 mg/kg, at
0 and 48 hr), and blood-brain barrier permeability
was measured.
(D) BMN were isolated from control and Atg7MD
mice and stimulated with 1 mg/ml LPS for 30 min,
and supernatant was subjected to gelatinase zy-
mography. A representative zymogram, in which
each lane denotes one mouse, and quantitative
analyses are shown. n = 6/group.
(E and F) Peritoneal exudate cells were collected
4 hr after thioglycollate injection. Neutrophils were
isolated and stained with Ly6G (E) and CD11b (F).
(G) Blood was collected by cardiac puncture, and
complete blood count was performed.
(H) BMN were isolated and counted. For all panels,
each symbol represents one mouse.
Data are mean ± SEM; *p < 0.05, **p < 0.01. MFI,
mean fluorescence intensity.

et al., 2010; Bennett et al., 2010). After LPS administration, Autophagy Deficiency in Myeloid Cells Reduced
we observed that Atg7MD mice showed substantially reduced Severity of Experimental Autoimmune
permeability of the blood-brain barrier (Figure 3C). Moreover, Encephalomyelitis
MMP-9 release, following stimulation of BMN with LPS, was To further test the inflammatory potential of autophagy-deficient
also reduced from autophagy-deficient neutrophils (Figure 3D). neutrophils, we used experimental autoimmune encephalomy-
These data suggested that autophagy-deficiency reduced over- elitis (EAE), an autoimmune model, which mimics several as-
all inflammatory potential of neutrophils. pects of multiple sclerosis disease in humans. Neutrophils
have been implicated as a critical mediator of CNS inflammation
Increased Circulating Neutrophils in Atg7MD Mice during the effector phase of EAE and neutrophil depletion at
To rule out defects in neutrophil migration, we used a peritonitis different phases of EAE inhibits or abolishes EAE (McColl et al.,
model in which peritoneal exudate cells were collected 4 hr after 1998; Rumble et al., 2015; Steinbach et al., 2013). Secretion of
intraperitoneal thioglycollate injection. Surprisingly, peritoneal proteases, such as MMP-9, by neutrophils facilitates breakdown
neutrophils were increased in Atg7MD mice, though no differ- of blood-CNS barrier during EAE (Opdenakker et al., 2001). We
ences in CD11b levels were observed in these neutrophils (Fig- found that, after EAE induction by active immunization, both
ures 3E and 3F). In vitro transwell migration assay showed no control and Atg7MD mice showed 100% incidence of EAE. How-
gross defect in Atg7MD neutrophil migration in response to ever, the severity of EAE was lower in Atg7MD mice (Figure 4A).
different stimuli (Figure S1D). Complete blood analysis of Atg7MD Additionally, weight loss and cumulative disease score (area
mice and their littermate controls showed similar total number of under the curve) were attenuated in Atg7MD mice (Figures 4B
white blood cells (Figure S2), but total and differential neutrophil and 4C).
counts were higher in the Atg7MD mice (Figures 3G and S2). Consistent with the roles of MMP-9 in breakdown of blood-
The increase in circulating neutrophils in Atg7MD mice was CNS barrier, we observed an increase in MMP-9 in spinal cord
likely the result of increased neutrophil content in the bone lysates, correlating with severity of EAE (Figure S3A). Previous
marrow of Atg7MD mice (Figure 3H). These data suggested that reports showed that blood-spinal cord barrier, rather than the
myeloid-specific autophagy deficiency increased the number blood-brain barrier, was predominantly affected in mouse model
of circulating neutrophils and, consequently, their recruitment of EAE (Bennett et al., 2010). At the peak of EAE, we observed
to the site of inflammation. However, because of their reduced reduced amount of Evan’s blue dye in the spinal cord, but not
effector functions, these neutrophils could not efficiently mediate in organs without permeability barrier, in Atg7MD mice (Figures
inflammation. 4D and S3B). Consistent with attenuation of breakdown of

1734 Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors
Figure 4. Autophagy Deficiency in Myeloid
Cells Reduced Severity of EAE
(A–C) EAE was induced in Atg7MD mice and
littermate controls, and daily clinical scores (A),
daily weight changes (B), and cumulative disease
scores (C) were recorded. n = 12–13/group.
(D) Evaluation of brain and spinal cord barrier
permeability at day 16 after EAE induction.
(E) Mice were sacrificed at day 16 after EAE in-
duction, and CNS-infiltrating cells were isolated by
Percoll gradient.
(F and G) EAE was induced in Atg5MD mice and
littermate controls, and daily clinical scores (F) and
cumulative disease scores (G) were monitored. n =
10–12/group.
(H) Contact hypersensitivity was induced in
Atg7MD and control mice by sensitization and
challenge with Oxazolone (OXA) or DNFB. Ear
swelling was measured 24 hr after challenge. Each
symbol represents one mouse. AUC, area under
the curve.
Data are mean ± SEM; *p < 0.05, **p < 0.01.

We also ruled out any gross defects in


T cell compartments in Atg7MD mice
that might account for the phenotype
observed (Figures S4C–S4J). Moreover,
we found that use of neutrophil-depleting
1A8 antibody, but not isotype control
blood-CNS barrier permeability, we found a significant reduction antibody, after the onset of EAE abolished the difference be-
in inflammatory cells in the CNS of the Atg7MD mice at the peak tween Atg7f/f and Atg7MD mice in disease severity, cumulative
of EAE (Figure 4E). Atg5MD mice also showed a significant reduc- disease score, and weight loss (Figure S5). Taken together, we
tion in severity and cumulative disease score of EAE compared concluded that the protective effect in the Atg7MD mice was
to their littermate controls (Figures 4F and 4G). Six out of ten caused by defective neutrophil function and not by changes in
Atg5f/f mice reached score 5 compared to one out of 12 Atg5MD peripheral T cell activation.
mice (*p = 0.02, chi-square test). Thus, deficiency of either Atg5
or Atg7 in myeloid cells resulted in attenuated EAE phenotype. Autophagy Deficiency Led to Reduced NADPH Oxidase-
Finally, to rule out global immunosuppression in these mice, Mediated ROS Production
we utilized models of hapten-induced contact hypersensitivity Flavocytochrome b558, a component of the NADPH oxidase ma-
(CHS). CHS is mediated primarily by CD8 T cells (Vocanson chinery, is present on the membrane of secondary granules of
et al., 2009). The severity of hapten-induced CHS in response neutrophils (Jesaitis et al., 1990). Therefore, NADPH oxidase-
to Oxazolone or DNFB remained undiminished in Atg7MD mice mediated ROS production in neutrophils depends on fusion of
(Figure 4H). Thus, the reduction in severity of EAE in Atg7MD granules with plasma membrane/phagosome. Our in vivo data
mice was not due to a global reduction in immune functions. suggested a fusion defect between granules and phagosome
as a cause of reduced degranulation. To further examine how
Peripheral Antigen-Specific T Cell Generation in EAE autophagy affected ROS generation by NADPH oxidase, we stim-
Was Unaltered in Atg7MD Mice ulated control or Atg7MD BMN with fMLF and measured extracel-
To rule out reduced peripheral T cell activation in Atg7MD mice, lular ROS generation by cytochrome c reduction assay. Atg7MD
we examined MOG-specific T cell proliferation at different neutrophils showed significant reduction in ROS production in
phases of EAE. There was no significant reduction in antigen- response to fMLF (Figure 5A). Similarly, FcOxyburst assay also re-
specific T cell proliferation in spleen of Atg7MD mice at any of vealed reduced phagosomal ROS production by Atg7MD neutro-
the doses or time points tested (Figure S4A). To test whether phils (Figure 5B). Similar results were found in Atg5MD BMN, in
macrophage-mediated antigen presentation was defective in which both phagosomal ROS generation and extracellular ROS
Atg7MD mice, we used DOBW cells, which recognize oval- generation, measured by OxyBURST Green H2 HFF BSA, were
bumin323–339 peptide in the context of MHC-II and produce IL- substantially reduced (Figures 5C and 5D). We did not find any
2 upon proliferation (Harding et al., 1991). We could not detect gross difference with respect to fMLF-induced F-actin organiza-
any significant differences in antigen presentation between resi- tion between control and Atg7MD neutrophils (data not shown).
dent peritoneal macrophages from control and Atg7MD mice Collectively, our data suggested that autophagy deficiency re-
(Figure S4B). duced NADPH oxidase-mediated ROS production by neutrophils.

Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors 1735
Figure 5. Autophagy Deficiency Reduced NADPH Oxidase-Mediated ROS Production in BMN
(A) BMN from control or Atg7MD mice were incubated with cytochrome c and extracellular ROS generation in response to 1 mM fMLF was monitored continuously
by measuring changes in absorbance (OD550). Data are from four independent experiments.
(B) BMN from control or Atg7MD mice were incubated with 125 mg/ml FcOxyburst reagent, and phagosomal ROS generation was measured by flow cytometry. A
representative plot and quantitative analyses are shown. Each symbol represents one mouse. Data are from three independent experiments.
(C) BMN from control or Atg5MD mice were incubated with OxyBURST H2HFF Green BSA, and extracellular ROS generation in response to 1 mM fMLF was
monitored by measuring changes in fluorescence. Time point 0 is the first time point measured after addition of fMLF. One representative curve for each genotype
and quantitative analyses are shown. n = 4/genotype.
(D) BMN from control or Atg5MD mice were incubated with 125 mg/ml FcOxyburst reagent, and phagosomal ROS generation was measured by flow cytometry. A
representative plot and quantitative analyses are shown. Each symbol represents one mouse. n = 4/genotype.
(E–H) BMN from wild-type mice were pre-treated with DPI (10 mM, 1 hr) and then stimulated with cytochalasin B and fMLF. Supernatant was collected and
b-glucuronidase was measured; n = 8/group. Each pair of symbols represents one mouse (E). Supernatant was also subjected to immunoblotting using lactoferrin
antibody (F) or gelatinase zymography (G). Representative immunoblots and zymogram are shown. (H) Supernatant was analyzed colorimetrically for LDH activity
to detect cell death.
Data are representative of three independent experiments and presented as mean ± SEM; *p < 0.05, **p < 0.01. AUC, area under the curve.

We then investigated the effects of NADPH oxidase-mediated functions of neutrophils remain incompletely understood. In
ROS generation on neutrophil degranulation. We pre-treated this regard, the current study provided several important findings
BMN from wild-type mice with vehicle only or with diphenyle- and established autophagy as a regulator of neutrophil func-
neiodonium chloride (DPI, 10 mM, 1 hr), a specific inhibitor of tions. It showed that inhibition of autophagy reduced degranula-
NADPH oxidase, and then stimulated these neutrophils with tion from neutrophils. Such reduction was not due to decreased
cytochalasin B and fMLF. Treatment with DPI reduced degranu- production of proteins but rather to a secretion defect. Moreover,
lation of primary, secondary, and tertiary granules and did not deficiency of either Atg5 or Atg7 led to similar defects in de-
alter cell viability (Figures 5E–5H). Taken together, these data granulation from neutrophils, suggesting a role of autophagy,
showed that autophagy deficiency led to reduced NADPH oxi- rather than autophagy-independent ATG functions, in mediating
dase-mediated ROS production and reduced ROS production degranulation from neutrophils. The in vivo relevance of these
by NADPH oxidase further contributed to reduced degranulation findings was confirmed using several mouse models of neutro-
from Atg7MD neutrophils. phil-mediated inflammatory and autoimmune processes, which
showed that autophagy deficiency reduced the inflammatory
DISCUSSION potential of neutrophils.
Autophagy deficiency produced maximal effects on degran-
Despite extensive characterization of neutrophil phagocytic ulation of tertiary and secondary granules. In contrast, we
activity and granules, the underlying mechanisms governing did not find any defect in upregulation of CD11b, suggesting

1736 Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors
intact degranulation of secretory vesicles in absence of auto- bone-marrow neutrophils in Atg7MD mice. We have previously
phagy. In neutrophils, secretory vesicles degranulate immedi- shown increased proliferative activity of leukocytes in bone
ately following mild stimulation. Tertiary and secondary granules marrow of Atg7MD mice (Abdel Fattah et al., 2015). Moreover,
follow secretory vesicles, in that order, and primary granules are ROS are critical for several neutrophil death mechanisms
the most difficult to mobilize. It is possible that the differential and reduced ROS generation in Atg7MD neutrophils may
effect of autophagy deficiency on different granules is a direct also contribute to better survival (Geering and Simon, 2011).
consequence of ease of mobilizing these granules. Together, these factors could explain higher numbers of circu-
We predominantly used fMLF, which is among the strongest lating neutrophils in Atg7MD mice.
inducers of neutrophil degranulation. It is possible that fMLF in- Emerging evidence suggests that neutrophils play a broad role
duction masked any potentially small defect in degranulation of in the immune system (Kumar and Sharma, 2010). Neutrophils
secretory vesicles. However, defective degranulation is unlikely have been implicated in autoimmunity and as one of the major
to be specific for fMLF, as both LPS and zymosan resulted in effector cells in EAE (Rumble et al., 2015). Autophagy has also
reduced MMP-9 release by autophagy-deficient neutrophils. been implicated in several autoimmune diseases (Bhattacharya
Thus, our data suggested an intrinsic problem in the degranula- and Eissa, 2013). In this regard, we found that autophagy defi-
tion mechanism in absence of autophagy. ciency in myeloid cells reduced severity of EAE. Reduced
Another important finding in this study is the role of NADPH ox- severity of EAE in myeloid-specific autophagy deficient mice
idase and ROS in mediating effects of autophagy on degranula- was not due to impairment of peripheral antigen-specific T cell
tion. Our data are consistent with a recent report, which showed activation. These results are consistent with our recent findings
similar effects of autophagy in mucus secretion by goblet cells supporting dendritic cells (DCs) as a major antigen presenting
(Patel et al., 2013). However, the potential roles of NADPH oxi- cells during EAE (Bhattacharya et al., 2014a). LysMCre-medi-
dase and ROS in mediating neutrophil degranulation remain ated deletion predominantly affects macrophages and neutro-
incompletely elucidated. Studies by different groups using neu- phils, whereas DCs remain largely unaffected (Clausen et al.,
trophils isolated from patients of chronic granulomatous dis- 1999). Normal peripheral T cell activation may reflect unaltered
ease, in which NADPH oxidase-mediated ROS generation is functions of DCs in these mice. Autophagy-deficient neutrophils
impaired, provided contrasting observations showing impaired, showed reduced secretion of MMP-9, a matrix metalloprotei-
augmented, or unaltered degranulation (Baehner et al., 1969; nase involved in breakdown of blood-CNS barrier in EAE.
Gold et al., 1974; Pak et al., 2007). Such a wide spectrum of re- MMP-9 is considered a potential therapeutic target in multiple
sults could be attributed to demography of patients studied, sclerosis and clinical trials with MMP-9 inhibitors are currently
types of stimuli used, methods employed, and granular contents under way (Muroski et al., 2008). Reduced severity of EAE in
measured as an index of degranulation. Although, in our study, Atg7MD mice correlated with reduced permeability of blood-spi-
inhibition of NADPH oxidase resulted in significant reduction nal cord barrier and reduced CNS cellular infiltration. Although
of degranulation, the effects were modest compared to that our studies utilized well-characterized neutrophil-mediated
observed in autophagy-deficient neutrophils, suggesting that in vivo models, they do not exclude possible roles of other
additional factors contributed to the effect of autophagy on myeloid cells in these models.
degranulation. In summary, this study revealed an important role of auto-
Assembly of NADPH oxidase requires fusion of secondary phagy in neutrophils during inflammatory and autoimmune
granules, the membrane of which contains b558 subunit, with diseases. Neutrophils constitute the major portion of circu-
phagosomal or plasma membrane (Jesaitis et al., 1990). Auto- lating leukocytes in humans and are considered the first line
phagy proteins are important for fusion of enzyme-containing of cellular defense and major inflammatory mediator in the
lysosomes with plasma membrane in osteoclasts (DeSelm immune system. Neutrophilic secretion is required not only
et al., 2011). The role of autophagy in mediating granule fusion during infection but also to maintain normal homeostasis.
is also observed in our study, using in vivo model of PMA- Increased neutrophilic secretion has been implicated in the
induced ear inflammation. In this model, bioluminescence im- pathogenesis of diseases such as chronic obstructive lung
aging of neutrophilic MPO by luminol depends on proper fusion disease and cystic fibrosis. Recent studies proposed alter-
of MPO-containing granules with phagosome (Gross et al., ation of the autophagy pathway as a therapeutic approach
2009). We found substantial reduction in MPO bioluminescence to multiple diseases. Our study uncovered autophagy as a
from the PMA-treated ear of Atg7MD mice, suggesting a poten- major regulator of neutrophil function and suggested the pos-
tial defect in fusion of these granules with phagosome. Thus, sibility that targeting the autophagy pathway might offer an
our study supports a model in which autophagy mediates important approach to treat neutrophil-mediated inflammatory
both fusion of granules with other membrane compartments processes.
and degranulation. Autophagy-mediated granule fusion helps
in the assembly of an active NADPH oxidase complex and EXPERIMENTAL PROCEDURES
NADPH oxidase-mediated ROS generation, in turn, further fa-
cilitates degranulation. Mice
C57BL/6 and LysM-Cre transgenic mice in a C57BL/6 background were pur-
Despite normal transwell migration in response to different
chased from Jackson Laboratory. Atg7MD and Atg5MD mice have been previ-
stimuli, Atg7MD mice showed increased neutrophils in the perito- ously described (Bonilla et al., 2013; Shin et al., 2013; Abdel Fattah et al.,
neal cavity following thioglycollate administration. This finding 2015). All mice used were housed in BCM vivarium (biosafety level 2). All animal
was likely the result of a higher number of circulating and protocols were approved by institutional boards of Baylor College of Medicine.

Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors 1737
In Vivo Disease Models SUPPLEMENTAL INFORMATION
EAE and contact CHS models were induced as described previously (Bhatta-
charya et al., 2014a). For EAE, active immunization was performed with Supplemental Information includes Supplemental Experimental Procedures
MOG35–55 peptide emulsified in Freund’s complete adjuvant, along with two and five figures and can be found with this article online at http://dx.doi.org/
doses of pertussis toxin. Mice were monitored daily for weight changes and 10.1016/j.celrep.2015.08.019.
clinical signs (Supplemental Experimental Procedures). CHS was induced by
sensitizing and challenging the mice with Oxazolone or DNFB. Ear thickness ACKNOWLEDGMENTS
was measured before and 24 hr after the challenge. Acute dermatitis was
induced by applying 20 mg of PMA in 20 ml of DMSO to one ear and DMSO We thank Dr. Clifford Harding for DOBW cells. We acknowledge current and
only (vehicle) to the other ear, and ear swelling was measured after 24 hr. Biolu- former members of the N.T.E. lab for stimulating discussions and technical
minescence imaging was performed as previously described (Gross et al., assistance. This study was supported by the National Heart, Lung, and Blood
2009). In brief, mice were anaesthetized by isoflurane and imaging was per- Institute and by the Cytometry and Cell Sorting Core at Baylor College of Med-
formed in a Xenogen In Vivo Imaging System (IVIS), 10 min after intraperitoneal icine with funding from the NIH (AI036211, CA125123, and RR024574) and the
injection of luminol. Quantitation was done by software (Living Image In Vivo expert assistance of Joel M. Sederstrom.
Imaging Software, PerkinElmer).
Received: March 6, 2015
Isolation of Neutrophils from Bone Marrow or Peritoneal Cavity Revised: May 7, 2015
Single-cell suspension from bone marrow, after ACK lysis, was subjected to Accepted: August 6, 2015
gradient centrifugation in 60%/80% Percoll, and neutrophils were collected Published: September 3, 2015
from the interface (Halpert et al., 2011), washed in HBSS without Ca2+ and
Mg2+, and stained for flow cytometry or used for in vitro stimulation. Peritoneal
REFERENCES
lavage was performed with 10 ml of ice-cold HBSS without Ca2+ and Mg2+ 4 hr
after injection of 1 ml thioglycollate, and neutrophils were isolated by Percoll
Abdel Fattah, E., Bhattacharya, A., Herron, A., Safdar, Z., and Eissa, N.T.
gradient.
(2015). Critical role for IL-18 in spontaneous lung inflammation caused by auto-
phagy deficiency. J. Immunol. 194, 5407–5416.
BMN Stimulation and Assessing Degranulation
Isolated BMN were allowed to settle for 20–30 min at 37 C, 5% CO2 and then Aid, S., Silva, A.C., Candelario-Jalil, E., Choi, S.H., Rosenberg, G.A., and
pre-treated for 5 min with 10 mg/ml of cytochalasin B. BMN were then stimu- Bosetti, F. (2010). Cyclooxygenase-1 and -2 differentially modulate lipopoly-
lated with 1 mM fMLF, 1 mg/ml of LPS or PMA, or 10 ng/ml of GM-CSF for saccharide-induced blood-brain barrier disruption through matrix metallopro-
various time points. In some experiments, neutrophils were pre-treated for teinase activity. J. Cereb. Blood Flow Metab. 30, 370–380.
2 hr with 50 mM CQ and then cytochalasin B was added for 5 min, followed Amulic, B., Cazalet, C., Hayes, G.L., Metzler, K.D., and Zychlinsky, A. (2012).
by fMLF stimulation. Supernatant was collected by centrifugation and cells Neutrophil function: from mechanisms to disease. Annu. Rev. Immunol. 30,
were prepared for flow cytometry. MMP-9 zymography was performed using 459–489.
Novex 10% Zymogram (Gelatin) Gel. For MPO detection, supernatant was
Baehner, R.L., Karnovsky, M.J., and Karnovsky, M.L. (1969). Degranulation of
incubated with TMB reaction mixture (1 mg of 3,30 ,5,50 -tetramethylbenzidine
leukocytes in chronic granulomatous disease. J. Clin. Invest. 48, 187–192.
in 1 ml of DMSO and 9 ml of 0.1 M NaH2PO4 (pH 5.5) with 2 ml of fresh 30%
hydrogen peroxide) for 15 min (in dark) and OD615 was measured immediately. Bennett, J., Basivireddy, J., Kollar, A., Biron, K.E., Reickmann, P., Jefferies,
For detection of b-glucuronidase, supernatant was incubated overnight with W.A., and McQuaid, S. (2010). Blood-brain barrier disruption and enhanced
3.15 mg/ml p-nitrophenyl-itroglucuronide in 0.17 M sodium acetate buffer vascular permeability in the multiple sclerosis model EAE. J. Neuroimmunol.
(pH 4.0), reaction was terminated by adding 0.4 M glycine (pH 10), and 229, 180–191.
OD405 was measured immediately. Bhattacharya, A., and Eissa, N.T. (2013). Autophagy and autoimmunity cross-
talks. Front. Immunol. 4, 88.
Measurement of ROS Production Bhattacharya, A., Parillon, X., Zeng, S., Han, S., and Eissa, N.T. (2014a). Defi-
Cytochrome c reduction assay and OxyBURST Green H2HFF BSA assay ciency of autophagy in dendritic cells protects against experimental autoim-
were performed to detect extracellular ROS production using 0.5 3 106 to 1 3 mune encephalomyelitis. J. Biol. Chem. 289, 26525–26532.
106 cells/well or 2 3 106 cells/well, respectively in a 96-well plate. Cytochrome
Bhattacharya, A., Prakash, Y.S., and Eissa, N.T. (2014b). Secretory function of
c or OxyBURST Green H2HFF BSA was added (final concentration of
autophagy in innate immune cells. Cell. Microbiol. 16, 1637–1645.
1.5 mg/ml or 10 mg/ml, respectively) to the cells and baseline absorbance/fluo-
rescence was recorded. fMLF was then added (final concentration: 1 mM in Bonilla, D.L., Bhattacharya, A., Sha, Y., Xu, Y., Xiang, Q., Kan, A., Jagannath,
200 ml/well) and absorbance/fluorescence was monitored continuously at C., Komatsu, M., and Eissa, N.T. (2013). Autophagy regulates phagocytosis by
550 nm (absorbance) or at 488 nm excitation and 530 nm emission (fluores- modulating the expression of scavenger receptors. Immunity 39, 537–547.
cence). FcOxyburst assays were performed according to the manufacturer’s Borregaard, N. (1997). Development of neutrophil granule diversity. Ann. N Y
instructions (Invitrogen). In brief, flow cytometric measurement was performed Acad. Sci. 832, 62–68.
using a total of 2 3 106 cells/tube. After baseline reading, FcOxyburst reagent Cadwell, K., Liu, J.Y., Brown, S.L., Miyoshi, H., Loh, J., Lennerz, J.K., Kishi, C.,
was added (125 mg/ml in a final volume of 400 ml), and data were collected contin- Kc, W., Carrero, J.A., Hunt, S., et al. (2008). A key role for autophagy and the
uously with LSR Fortessa (Becton Dickinson). FACSDiva software was used for autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature
data collection, and FlowJo software was used for analysis. Cell viability assays 456, 259–263.
were performed using lactate dehydrogenase (LDH) detection kit (Roche).
Clausen, B.E., Burkhardt, C., Reith, W., Renkawitz, R., and Förster, I. (1999).
Conditional gene targeting in macrophages and granulocytes using LysMcre
Statistical Analysis
mice. Transgenic Res. 8, 265–277.
Normally distributed data were analyzed by Student’s t test (Welch correction
was applied in case of significant difference in variance between the groups), DeSelm, C.J., Miller, B.C., Zou, W., Beatty, W.L., van Meel, E., Takahata, Y.,
and non-parametric comparisons were performed by Mann-Whitney test. For Klumperman, J., Tooze, S.A., Teitelbaum, S.L., and Virgin, H.W. (2011). Auto-
EAE data, curves were compared by ANOVA, daily clinical scores and weight phagy proteins regulate the secretory component of osteoclastic bone resorp-
changes by Student’s t test. Paired t test was used for comparisons between tion. Dev. Cell 21, 966–974.
matched pairs (in vivo bioluminescence and degranulation of primary granules Geering, B., and Simon, H.U. (2011). Peculiarities of cell death mechanisms in
after DPI treatment). Analyses were performed by GraphPad Prism 6. neutrophils. Cell Death Differ. 18, 1457–1469.

1738 Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors
Gold, S.B., Hanes, D.M., Stites, D.P., and Fudenberg, H.H. (1974). Abnormal teinase-9/gelatinase B is a putative therapeutic target of chronic obstructive
kinetics of degranulation in chronic granulomatous disease. N. Engl. J. Med. pulmonary disease and multiple sclerosis. Curr. Pharm. Biotechnol. 9, 34–46.
291, 332–337. Opdenakker, G., Van den Steen, P.E., Dubois, B., Nelissen, I., Van Coillie, E.,
Gross, S., Gammon, S.T., Moss, B.L., Rauch, D., Harding, J., Heinecke, J.W., Masure, S., Proost, P., and Van Damme, J. (2001). Gelatinase B functions as
Ratner, L., and Piwnica-Worms, D. (2009). Bioluminescence imaging of mye- regulator and effector in leukocyte biology. J. Leukoc. Biol. 69, 851–859.
loperoxidase activity in vivo. Nat. Med. 15, 455–461.
Pak, V., Budikhina, A., Pashenkov, M., and Pinegin, B. (2007). Neutrophil ac-
Halpert, M.M., Thomas, K.A., King, R.G., and Justement, L.B. (2011). TLT2 po- tivity in chronic granulomatous disease. Adv. Exp. Med. Biol. 601, 69–74.
tentiates neutrophil antibacterial activity and chemotaxis in response to G pro-
Patel, K.K., Miyoshi, H., Beatty, W.L., Head, R.D., Malvin, N.P., Cadwell, K.,
tein-coupled receptor-mediated signaling. J. Immunol. 187, 2346–2355.
Guan, J.L., Saitoh, T., Akira, S., Seglen, P.O., et al. (2013). Autophagy proteins
Harding, C.V., Collins, D.S., Kanagawa, O., and Unanue, E.R. (1991). Liposome-
control goblet cell function by potentiating reactive oxygen species produc-
encapsulated antigens engender lysosomal processing for class II MHC pre-
tion. EMBO J. 32, 3130–3144.
sentation and cytosolic processing for class I presentation. J. Immunol. 147,
2860–2863. Rumble, J.M., Huber, A.K., Krishnamoorthy, G., Srinivasan, A., Giles, D.A.,
Zhang, X., Wang, L., and Segal, B.M. (2015). Neutrophil-related factors as bio-
Jesaitis, A.J., Buescher, E.S., Harrison, D., Quinn, M.T., Parkos, C.A., Livesey,
markers in EAE and MS. J. Exp. Med. 212, 23–35.
S., and Linner, J. (1990). Ultrastructural localization of cytochrome b in the
membranes of resting and phagocytosing human granulocytes. J. Clin. Invest. Shin, J.N., Fattah, E.A., Bhattacharya, A., Ko, S., and Eissa, N.T. (2013). Inflam-
85, 821–835. masome activation by altered proteostasis. J. Biol. Chem. 288, 35886–35895.
Kumar, V., and Sharma, A. (2010). Neutrophils: Cinderella of innate immune Steinbach, K., Piedavent, M., Bauer, S., Neumann, J.T., and Friese, M.A.
system. Int. Immunopharmacol. 10, 1325–1334. (2013). Neutrophils amplify autoimmune central nervous system infiltrates by
Levine, B., Mizushima, N., and Virgin, H.W. (2011). Autophagy in immunity and maturing local APCs. J. Immunol. 191, 4531–4539.
inflammation. Nature 469, 323–335. Ushio, H., Ueno, T., Kojima, Y., Komatsu, M., Tanaka, S., Yamamoto, A., Ichi-
McColl, S.R., Staykova, M.A., Wozniak, A., Fordham, S., Bruce, J., and Willen- mura, Y., Ezaki, J., Nishida, K., Komazawa-Sakon, S., et al. (2011). Crucial role
borg, D.O. (1998). Treatment with anti-granulocyte antibodies inhibits the for autophagy in degranulation of mast cells. J. Allergy Clin. Immunol. 127,
effector phase of experimental autoimmune encephalomyelitis. J. Immunol. 1267–1276.e6.
161, 6421–6426. Vocanson, M., Hennino, A., Rozières, A., Poyet, G., and Nicolas, J.F. (2009).
Muroski, M.E., Roycik, M.D., Newcomer, R.G., Van den Steen, P.E., Opdenak- Effector and regulatory mechanisms in allergic contact dermatitis. Allergy
ker, G., Monroe, H.R., Sahab, Z.J., and Sang, Q.X. (2008). Matrix metallopro- 64, 1699–1714.

Cell Reports 12, 1731–1739, September 22, 2015 ª2015 The Authors 1739
Cell Reports
Supplemental Information

Autophagy Is Required
for Neutrophil-Mediated Inflammation
Abhisek Bhattacharya, Qin Wei, Jin Na Shin, Elmoataz Abdel Fattah, Diana L. Bonilla,
Qian Xiang, and N. Tony Eissa
Supplemental Data

Figure S1 (Related to figure 1 & 3). Deficiency of Autophagy reduced


degranulation, but did not alter global neutrophil functions. (A-C) BMN from control
or Atg7M∆ mice were incubated with FITC-zymosan A particles for 15 min at 37°C. Flow
cytometric analysis was performed to evaluate phagocytosis of zymosan by BMN (A)
and surface levels of CD11b (B). Supernatants were collected and subjected to
gelatinase zymography (C). Densitometry analysis and a representative zymogram are
shown. Each lane or symbol represents one mouse. N=8-10/group. (D) Transwell
migration assays were performed for 1 hr using BMN and medium (Med) with or without
1 µM fMLF or 10 nM GM-CSF in the lower chamber. N=3/ group. Data are mean ±
SEM; *p <0.05, **p<0.01. MFI, mean fluorescence intensity. NS: Not significant.

  1  
Figure S2 (Related to figure 3). No gross abnormalities in numbers and
percentages of circulating blood cells in Atg7M∆ mice. Blood was collected by
cardiac puncture and analysis was performed by automated analyzer. Total numbers of
red blood cells (RBC), white blood cells (WBC) and platelets are shown (A). Total (B)
and differential (C) counts of circulating blood cells are shown. Data are mean ± SEM.
Each symbol represents one mouse, *p<0.05. LUC, large unstained cells.  

  2  
Figure S3 (Related to figure 4). Characterization of permeability during EAE. (A)
Spinal cord lysates from naïve or mice during EAE were subjected to gelatinase
zymography. Representative image is shown, each lane represents one mouse. N=7-9/
group. (B) Sixteen days after EAE induction (at the peak of EAE), mice were injected
intraperitoneally with Evan’s blue dye for 4 hr and organs were collected and weighed.
Dye was extracted by formamide and quantified by colorimetric assay. Each symbol
represents one mouse. Data are mean ± SEM. NS, not significant.  

  3  
  4  
Figure S4 (Related to figure 4). Myeloid specific deletion of Atg7 did not produce
any gross defect in T cell compartments (A) Single cell suspensions from spleens
were prepared at the peak (day 16) or remission (day 26) phase of EAE and incubated
with the indicated concentration of MOG peptide for 72 hr. Proliferation was measured
by incorporating tritiated thymidine for the last 18 hr of the assay. N=6-7/ group. (B)
Naïve peritoneal macrophages were collected, purified by adherence, stimulated with
LPS (1µg/ ml) for 40 hr and incubated for 24 hr with the indicated concentration of
ovalbumin and DOBW hybridoma cells. Supernatant was collected and assayed for IL-2
by ELISA. N=8-10/ group. (C-F) Spleen and thymus were collected from naïve control or
Atg7M∆ mice. Single cell suspensions were prepared and stained for different T cell
markers followed by intracellular staining for Foxp3. Total cell count (C), percent of
CD3+ cells (D), different T cell compartments in thymus (E) and Treg cells in thymus
and spleen (F) were enumerated. (G) MACS-purified CD4 T cells were stained with
CFSE and stimulated for 5 days with plate bound anti-CD3, anti-CD3 and soluble anti-
CD28 or 2 µg/ml of concanavalin A. Proliferation was measured by flow cytometric
estimation of CFSE dilution. N=3-8/ group. (H-I) Spleens were collected from naïve
control or Atg7M∆ mice. Single cell suspensions were prepared and transwell migration
assays were performed in response to various concentrations of sphingosine-1-
phosphate in the lower chambers of the transwell. T cell populations were identified in
both input cells and migrating cells using flow cytometry. Data are expressed for CD4
(H) and CD8 (I), as chemotactic index (fold increase over medium only). (J) Single cells
suspensions from spleens of control and Atg7-deficient mice were stained with green
and red cell trackers respectively, mixed in equal number and then injected into control
and Atg7-defiicent mice. Mice were sacrificed after 18-20 hr, blood and spleen were
collected and flow cytometry was performed to identify stained cells. N=3/group. Data
are mean ± SEM.  

  5  
Figure. S5 (Related to figure 4). Neutrophil depletion abolished the difference in
severity of EAE between control and Atg7M∆ mice. EAE was induced in mice. After
the onset of EAE, isotype control or neutrophil-depleting (1A8) antibodies were injected
intraperitoneally every alternate day starting from day 10 (A). Daily clinical scores (B),
cumulative scores (C) and weight (D) were monitored. N=4-8/ group, data represent
mean ± SEM. *p <0.05, **p<0.01, NS, not-significant  

  6  
Supplemental experimental procedures

EAE Induction and in vivo treatments  

EAE was induced as described (Bhattacharya A et al., 2014a). Briefly, eight to twelve

week-old mice were actively immunized with 100 µl of 1 mg/ml MOG35-55 peptide (EZ

Biolab) emulsified in Freund’s complete adjuvant (5 mg/ ml of Mycobacterium

tuberculosis extract H37Ra in incomplete Freund’s adjuvant). At and 48 hr after

immunization, all mice received intraperitoneal injections of 500 ng of pertussis toxin

(List Biological Laboratories). Mice were monitored daily for weight changes and clinical

symptoms, which were scored as follows: 0: no overt abnormalities, 1: limp tail or hind

limb weakness, 2: limp tail and hind limb weakness, 3: partial paralysis of hind limbs, 4:

complete hind limb paralysis, 5: moribund or death from EAE. At grade 5, mice were

sacrificed for humane reasons. Disease incidence was marked when an animal showed

clinical signs for at least two consecutive days. Neutrophil depletion experiments were

done 8-10 days after immunization for EAE. Mice were injected intraperitoneally every

other day for 10 days (5 doses) with either 500 µg of 1A8 antibody or 250 µg of the 2A3

isotype control (BioXcell). Investigators scoring the mice for all EAE experiments were

blinded to mice genotype.  

Induction of Contact hypersensitivity

Contact hypersensitivity was induced as described (Bhattacharya A et al., 2014a).

Briefly, mice were sensitized (day 0) by applying 50 µl of 5% (wt/vol) Oxazolone (4-

Ethoxymethylene-2-phenyl-2-oxazolin-5-one) in absolute ethanol, or by applying, on day

0 and day 1, 50 µl of 0.5% DNFB in acetone: olive oil (4:1) to the shaved abdomen.

  7  
Mice were challenged on day 5 by applying 20 µl of 0.2% DNFB on one ear (10 µl on

both sides) or on day 6 by applying 20 µl of 1% Oxazolone on one ear and solvent

alone to the other ear. For mice with ear tags, the tagged ear was always used for

vehicle treatment. Ear thickness was measured before and 24 hours after the challenge

with a micrometer (Mitutoyo) and ear swelling was calculated by using the formula:  

(Challenged ear after- Challenged ear before)- (Vehicle treated ear after-Vehicle treated ear

before).  

In vivo assessment of permeability  

EAE was induced in C57Bl/6 mice and permeability was measured as described

(Bennett J et al., 2010) with minor modifications. Briefly, 0.5% Evans blue dye was

injected intraperitoneally (50-100 mg/ kg body weight) at the peak of the disease (~16

days following immunization). For LPS-induced breakdown of blood-brain barrier, LPS

was injected intraperitoneally (5 mg/kg body weight) at 0 and 48 hr. Evan’s blue dye

was injected 16-20 hr after the second LPS injection. Four hours after Evan’s blue

injection, mice were deeply anaesthetized, perfused through the left ventricle with 30 ml

cold PBS and brain, spinal cord, spleen, a part of the liver (medial lobe) and left kidney

were collected. Organs were weighed and dye was extracted with 1 ml of formamide for

5 days at 37°C. The amount of dye was computed from a standard curve of Evans blue

by spectrophotometric measurement (OD650).  

In vivo competitive homing assay  

Single cell suspension was prepared from spleen mechanically and stained with

CellTrackers Green or Red (Molecular Probes). Equal numbers of labeled cells from

autophagy-deficient and littermate control mice were mixed and injected in mice retro-

  8  
orbitally. Mice were sacrificed after 18-20 hr, spleens were collected, single cell

suspension prepared and flow cytometry was performed to identify red and green cells.  

Isolation of CNS infiltrating lymphocytes  

Mice were sacrificed at the peak of EAE (~16 days after immunization) and

mononuclear cells from brain and spinal cord were isolated by gradient centrifugation

using 37/70% Percoll gradient (GE Healthcare) after digestion with collagenase and

DNase I. Cells were counted and stained for flow cytometric analysis after Fc blocking.

HL-60 culture and neutrophil differentiation  

HL-60 cells were obtained from ATCC. Neutrophil differentiation was performed by

culturing HL-60 cells for 5-7 days in complete RPMI-1640 medium supplemented with

1.3% DMSO. Cells were stimulated with cytochalasin B (10 µg/ ml) and 100 nM fMLF,

and assays for degranulation were performed as described above. For pharmacological

inhibition of class III PI3K pathway, differentiated HL-60 (neutHL-60) cells were treated

with 3-methyladenine (3-MA, 5 mM), LY-294002 (50 µM) or wortmanin (800 nM) for 2 hr

and then stimulated with cytochalasin B and fMLF.  

Culture of macrophages derived from bone marrow or spleen  

Macrophages from bone marrow or spleen were prepared as previously described

(Bonilla et al., 2013). Briefly, single cell suspension from bone marrow was treated with

ACK lysis buffer and cells were cultured for 6-7 days in presence of 10% conditioned

medium from L929 cells. Half of the medium was discarded and fresh medium added

after 3-4 days.  

Zymosan phagocytosis  

  9  
FITC-zymosan A particles (Invitrogen) were added to BMN at a ratio of 1:5-1:10.

Synchronous binding was performed by centrifuging the plates at 100g for 2 min at 4°C.

After washing with ice cold PBS, phagocytosis was initiated by adding pre-warmed

complete RPMI-1640. Plates were incubated for 15 min at 37°C and supernatant was

collected for zymography. Cells were treated with Trypsin-EDTA to remove zymosan

particles adherent to the surface, washed and then stained for flow cytometry.

Complete Blood Count  

Blood was collected into EDTA-coated tubes (BD Microtainer) by cardiac puncture from

deeply anaesthetized mice and complete and differential blood counts were performed

by automated blood analyzer (Advia 120).  

Single cell suspension and flow cytometry  

Single cell suspension from spleen, thymus and lymph nodes were prepared by

mechanical disruption over cell strainer (BD Falcon) followed by RBC lysis using ACK

lysis buffer (Quality Biological, Inc). Intracellular staining for Foxp3 was performed using

Cytofix/Cytoperm Plus (BD) kit according to manufacturer’s instructions. In all neutrophil

experiments for CD11b upregulation, analysis was performed on Ly6G+CD11b+ cells.

The following antibodies were used: CD8-APC (17-0081-81), F4/80APC-eFluor780 (47-

4801-82), all from eBioscience; CD11b-PerCP-Cy5.5 (550993), CD11b-APC-Cy7

(561039), CD4-APC (553051), CD4-PE (553048), Ly6G-PE (551461), CD8-FITC

(553030), CD3e-PE-Cy7 (561100) and Foxp3-PE (560414), all from BD Biosciences.

For F-actin reorganization studies, BMN were stimulated with 1 µM fMLF, fixed with ice

cold 4% paraformaldehyde, stained with AlexaFluor488-phalloidin for intracellular F-

actin and analyzed by flow cytometry. Flow cytometry was performed with LSR Fortessa

  10  
(Becton Dickinson), FACSDiva software was used for data collection and FlowJo

software was used for analysis.  

Transwell migration  

After 3-hr serum-starvation at 37°C, 106 splenocytes were placed on the top well of a 5-

µm polycarbonate insert (Costar) in 100 µl of medium and the bottom wells were set up

with 600 µl medium alone or with various concentrations of Sphingosine-1-phosphate.

After 3-hr incubation at 37°C, cells from the bottom wells were recovered, counted and

stained along with input splenocytes by flow cytometry. Migration was calculated as

percentage of number of input cells and chemotactic index was presented with respect

to the control well with medium only. Migration experiments for splenocytes were

conducted in serum-free RPMI 1640 medium supplemented with 0.5% BSA (fatty-acid

depleted, Sigma).

In transwell migration assay for neutrophils, complete RPMI 1640 medium and 3-µm

polycarbonate insert (Costar) were used with chemoattractants in the bottom wells.

After 1 hr at 37°C, media from the lower chamber were collected, 300 µl of HBSS (Ca2+

and Mg2+ free) containing 5 mM EDTA was added to the wells and the plate was

incubated on ice for 30 minutes. Wells were rinsed with 100 µl HBSS-EDTA and total

volume was made to 1 ml. Cells were counted using Countess automated cell counter

(Invitrogen).

T cell proliferation  

Antigen-specific T cell proliferation was measured by radioactive thymidine

incorporation. Single cell suspensions from spleen or lymph nodes were incubated with

the indicated concentration of MOG35-55 peptide for 3 days and 1 µCi/well of tritiated

  11  
thymidine was incorporated for the last 18 hours and measured by a scintillation

counter. For non-specific stimulation, CFSE dilution (Invitrogen) was used to measure

proliferation of total splenocytes or MACS-purified (Miltenyi) CD4 T cells as per

manufacturer’s instructions. Anti-CD3 and anti-CD28 (both from eBioscience) and

concanavalin A (Sigma) were used for stimulation.  

Antigen presentation assay  

Naïve peritoneal exudate cells were collected with ice cold medium and plated at 3X105

cells/ well in a 96-well plate, macrophages were isolated by adherence and stimulated

with LPS (1µg/ ml) for 40 hr. Antigen presentation to DOBW cells were performed with

Ovalbumin protein (Sigma) as described (Harding et al., 1991).

Immunoblotting  

Cell lysis was performed on ice using RIPA buffer with protease inhibitor cocktail (BD).

Lysates were collected after centrifugation, protein concentrations measured by BCA

protein assay kit (Pierce), boiled in Laemmli sample buffer for 5 minutes at 95°C and

resolved by SDS-PAGE. Following transfer onto a nitrocellulose membrane by Mini

Trans-Blot Cell (Bio-Rad), membranes were incubated with antibodies and imaging was

performed using an Odyssey Infrared Imaging System (LI-COR Bioscience). The

following antibodies were used: anti-ATG7 (600-401-487, Rockland Immunochemicals)

anti--actin (AM43020, Ambion), anti-p62 (03-GP62-C-1, American Research Products)

and anti-lactoferrin (ab77705, abcam).  

  12  

You might also like