You are on page 1of 10

Article

An essential role for Rab27a GTPase in


eosinophil exocytosis
John Dongil Kim,* Lian Willetts,* Sergei Ochkur,† Nutan Srivastava,* Rudolf Hamburg,*
Anooshirvan Shayeganpour,* Miguel C. Seabra,‡ James J. Lee,†,1 Redwan Moqbel,§,1
and Paige Lacy*,1,2
*Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada; †Division of
Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Arizona, USA; ‡Clinical
Neuroscience, Division of Neuroscience and Mental Health, National Heart and Lung Institute, Imperial College London,
London, UK; and §Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
RECEIVED AUGUST 30, 2012; REVISED JULY 31, 2013; ACCEPTED AUGUST 1, 2013. DOI: 10.1189/jlb.0812431

ABSTRACT eosinophils, both in vitro and in vivo. In mouse models,


Eosinophil degranulation has been implicated in inflam- Ashen mice demonstrated reduced EPX release in BAL
matory processes associated with allergic asthma. fluid. These findings suggest that Rab27a has a key role
Rab27a, a Rab-related GTPase, is a regulatory intracel- in eosinophil degranulation. Furthermore, these findings
lular signaling molecule expressed in human eosino- have implications for Rab27a-dependent eosinophil de-
phils. We postulated that Rab27a regulates eosinophil granulation in airway inflammation. J. Leukoc. Biol. 94:
degranulation. We investigated the role of Rab27a in 1265–1274; 2013.
eosinophil degranulation within the context of airway
inflammation. Rab27a expression and localization in eo-
sinophils were investigated by using subcellular frac- Introduction
tionation combined with Western blot analysis, and the Eosinophils are inflammatory granulocytes implicated in
results were confirmed by immunofluorescence analy- asthma and parasitic helminth infections [1– 4]. They are
sis of Rab27a and the granule membrane marker highly granulated white blood cells enriched in cationic gran-
CD63. To determine the function of eosinophil Rab27a, ule proteins that are thought to contribute to tissue damage in
we used Ashen mice, a strain of Rab27a-deficient ani-
inflammatory airway diseases, including atopic asthma [5].
mals. Ashen eosinophils were tested for degranulation
Granule proteins from eosinophils are associated with pheno-
in response to PAF and calcium ionophore by measur-
ing released EPX activity. Airway EPX release was also types of asthma, which include, but are not limited to, hyper-
determined by intratracheal injection of eosinophils into secretion of mucus, increased vascular permeability, bronchial
mice lacking EPX. Rab27a immunoreactivity colocalized epithelial damage, and smooth muscle contraction [4]. In hu-
with eosinophil crystalloid granules, as determined by mans, however, a proinflammatory role for eosinophils in al-
subcellular fractionation and immunofluorescence anal- lergic asthma remains controversial. Early studies using me-
ysis. PAF induced eosinophil degranulation in correla- polizumab, a humanized anti-IL-5 monoclonal antibody, pro-
tion with redistribution of Rab27a⫹ structures, some of posed a lack of efficacy in the asthma patients studied, thus
which colocalized with CD63⫹ crystalloid granules at concluding that eosinophils may not play a direct effector role
the cell membrane. Eosinophils from mice had signifi-
in asthma [6 – 8]. However, 2 studies in 2009, using the same
cantly reduced EPX release compared with normal WT
antibody therapy, indicated that eosinophil ablation signifi-
cantly reduces the number of asthma exacerbations in patients
with sputum eosinophilia [9, 10]. Another 2 studies in eosino-
Abbreviations: BAL⫽bronchoalveolar lavage; bmEos⫽bone marrow– de- phil-deficient mice have supported a proinflammatory role for
rived eosinophils; CIHR⫽Canadian Institutes of Health Research;
eosinophils in asthma [11, 12]. On the basis of these studies
DAPI⫽4=6=diamidino-2-phenylindole; EDN⫽eosinophil-derived neurotoxin;
eos. equiv./microliter⫽eosinophil equivalents per microliter; EPX⫽eosinophil and others, a role for eosinophils in airway hyperresponsive-
peroxidase; GTPase⫽guanosine triphosphatase; FLT3L⫽Fms-related ty- ness and remodeling in atopic asthma continues to be the fo-
rosine kinase 3 ligand; hE⫽human eotaxin; LDH⫽lactate dehydrogenase; cus of intensive research. Furthermore, the precise contribu-
NIH⫽National Institutes of Health; NSF⫽N-ethylmaleimide-sensitive factor;
NWT⫽Northwest Territories; OPD⫽o-phenylenediamine; OVA⫽ovalbumin;
tion of Rab27a to eosinophil effector functions, principally de-
PAF⫽platelet-activating factor; Q-SNARE⫽glutamine-soluble SNARE; granulation, has not been definitively established.
RabGEF⫽Rab guanine nucleotide exchange factor; RPMI⫽Rosewell Park
Medical Institute; R-SNARE⫽arginine -soluble SNARE; SCF⫽stem cell fac-
tor; Slp/Slac2⫽synaptotagmin-like protein (Slp) homologue lacking C2 do- 1. These authors contributed equally to this work.
mains; SNAP⫽soluble NSF attachment protein; SNARE⫽soluble NSF at- 2. Correspondence: Pulmonary Research Group, 559 HMRC, Department of
tachment protein receptor; VAMP⫽ vesicle-associated membrane protein; Medicine, University of Alberta, Edmonton, AB, T6G 2S2, Canada. E-mail:
WT⫽wild-type paige.lacy@ualberta.ca

0741-5400/13/0094-1265 © Society for Leukocyte Biology Volume 94, December 2013 Journal of Leukocyte Biology 1265
In the midst of this ongoing debate, Coughlin et al. [13] (IL-5) [23] and chemotaxis (hE2, including egress from bone
shed new light on a potential effector role for eosinophil marrow) [24]. IL-5/hE2 double-transgenic mice exhibit eosin-
Rab27a in asthma. Rab27a, a Rab-related GTPase, is involved ophilia and spontaneously develop severe asthma-like symp-
in vesicular transport, budding, and movement, upstream of toms with significant airway hyperresponsiveness. Thus, IL-5/
docking and fusion [14]. This secretory GTPase is usually hE2 double-transgenic mice serve as an abundant source of
membrane bound and becomes activated after displacement of spontaneously degranulating eosinophils. In this study, IL-5/
bound GDP and association with GTP by the action of Rab- hE2 mice were crossed with the Ashen strain [18, 19], which
GEFs [15]. A rare, autosomal recessive disorder known as Gris- then provided a Rab27a-deficient, eosinophil-producing
celli syndrome type 2 is caused by a deficiency in Rab27a, model.
characterized by partial albinism and immunodeficiency result-
ing in early childhood death, unless a bone marrow transplant
is performed [16, 17]. This disorder is specifically associated MATERIALS AND METHODS
with deficient exocytosis from immune and other secretory
cells, including melanocytes. In our recent study, the expres- Animals
sion and magnitude of activity of eosinophil Rab27a was com- All animals in this study were on a C57BL/6 background, and C57BL/6
pared between patients with asthma and control subjects. We WT mice were purchased from Jackson Laboratory (Bar Harbor, ME,
USA). NJ.1638 IL-5 [23], hE2 [22], and IL-5/hE2 [22] transgenic mice
reported significantly higher expression and activity of Rab27a
were bred in house. These strains were crossed with Ashen [19] to generate
in eosinophils from those with asthma [13]. From our data, we IL-5/Ashen, hE2/Ashen, and IL-5/hE2/Ashen mice. The Ashen mice pos-
concluded that Rab27a and possibly eosinophil exocytosis are sessed similar numbers of total and differential peripheral blood eosino-
important contributors to an asthma phenotype. phils and leukocytes to that of WT C57BL/6 animals (Fig. 1). The IL-5/
The Ashen mouse strain was used to examine the in vivo rel- hE2 mice were hemizygous for both transgenes and served as controls for
evance of this observation. The Ashen mouse is a Rab27a-defi- the IL-5/hE2/Ashen mice. Eosinophil peroxidase knockout (EPX⫺/⫺) mice
cient strain that produces a truncated Rab27a protein lacking were generated that lacked EPX protein and activity [25]. (The acronym
EPX is used here to avoid confusion with EPO, denoting erythropoietin.)
critical GTP binding domains [18, 19]. These mice have di-
IL-5/hE2 mice were also crossed with EPX⫺/⫺ mice to generate IL-5/hE2/
luted coat colors, related to reduced melanocyte secretion, EPX⫺/⫺ mice. The mice were maintained in ventilated microisolator cages
and exhibit a profound immunodeficiency due to a failure to housed in specific-pathogen–free environments at the University of Alberta
release cytolytic granules from CTLs and NK cells [20, 21]. (Edmonton, AB, Canada) and Mayo Clinic Scottsdale (Scottsdale, AZ,
Thus, many secretory cells from Ashen mice are characterized USA). All experiments were performed were in accordance with the guide-
by abnormal exocytosis. We hypothesized that Rab27a defi- lines of the University of Alberta’s Animal Care and Use Committee, the
Canadian Council of Animal Care, the National Institutes of Health, and
ciency reduces eosinophil exocytotic capacity. Eosinophils were
the Mayo Foundation.
isolated from Ashen mice and compared for their degranula-
tion responses with IL-5 transgenic eosinophils. Ashen mice
were also crossed with IL-5/hE2 double-transgenic mice to de- Preparation of eosinophils
termine the role of Rab27a in in vivo eosinophil exocytosis. HL-60 clone 15 (HL-60c15) cells (ATCC, Manassas VA, USA), chosen for
this study based on their propensity to differentiate into highly granulated
IL-5/hE2 double-transgenic mice are characterized by high
eosinophil-like cells, were treated with 500 ␮M n-butyrate [26]. Human eo-
expression of T-cell-derived IL-5, driven by the CD3 promoter, sinophils were purified from peripheral blood of atopic donors [27–29].
along with hE2 from Clara cells residing in the lungs, by fu- The purity and viability of human eosinophils was typically ⬎98%. Splenic
sion of the hE2 gene with the CC10 promoter [22]. These cy- eosinophils were isolated from IL-5 transgenic mice by using a published
tokines contribute significantly to eosinophil differentiation method [30, 31]. For peripheral blood mouse eosinophils, the cells were

A B C
ood Cells

100
10 3
WT
80 Ashen
8
o Total White Blo

2
6 60

4 40
1
2 20
% of

0 0 0
T

en
T

en

es

es

ils
ls
W

hi
sh

sh

yt

yt

ph
op
oc

oc
A

no
tr
ph

on

si
eu
m

Eo
N
Ly

Figure 1. Total and differential peripheral blood leukocyte counts were similar between WT C57BL/6 and Ashen mice. (A) Total cell counts from
the peripheral blood of WT C57BL/6 and Ashen mice were similar. (B) Percentages of peripheral blood eosinophils were identical in WT and
Ashen mice. (C) Differential counts of leukocytes in WT and Ashen mice showed similar proportions (n⫽3).

1266 Journal of Leukocyte Biology Volume 94, December 2013 www.jleukbio.org


Kim et al. Rab27a in eosinophil exocytosis

recovered from the peripheral blood of IL-5 transgenic mice at ⬎98% pu- Intratracheal instillation of mouse eosinophils
rity [23]. Cultured, in vitro bmEos were also generated from WT C57BL/6
Peripheral blood eosinophils isolated from IL-5 transgenic mice were re-
mice and differentiated in the presence of recombinant mouse SCF,
constituted at 10 ⫻ 106 cells in 30 ␮l of PBS and adoptively transferred
FLT3L, and IL-5 [32].
into the lungs of lightly anesthetized mice (3% isoflurane). BAL samples
from these mice were collected 24 h after instillation of eosinophils.
Subcellular fractionation and marker enzyme assays
Differentiated HL-60c15 cells and peripheral blood mouse eosinophils were ELISA of EPX
homogenized in a cell cracker (a ball-bearing– containing cell homoge- Quantification of EPX was achieved by an EPX ELISA method developed
nizer), and the resulting organelles were separated by a linear Nycodenz or by Mayo Clinic Arizona [37]. EPX amounts were quantified by comparing
Histodenz (Sigma-Aldrich St. Louis, MO, USA) gradient (0 – 45%) [33]. them to the standard curve generated from measuring EPX in pure mouse
Marker enzyme assays were used on fractionated samples. EPX activity in eosinophil lysates. The results are shown as eos. equiv./microliter, to indi-
the resulting fractions was measured by the reactivity with tetramethylbenzi- cate the absorbance measured per eosinophil count.
dine substrate, and cytosol fractions were detected with LDH or protein
assay [33] (Bio-Rad, Hercules, CA, USA).
Statistical analysis
Data were analyzed and graphed with Prism (version 5; GraphPad, San Di-
Immunofluorescence ego, CA, USA). The results are represented as the mean ⫾ se and analyzed
Image analysis of the immunofluorescence of adherent eosinophils was per- by 1-way ANOVA with Tukey’s post hoc analysis. For all experiments, P ⬍
formed after plating of peripheral blood mouse eosinophils from IL-5 0.05 was considered to be statistically significant.
transgenic NJ.1638 mice or human eosinophils from atopic donors in
8-well chamber slides (Lab-Tek Chambered Coverglass System; Nunc, Roch-
ester, NY, USA) [34]. The cells were resuspended in serum-free RPMI me-
RESULTS
dium, added to the glass surfaces of the chamber slides, and incubated for
15 min at 37°C, to allow adhesion. PAF (5 ␮M) was added to each well for Rab27a was expressed in eosinophils and colocalized
5 or 15 min. Stimulation was terminated by removal of the medium and
with EPX-containing granules
fixation of the cells with 4% paraformaldehyde for 20 min. The cells were
permeabilized with 0.1% Triton X-100 in PBS for 3 min before staining. To determine whether mouse eosinophils express Rab27a,
The mouse eosinophils were then blocked for 20 min with 2% BSA⫹2% bmEos and peripheral blood mouse eosinophils were tested
goat serum in PBS, to reduce nonspecific binding by antibodies, whereas for expression of Rab27a mRNA and protein. We determined
the human eosinophils were blocked with 2% human IgG in PBS [35]. that Rab27a mRNA and protein were expressed in bmEos
Mouse monoclonal anti-Rab27a (5 ␮g/ml; 20/RAB27 clone; BD Biosci- from WT C57BL/6 mice as well as bmEos and peripheral
ences, Inc., San Jose, CA, USA) was then added and detected with anti-
blood eosinophils from IL-5 transgenic NJ.1638 mice (Fig. 2).
mouse IgG-Cy3, followed by fluorescein isothiocyanate (FITC)– mouse
monoclonal anti-CD63 (Serotec, Raleigh, NC, USA). After the cells were
The levels of expression of Rab27a mRNA and protein, as well
washed with PBS, ProLong Gold mounting medium containing DAPI (In- as the molecular weight of Rab27a protein, in mouse eosino-
vitrogen-Molecular Probes, Eugene, OR, USA) was added to each well. Im- phils was similar to that in human eosinophils and butyrate-
munofluorescence labeling was imaged with a Deltavision OMX microscope treated HL-60c15 cells differentiated into an eosinophilic phe-
equipped with a ⫻60 objective (1.43 NA; Applied Precision, Issaquah, WA, notype.
USA). To determine the subcellular localization of Rab27a, the
eosinophils were subjected to lysis through a cell cracker, and
Gel electrophoresis and Western blot analysis the resulting intact organelles were separated across a linear
Proteins were separated by SDS-PAGE gels and transferred onto nitrocellu- density gradient consisting of Histodenz (0 – 45%). Peripheral
lose membranes. The membranes were blocked with Odyssey blocking buf- blood eosinophils from IL-5 Tg mice expressed high EPX ac-
fer (Li-Cor Bioscience, Lincoln, NE, USA) diluted 1:2 in PBS. Rab27a pro- tivity in crystalloid granule-containing fractions (high-density
tein was probed with an anti-Rab27a rabbit antibody (Santa Cruz Biotech- fractions 6 –10) that substantially overlapped with fractions
nology Inc., Dallas, TX, USA) [36]. Immunofluorescent protein bands were
containing Rab27a (fractions 6 –13) as determined by Western
quantified by using IRDye-conjugated secondary antibodies against primary
antibodies with a Li-Cor Odyssey infrared imaging system (Li-Cor Biosci- blot analysis. Most of the Rab27a immunoreactivity appeared
ence). in fractions enriched in membrane-bound organelles, which
typically entered the gradient and was separate from cytosolic
LDH-containing fractions above the gradient. The colocaliza-
In vitro stimulation of mouse eosinophils
tion of Rab27a with membrane-bound organelles in fraction-
Eosinophils (2⫻105) were stimulated with 0.1 ␮M PAF, 5 ␮M ionomycin,
ated eosinophils confirmed its membrane-associated proper-
or both. The cells were initially centrifuged at 400 g for 10 min, and then
supernatants from this step were centrifuged again at 10,000 g for 10 min
ties, on the one hand [15]. Plasma membrane gp91phox expres-
at 4°C to clarify the supernatants for the assay. The resulting supernatants sion, on the other hand, corresponded to lower density,
were placed in 96-well plates for detection of EPX by ELISA. plasma membrane-containing fractions (fractions 11–15).
These findings suggest that Rab27a colocalizes with EPX-con-
Preparation of BAL fluid taining crystalloid granules, as well as other lower density
membrane-bound organelles, in unstimulated eosinophils.
BAL fluid was recovered by introducing and collecting 1 ml of 2% FCS in
PBS. BAL fluid was initially centrifuged at 400 g for 10 min at 4°C, after
Next, we determined the effects of PAF stimulation on the
which the supernatant was recovered for a further centrifugation at 10,000 intracellular localization of Rab27a. PAF is a potent eosinophil
g for 10 min at 4°C to remove any remaining debris. Pellets from BAL sam- secretagogue that induces EPX release [38]. Upon stimulation
ples were analyzed for total and differential cell counts. by PAF, both EPX activity and Rab27a expression shifted to a

www.jleukbio.org Volume 94, December 2013 Journal of Leukocyte Biology 1267


A B

R b27
Rab27a Rab27a 27 kDa

β-Actin 42 kDa

Figure 2. Rab27a was expressed in mouse eosin- β-Actin


ophils and colocalized with crystalloid granule
fractions. (A) RT-PCR analysis of differentiated C Peripheral
p blood mouse IL-5 Tgg eosinophils
p
HL-60c15 cells and bmEos cultured from
Plasma
C57BL/6 mice. (B) Western blot analysis of
Granules membrane Cytosol
Rab27a expression in eosinophils from mouse
and human sources. BmEos were isolated from
C57BL/6 mice (left) and bmEos, and peripheral
blood (PB) eosinophils were prepared from IL-5 30
Tg mice (right), to compare Rab27a expression EPX Resting
with that in human eosinophils and differenti- EPX Stimulated
ated HL-60c15 cells. A total of 20 ␮g protein LDH
was loaded in each lane. (C) Subcellular frac-
20
tionation of peripheral blood eosinophils from
IL-5 Tg mice. An enzymatic assay was used to
detect EPX in fractions containing crystalloid
granules (fractions 6 –10), and LDH was assayed
to identify cytosolic fractions (fractions 16 –23). 10
Western blot analysis showed Rab27a (fractions
6 –13) and gp91phox (a marker for plasma mem-
brane, fractions 11–15).
0
0 5 10 15 20
Fraction #

Resng
Rab27a PAF-Smulated

gp91phox

slightly higher density fraction (fraction 6) compared with that staining indicating granular and vesicular localization, some of
in the resting cells (Fig. 2C). The amount of total granule-as- which colocalized with CD63⫹ crystalloid granules. These find-
sociated EPX activity was also reduced in stimulated cells, sug- ings are in agreement with subcellular fractionation results.
gesting degranulation of EPX. However, the distribution of Upon stimulation with PAF, several CD63⫹ granules redis-
Rab27a was not markedly altered after PAF stimulation and tributed toward the periphery and plasma membrane in both
exhibited expression in both low- and high-density fractions mouse (Fig. 3C) and human (Fig. 3D) eosinophils. Granule
(fractions 6 –13) similar to that of unstimulated cells. polarization to leading edges in both human and mouse eosin-
We confirmed colocalization of Rab27a with eosinophil crys- ophils was evident within 5 min of stimulation with PAF, be-
talloid granules by performing immunofluorescence analysis of fore EPX release. Previous studies showed that PAF at 5 ␮M
mouse and human eosinophils. CD63, used as a granule mem- induces significant EPX release after 30 min of stimulation
brane marker for eosinophil crystalloid granules [39], showed [38]. PAF was applied to cell supernatants and not as a che-
that Rab27a colocalized with CD63⫹ granules in the cytoplasm motactic gradient, suggesting that polarization of granules oc-
of mouse and human eosinophils (Fig. 3A, B). CD63 is a curred within the cells and that degranulation from PAF-stimu-
membrane-bound tetraspanin molecule [39]. As expected, im- lated eosinophils involves polarized exocytosis.
munofluorescence for CD63 showed characteristic ring-like In parallel with that finding, immunofluorescence for
structures in human eosinophils, suggesting granule mem- Rab27a showed colocalization with CD63⫹ granules in un-
brane localization of CD63. These intracellular CD63⫹ crystal- stimulated cells (Fig. 3). However, consistent with subcellular
loid granules were densely packed in human eosinophils and fractionation data, Rab27a was not exclusively localized to
showed minimal plasma membrane localization. In mouse eo- CD63⫹ granules in mouse and human eosinophils, as other
sinophils, CD63⫹ ring structures were less prevalent, but simi- Rab27a⫹ structures were evident that did not colocalize with
lar to human eosinophils, CD63 was found only in intracellu- CD63. Upon stimulation of mouse eosinophils with PAF (5
lar structures resembling granules. In contrast, Rab27a was min), Rab27a appeared to redistribute away from the CD63⫹
more widely distributed throughout the cells, with punctate granules found within cytoplasmic sites to other locations in

1268 Journal of Leukocyte Biology Volume 94, December 2013 www.jleukbio.org


Kim et al. Rab27a in eosinophil exocytosis

A Rab27 CD63 Merge

Figure 3. Rab27a colocalized with


crystalloid granules in mouse and
human eosinophils. (A) Mouse eo-
sinophils from the peripheral
blood of IL-5 Tg mice were plated
onto glass coverslips in 8-chamber
slides and allowed to adhere be-
B Rab27 CD63 Merge fore fixation and staining for
Rab27a and CD63, a membrane
marker for crystalloid granules.
Arrowhead indicates colocalization
of Rab27a and CD63 to the mem-
brane of a single crystalloid gran-
ule. (B) Human eosinophils (ar-
rowhead) from peripheral blood
were labeled similarly. (C) Mouse
IL-5 Tg eosinophils were treated
C 0 min 5 min 15 min
with PAF (5 ␮M) for 0, 5, and 15
min before fixation and staining.
Arrowhead and inset show strong
colocalization of Rab27a and CD63
at the cell periphery. (D) Human
peripheral blood eosinophils also
demonstrated cell-stretching, along
with granule polarization at the
leading edge, with colocalization of
D 0 min 5 min 15 min Rab27a and CD63 at the edges of
the cells during stimulation. Im-
ages of stimulated cells are repre-
sentative of 30 – 40% of cells in 10
high-powered fields each. Scale
bar ⫽ 5 ␮m.

the cell, including the cell membrane (Fig. 3C). This selective expression. We then compared degranulation responses of eo-
redistribution of Rab27a⫹ organelles is suggestive of piecemeal sinophils from the resulting IL-5/Ashen offspring with those of
degranulation, in which small, rapidly mobilized secretory vesi- the respective WT controls against the same background strain
cles shuttle granule products from the crystalloid granules to (C57BL/6) as was crossed with IL-5 transgenic mice.
the cell membrane, as previously shown in human eosinophils Peripheral blood eosinophils from IL-5/WT and IL-5/Ashen
[29]. However, at 15 min of PAF stimulation, Rab27a and mice were stimulated with the secretagogues PAF, ionomycin,
CD63 colocalized to highly focused, punctate regions at or or PAF⫹ionomycin, with EPX release being quantified as eos.
near the cell membrane in mouse eosinophils. In human eo- equiv./microliter. This unit of measurement was used to en-
sinophils, the pattern of Rab27a redistribution was distinct. sure that peripheral eosinophils isolated from both types of
PAF induced a redistribution of Rab27a that colocalized with mice contained equal amounts of EPX, as measured by an in-
CD63⫹ structures in polarized regions of cells, with relatively house ELISA [37]. Using this approach, we were able to detect
little concentration at the cell membrane (Fig. 3D, arrow- increased EPX release in supernatants from eosinophils stimu-
head). lated with PAF, the calcium ionophore ionomycin, and a
combination of PAF and ionomycin (Fig. 4). In contrast,
Ashen eosinophils showed deficient EPX release IL-5/Ashen exhibited significantly reduced EPX release in
compared with WT eosinophils in vitro response to all stimuli, compared with that in IL-5/WT eo-
Rab27a is a critical regulatory protein in granule exocytosis in sinophils (Fig. 4).
other secretory cells, and so we examined the role of Rab27a
in degranulation from eosinophils. We isolated eosinophils Ashen eosinophils exhibited defective EPX release
from Ashen mice, a substrain of C57BL/6 mice that contains a in vivo
splicing mutation in Rab27a [18], that were crossed with IL-5- We next confirmed that Ashen eosinophils are defective in
overexpressing mice (NJ.1638) [23], to obtain a large number their degranulation responses when administered in vivo. WT
of splenic and peripheral blood eosinophils lacking Rab27a and Ashen mice were sensitized and challenged with OVA, and

www.jleukbio.org Volume 94, December 2013 Journal of Leukocyte Biology 1269


Thus, we compared EPX levels from BAL supernatants of
IL-5 mice with intratracheal instillation of IL-5/WT and IL-5/Ashen
IL-5/Ashen peripheral blood eosinophils (Fig. 6). BAL from mice that re-
ceived IL-5/WT eosinophils exhibited robust EPX release. In
contrast, BAL from IL-5/Ashen eosinophil-instilled mice
showed significantly reduced amounts of EPX (P⬍0.001) (Fig.
6A). When differential cell counts were performed on these
samples, no differences were observed in total BAL cellularity
and eosinophil count (Fig. 6B, C). Thus, these results support
and confirm our data from EPX levels in BAL of OVA-treated
mice and suggest that Rab27a-deficient Ashen eosinophils have
defective degranulation responses in vivo.
Vehicle PAF +
PAF ionomycin We tested the apparent degranulation deficiency of Ashen
control ionomycin
eosinophils further in IL-5/hE2/WT and IL-5/hE2/Ashen
Figure 4. Eosinophils from IL-5/Ashen mice were deficient in EPX mice. These were generated by crossing double-transgenic IL-
release in vitro. In vitro degranulation was observed in purified pe- 5/hE2 mice with either WT C57BL/6 or Ashen mice and using
ripheral blood eosinophils from IL-5 Tg and IL-5/Ashen mice. Super- the offspring that were positive for the appropriate genotypes.
natants from stimulated cells were measured by EPX ELISA. Cells were For comparison, a third strain of IL-5/hE2/EPX⫺/⫺ mice was
stimulated with 50 ng/ml PAF and/or 1 ␮M ionomycin for 6 h. The
generated in which EPX expression was absent. BAL superna-
data are the mean ⫾ se (nⱖ10). *P ⬍ 0.05, ***P ⬍ 0.001.
tants from these 3 strains of mice (6 – 8 wk) were collected,
and spontaneously secreted EPX levels were measured (Fig. 7).
EPX release was determined by ELISA of BAL samples ob- Again, IL-5/hE2/Ashen mice showed markedly reduced EPX
tained on day 28 (Fig. 5). WT and Ashen mice treated with sa- levels in BAL than in IL-5/hE2/WT BAL. Taken together, our
line alone did not show any difference in EPX levels in their data demonstrate that eosinophils lacking Rab27a have a defi-
BAL. However, OVA sensitization and challenge of Ashen mice ciency in secretion of EPX-containing crystalloid granules.
led to a significant (P⬍0.001) decrease in EPX content in BAL
samples compared with those from OVA-treated WT mice. These
findings suggest that airway eosinophil degranulation is deficient
after OVA sensitization and challenge in Ashen mice.
However, upon further assessment of total BAL cellularity,
A B
we observed a reduced total number of cells and eosinophils
in the BAL of OVA-treated Ashen compared with that of simi-
larly treated WT mice, even though the percentages of eosino-
phils of total BAL cellularity were similar in both (Fig. 5B–D).
Thus, reduced EPX concentrations in BAL samples from Ashen
mice may be related to a decrease in eosinophils rather than a
degranulation defect.
Saline OVA Saline OVA Saline OVA Saline OVA
The question of whether the reduced BAL EPX quantities
Wild type Ashen Wild type Ashen
observed in OVA-treated Ashen mice was due to lower eosino-
phil counts was addressed in a novel ex vivo eosinophil de-
C D
granulation experiment. Peripheral blood eosinophils from
IL-5/WT and IL-5/Ashen mice were isolated, purified, and
adoptively transferred into the lungs of triple-transgenic IL-5/
hE2/EPX⫺/⫺ mice. These mice were generated to evoke maxi-
mally stimulating eosinophil degranulation conditions in vivo,
with a complete absence of endogenous EPX secreted by the
recipient animal. Typically, a large number of eosinophils
Saline OVA Saline OVA Saline OVA Saline OVA
transmigrate to the airway spaces in IL-5/hE2 double-trans-
Wild type Ashen Wild type Ashen
genic animals and spontaneously degranulate in response to
tissue-overexpressed IL-5 and eotaxin-2 [22]. Thus, intratrache-
ally introduced WT eosinophils would be expected to degranu- Figure 5. OVA-sensitized and -challenged Ashen mice exhibited de-
late in the airways of these animals (due to transgenic overex- creased BAL EPX release. (A) EPX was detected in BAL samples ob-
pression of eosinophil-specific activating cytokine IL-5 and tained on day 28 of OVA sensitization and challenge or saline controls
from WT and Ashen mice. EPX quantity was measured by EPX ELISA.
chemokine eotaxin-2). In this ex vivo degranulation assay sys-
(B) BAL cellularity, (C) number of BAL eosinophils, and (D) BAL
tem, any EPX that is detected in BAL samples would be exclu- percentage of eosinophils in saline- and OVA-challenged WT and
sively derived from donor eosinophils instilled into the trachea Ashen mice. The data are the mean ⴞ se (n>5). *P ⬍ 0.05;
of IL-5/hE2/EPX⫺/⫺ mice. **P ⬍ 0.01; ***P ⬍ 0.001.

1270 Journal of Leukocyte Biology Volume 94, December 2013 www.jleukbio.org


Kim et al. Rab27a in eosinophil exocytosis

A B C

PBS IT IL-5/WT IL-5/Ashen IL-5/WT IL-5/Ashen IL-5/WT IL-5/Ashen


Eos IT Eos IT Eos IT Eos IT Eos IT Eos IT

Figure 6. IL-5/Ashen eosinophils were defective in EPX release when injected intratracheally. (A) Purified peripheral blood eosinophils (107)
from IL-5/WT or IL-5/Ashen mice were intratracheally instilled into IL-5/hE2/EPX⫺/⫺ mice, and BAL was collected from euthanized mice 24 h
after instillation. (B) BAL cellularity and (C) BAL eosinophils after intratracheal injection of eosinophils into IL-5/hE2/EPX⫺/⫺ mice. The data
are the mean ⴞ se (n⫽4). ***P ⬍ 0.001.

DISCUSSION been shown for Rab27a in exocytosis of crystalloid granules in


eosinophils.
This report describes, for the first time, a direct role for
The function of Rab27a in granule exocytosis is dependent
Rab27a in eosinophil degranulation, identified in the Ashen
on a wide range of specific effector molecules [44]. Proteins
Rab27a gene knockout strain. The role of Rab27a in granule
containing an N-terminal Rab27a-binding region, known as
exocytosis and the development of inflammatory processes has
Slp/Slac2 proteins or exophilins, have been shown to bind
been investigated in CTLs [20, 21, 40], NK cells [16, 41], and
Rab27a and mediate respiratory burst in neutrophils [42, 45].
neutrophils [42], whereas Rab27b has been implicated in mast
Rab27a may also bind directly to SNARE fusion proteins, to
cells [43]. Our previous report on the expression and function
initiate exocytotic membrane fusion between granules and the
of Rab27a in human eosinophils demonstrated that eosino-
plasma membrane. The R-SNAREs VAMP-2, -7, and -8, along
phils express and activate Rab27a in response to artificial ago-
with their respective binding partners, the Q-SNAREs
nists (phorbol esters and calcium ionophore) as well as physio-
SNAP-23 and syntaxin-4, are expressed in human eosino-
logical stimuli (fMLF and serum-coated zymosan) [13]. Hu-
phils, and VAMP-7 has been shown to regulate exocytotic
man peripheral blood eosinophils expressed mRNA for Rab8a,
release of the granule proteins EPX and EDN [46 – 49]. Fi-
-8b, -10, -11a, -27a, and -37, with weak signals for Rab11b and
nally, another putative effector molecule for Rab27a may be
-13 and no detectable signal for Rab27b. We also found in-
the Sec/Munc family member Munc13-4, which has been
creased expression of Rab27a protein, with greater activity ob-
demonstrated in other cell types, including CTL cells, NK
served in eosinophils from patients with asthma compared
cells, mast cells, and platelets, to regulate Rab27a-induced
with normal subjects [13]. However, a direct role had not
exocytosis [50 –52].
The expression and function of Rab27a and its effector mol-
ecules have not yet been explored in eosinophils. These obser-
vations led to the current study with a view to further elucidate
the expression and function of Rab27a in eosinophils and to
determine how this protein may influence degranulation. Us-
ing Western blot analysis, subcellular fractionation, and immu-
nofluorescence analysis, we established that Rab27a was ex-
pressed in both mouse and human eosinophils and that it co-
localized with eosinophil crystalloid granules, as well as other
membrane-bound organelles. To pursue this finding, we chose
the Ashen strain, which exhibits a functional deficiency in
Rab27a, and derived mouse bmEos from those animals, to
confirm the function of Rab27a in mouse eosinophils and vali-
IL-5/hE2/EPX-/- IL-5/hE2 IL-5/hE2/Ashen
date ensuing mouse models of airway inflammation. Previous
studies in neutrophils have shown that Rab27a is mostly local-
Figure 7. Spontaneous release of EPX was reduced in BAL fluid from
ized to specific and gelatinase-enriched tertiary granules and is
IL-5/hE2/Ashen mice. IL-5/hE2/Ashen mice (6 – 8 wk) were generated
to determine spontaneous EPX release in BAL fluid in comparison not essential for myeloperoxidase release from azurophilic
with that in age-matched IL-5/hE2 mice. EPX was measured by EPX granules [53]. In contrast, our results provide strong evidence
ELISA. The data are the mean ⴞ se (n⫽3– 4). ***P ⬍ 0.001. that Rab27a associates with eosinophil crystalloid granules con-

www.jleukbio.org Volume 94, December 2013 Journal of Leukocyte Biology 1271


taining CD63 and EPX and that Rab27a is necessary for EPX cytokine and chemokine secretion from other cell types that
release. are dependent on Rab27a.
To determine whether the localization of Rab27a and EPX To further explore the possibility that Rab27a is involved in
change with stimulation, eosinophils were treated with the po- eosinophil degranulation, a novel ex vivo degranulation assay
tent secretagogue PAF [38]. After PAF activation, the total for eosinophils was designed in which eosinophils were intra-
EPX level across subfractions was significantly reduced, sug- tracheally injected into IL-5/hE2/EPX⫺/⫺ mice. The use of
gesting extracellular release of EPX, and EPX activity was split IL-5/hE2/EPX⫺/⫺ mice provided a unique lung environment
between 2 fractions (6 and 8). A possible explanation for the where intratracheally instilled eosinophils are induced to de-
double peak of EPX activity in stimulated eosinophils is that granulate by in vivo secretagogues generated from tissue
stimulated cells release EPX through piecemeal degranulation sources. These mice lack systemically expressed EPX, and any
[29, 54]. In confirmation of this finding, immunofluorescence detected EPX in BAL samples must therefore originate exclu-
images showed that a proportion of Rab27a redistributed away sively from donor, not recipient, eosinophils. We found signifi-
from CD63⫹ granules after 5 min of PAF stimulation in mouse cantly less EPX in BAL from IL-5/hE2/EPX⫺/⫺ mice with IL-
eosinophils. Some Rab27a immunoreactivity was found at the 5/Ashen eosinophils instilled when compared with those with
cell membrane, colocalizing with a proportion of CD63⫹ gran- IL-5/WT eosinophils instilled, and differential cell counts per-
ules in mouse eosinophils. However, at 15 min of PAF stimula- formed on BAL from these 2 groups were not significantly dif-
tion, there was a redistribution of Rab27a⫹ structures colocal- ferent. These data provide supportive evidence for Rab27a in
izing with CD63⫹ granules at the cell membrane in highly fo- eosinophil degranulation, using a model in which exogenously
cused, punctate regions. This suggests that Rab27a may be administered eosinophils are introduced into a highly stimula-
involved in piecemeal degranulation in early cell stimulation, tory airway environment. Consistent with these findings, dou-
as well as crystalloid granule fusion with the cell membrane in ble-transgenic IL-5/hE2 mice crossed with Ashen mice to gen-
erate a large number of eosinophils lacking Rab27a also
mouse eosinophils at later stages of degranulation (within 15
showed a significant decrease in EPX in BAL samples from
min of PAF stimulation).
IL-5/hE2/Ashen mice.
In human eosinophils, PAF induced a marked polarization,
In conclusion, our findings provide strong evidence support-
whereupon CD63⫹ granules mobilized to the leading edges of
ing a role for Rab27a in eosinophil degranulation, both in
the cells. Rab27a was found to colocalize with some CD63⫹
vitro and in vivo, in the mouse model. The findings support
granules at the leading edge of the cells. Taken together,
our study on Rab27a function in human eosinophils [13] and
these findings suggest that Rab27a coordinates fusion between
suggest that eosinophils are dependent on Rab27a for degran-
crystalloid granules and plasma membrane to regulate granule
ulation responses in allergic airway inflammation and asthma.
exocytosis in eosinophils, although there were subtle differ-
ences in the manner in which Rab27a was redistributed after
PAF stimulation in mouse and human cells. AUTHORSHIP
The role of Rab27a in eosinophil degranulation was further
J.D.K., L.W., N.S., R.H., and A.S. conducted the experiments
investigated by stimulating peripheral blood eosinophils iso-
and were supervised by J.J.L., R.M., and P.L. P.L. also con-
lated from WT and Ashen mice. In this system, a novel EPX
ducted experiments for the study. S.O. contributed to experi-
ELISA [37] was used to measure the amount of released
ments and training of users and was supervised by J.J.L. M.C.S.
mouse EPX after stimulation. This EPX ELISA was developed
provided the Ashen strain. This study was supervised by J.J.L.,
in house and provides a 10-fold higher sensitivity than does
R.M., and P.L., who obtained the funding.
the widely used OPD assay [55]. Some studies have suggested
that mouse eosinophils do not readily degranulate either in
vitro or in vivo [13, 56, 57]. However, we have recently re- ACKNOWLEDGMENTS
ported that mouse eosinophils degranulate in vitro in response
to at least 2 secretagogues: PAF and calcium ionophore This study was funded by CIHR (R.M., P.L.), the Lung Associa-
[34, 38]. tion of Alberta and NWT (to J.D.K.), and U.S. National Insti-
tutes of Health, Bethesda, MD (J.J.L.). J.D.K. also received a
Our in vivo findings confirmed that Rab27a plays an impor-
CIHR Banting & Best Canada Graduate Scholarship–Master’s
tant role in eosinophil exocytosis in association with EPX-con-
Award for this project. The authors thank Elizabeth Jacobsen
taining crystalloid granules. OVA-treated Ashen BAL samples
and others at the Mayo Clinic Scottsdale for lending excellent
exhibited significantly diminished levels of EPX when com-
technical support and expertise and Renjith Pillai, Caroline
pared to OVA-treated WT BAL. However, when differential
Ethier, Dr. Francis Davoine, Vivek Gandhi, and Dr. Stephen
cell counts were performed, OVA-treated Ashen mice had
Ogg for support contributing to findings in this study. Images
lower BAL cellularity, despite having a percentage of eosino-
were acquired using resources and expertise provided by the
phils similar to that in OVA-treated WT mice. This result sug-
Faculty of Medicine and Dentistry’s core imaging center at the
gests that the reduction of EPX observed in the BAL of OVA-
University of Alberta.
treated Ashen mice is related to a reduction in airway eosino-
phils. The finding was unexpected, but was not surprising,
given that global gene deletion of Rab27a may indirectly affect DISCLOSURES
eosinophil recruitment and accumulation in the lungs through The authors declare no conflicts of interest.

1272 Journal of Leukocyte Biology Volume 94, December 2013 www.jleukbio.org


Kim et al. Rab27a in eosinophil exocytosis

REFERENCES phil-dependent model of respiratory inflammation with characteristics


1. Lacy, P., Moqbel, R. (2001) Immune effector functions of eosinophils in of severe asthma. J. Immunol. 178, 7879 –7889.
allergic airway inflammation. Curr. Opin. Allergy Clin. Immunol. 1, 79 –84. 23. Lee, N. A., McGarry, M. P., Larson, K. A., Horton, M. A., Kristensen,
A. B., Lee, J. J. (1997) Expression of IL-5 in thymocytes/T cells leads to
2. Adamko, D., Lacy, P., Moqbel, R. (2004) Eosinophil function in allergic
the development of a massive eosinophilia, extramedullary eosinophilo-
inflammation: from bone marrow to tissue response. Curr. Allergy Asthma
poiesis, and unique histopathologies. J. Immunol. 158, 1332–1344.
Rep. 4, 149 –158.
24. Garcia-Zepeda, E. A., Rothenberg, M. E., Ownbey, R. T., Celestin, J.,
3. Rothenberg, M. E., Hogan, S. P. (2006) The eosinophil. Annu. Rev. Im-
Leder, P., Luster, A. D. (1996) Human eotaxin is a specific chemoattrac-
munol. 24, 147–174. tant for eosinophil cells and provides a new mechanism to explain tissue
4. Hogan, S. P., Rosenberg, H. F., Moqbel, R., Phipps, S., Foster, P. S., eosinophilia. Nat. Med. 2, 449 –456.
Lacy, P., Kay, A. B., Rothenberg, M. E. (2008) Eosinophils: biological 25. Denzler, K. L., Borchers, M. T., Crosby, J. R., Cieslewicz, G., Hines,
properties and role in health and disease. Clin. Exp. Allergy 38, 709 –750. E. M., Justice, J. P., Cormier, S. A., Lindenberger, K. A., Song, W., Wu,
5. Jacobsen, E. A., Taranova, A. G., Lee, N. A., Lee, J. J. (2007) Eosino- W., Hazen, S. L., Gleich, G. J., Lee, J. J., Lee, N. A. (2001) Extensive
phils: singularly destructive effector cells or purveyors of immunoregula- eosinophil degranulation and peroxidase-mediated oxidation of airway
tion? J. Allergy Clin. Immunol. proteins do not occur in a mouse ovalbumin-challenge model of pulmo-
6. Leckie, M. J., ten Brinke, A., Khan, J., Diamant, Z., O’Connor, B. J., nary inflammation. J. Immunol. 167, 1672–1682.
Walls, C. M., Mathur, A. K., Cowley, H. C., Chung, K. F., Djukanovic, R., 26. Ishihara, K., Hong, J., Zee, O., Ohuchi, K. (2005) Mechanism of the
Hansel, T. T., Holgate, S. T., Sterk, P. J., Barnes, P. J. (2000) Effects of eosinophilic differentiation of HL-60 clone 15 cells induced by n-bu-
an interleukin-5 blocking monoclonal antibody on eosinophils, airway tyrate. Int. Arch. Allergy Immunol. 137(Suppl. 1), 77–82.
hyper-responsiveness, and the late asthmatic response. Lancet 356, 2144 – 27. Lacy, P., Abdel-Latif, D., Steward, M., Musat-Marcu, S., Man, S. F., Moq-
2148. bel, R. (2003) Divergence of mechanisms regulating respiratory burst in
7. Flood-Page, P. T., Menzies-Gow, A. N., Kay, A. B., Robinson, D. S. blood and sputum eosinophils and neutrophils from atopic subjects. J.
(2003) Eosinophil’s role remains uncertain as anti-interleukin-5 only Immunol. 170, 2670 –2679.
partially depletes numbers in asthmatic airway. Am. J. Respir. Crit. Care 28. Levi-Schaffer, F., Lacy, P., Severs, N. J., Newman, T. M., North, J., Gom-
Med. 167, 199 –204. perts, B., Kay, A. B., Moqbel, R. (1995) Association of granulocyte-mac-
8. Kips, J. C., O’Connor, B. J., Langley, S. J., Woodcock, A., Kerstjens, rophage colony-stimulating factor with the crystalloid granules of hu-
H. A., Postma, D. S., Danzig, M., Cuss, F., Pauwels, R. A. (2003) Effect man eosinophils. Blood 85, 2579 –2586.
of SCH55700, a humanized anti-human interleukin-5 antibody, in severe 29. Lacy, P., Mahmudi-Azer, S., Bablitz, B., Hagen, S. C., Velazquez, J. R.,
persistent asthma: a pilot study. Am. J. Respir. Crit. Care Med. 167, 1655– Man, S. F., Moqbel, R. (1999) Rapid mobilization of intracellularly
1659. stored RANTES in response to interferon-␥ in human eosinophils. Blood
9. Nair, P., Pizzichini, M. M., Kjarsgaard, M., Inman, M. D., Efthimiadis, A., 94, 23–32.
Pizzichini, E., Hargreave, F. E., O’Byrne, P. M. (2009) Mepolizumab for 30. Rothenberg, M. E., Luster, A. D., Leder, P. (1995) Murine eotaxin: an
prednisone-dependent asthma with sputum eosinophilia. N. Engl. J. Med. eosinophil chemoattractant inducible in endothelial cells and in inter-
360, 985–993. leukin 4-induced tumor suppression. Proc. Natl. Acad. Sci. U. S. A. 92,
10. Haldar, P., Brightling, C. E., Hargadon, B., Gupta, S., Monteiro, W., 8960 –8964.
Sousa, A., Marshall, R. P., Bradding, P., Green, R. H., Wardlaw, A. J., 31. Lacy, P., Willetts, L., Kim, J. D., Lo, A., Lam, B., MacLean, E. I., Moqbel,
Pavord, I. D. (2009) Mepolizumab and exacerbations of refractory eosin- R., Rothenberg, M. E., Zimmermann, N. (2011) Agonist activation of
ophilic asthma. N. Engl. J. Med. 360, 973–984. F-actin-mediated eosinophil shape change and mediator release is de-
11. Lee, J. J., Dimina, D., Macias, M. P., Ochkur, S. I., McGarry, M. P., pendent on Rac2. Int Arch Allergy Immunol. 156, 137–147.
O’Neill, K. R., Protheroe, C., Pero, R., Nguyen, T., Cormier, S. A., Len- 32. Dyer, K. D., Moser, J. M., Czapiga, M., Siegel, S. J., Percopo, C. M.,
kiewicz, E., Colbert, D., Rinaldi, L., Ackerman, S. J., Irvin, C. G., Lee, Rosenberg, H. F. (2008) Functionally competent eosinophils differenti-
N. A. (2004) Defining a link with asthma in mice congenitally deficient ated ex vivo in high purity from normal mouse bone marrow. J. Immu-
in eosinophils. Science 305, 1773–1776. nol. 181, 4004 –4009.
12. Humbles, A. A., Lloyd, C. M., McMillan, S. J., Friend, D. S., Xanthou, 33. Lacy, P., Levi-Schaffer, F., Mahmudi-Azer, S., Bablitz, B., Hagen, S. C.,
G., McKenna, E. E., Ghiran, S., Gerard, N. P., Yu, C., Orkin, S. H., Ge- Velazquez, J., Kay, A. B., Moqbel, R. (1998) Intracellular localization of
rard, C. (2004) A critical role for eosinophils in allergic airways remod- interleukin-6 in eosinophils from atopic asthmatics and effects of inter-
eling. Science 305, 1776 –1779. feron ␥. Blood 91, 2508 –2516.
13. Coughlin, J. J., Odemuyiwa, S. O., Davidson, C. E., Moqbel, R. (2008) 34. Lacy, P., Willetts, L., Kim, J. D., Lo, A. N., Lam, B., Maclean, E. I., Moq-
Differential expression and activation of Rab27A in human eosinophils: bel, R., Rothenberg, M. E., Zimmermann, N. (2011) Agonist activation
relationship to blood eosinophilia. Biochem. Biophys. Res. Commun. 373, of f-actin-mediated eosinophil shape change and mediator release is de-
382–386. pendent on Rac2. Int. Arch. Allergy Immunol. 156, 137–147.
14. Seabra, M. C., Mules, E. H., Hume, A. N. (2002) Rab GTPases, intracel- 35. Mahmudi-Azer, S., Lacy, P., Bablitz, B., Moqbel, R. (1998) Inhibition of
lular traffic and disease. Trends. Mol. Med. 8, 23–30. nonspecific binding of fluorescent-labelled antibodies to human eosino-
15. Seabra, M. C., Coudrier, E. (2004) Rab GTPases and myosin motors in phils. J. Immunol. Methods 217, 113–119.
organelle motility. Traffic 5, 393–399. 36. Hume, A. N., Collinson, L. M., Rapak, A., Gomes, A. Q., Hopkins, C. R.,
16. Wood, S. M., Meeths, M., Chiang, S. C., Bechensteen, A. G., Boelens, Seabra, M. C. (2001) Rab27a regulates the peripheral distribution of
J. J., Heilmann, C., Horiuchi, H., Rosthoj, S., Rutynowska, O., Winiarski, melanosomes in melanocytes. J. Cell Biol. 152, 795–808.
37. Ochkur, S. I., Kim, J. D., Protheroe, C. A., Colbert, D., Moqbel, R., Lacy,
J., Stow, J. L., Nordenskjold, M., Henter, J. I., Ljunggren, H. G., Bryce-
P., Lee, J. J., Lee, N. A. (2012) The development of a sensitive and spe-
son, Y. T. (2009) Different NK cell-activating receptors preferentially
cific ELISA for mouse eosinophil peroxidase: assessment of eosinophil
recruit Rab27a or Munc13-4 to perforin-containing granules for cytotox-
degranulation ex vivo and in models of human disease. J. Immunol. Meth-
icity. Blood 114, 4117–4127. ods 375, 138 –147.
17. Arico, M., Zecca, M., Santoro, N., Caselli, D., Maccario, R., Danesino, C., 38. Dyer, K. D., Percopo, C. M., Xie, Z., Yang, Z., Kim, J. D., Davoine, F.,
de Saint Basile, G., Locatelli, F. (2002) Successful treatment of Griscelli Lacy, P., Druey, K. M., Moqbel, R., Rosenberg, H. F. (2010) Mouse and
syndrome with unrelated donor allogeneic hematopoietic stem cell human eosinophils degranulate in response to platelet-activating factor
transplantation. Bone Marrow Transplant. 29, 995–998. (PAF) and lysoPAF via a PAF-receptor-independent mechanism: evi-
18. Wilson, S. M., Yip, R., Swing, D. A., O’Sullivan, T. N., Zhang, Y., Novak, dence for a novel receptor. J. Immunol. 184, 6327–6334.
E. K., Swank, R. T., Russell, L. B., Copeland, N. G., Jenkins, N. A. 39. Mahmudi-Azer, S., Downey, G. P., Moqbel, R. (2002) Translocation of
(2000) A mutation in Rab27a causes the vesicle transport defects ob- the tetraspanin CD63 in association with human eosinophil mediator
served in ashen mice. Proc. Natl. Acad. Sci. U. S. A. 97, 7933–7938. release. Blood 99, 4039 –4047.
19. Barral, D. C., Ramalho, J. S., Anders, R., Hume, A. N., Knapton, H. J., 40. Bahadoran, P., Aberdam, E., Mantoux, F., Busca, R., Bille, K., Yalman,
Tolmachova, T., Collinson, L. M., Goulding, D., Authi, K. S., Seabra, N., de Saint-Basile, G., Casaroli-Marano, R., Ortonne, J. P., Ballotti, R.
M. C. (2002) Functional redundancy of Rab27 proteins and the patho- (2001) Rab27a: a key to melanosome transport in human melanocytes.
genesis of Griscelli syndrome. J. Clin. Invest. 110, 247–257. J. Cell Biol. 152, 843–850.
20. Haddad, E. K., Wu, X., Hammer, J. A., 3rd, Henkart, P. A. (2001) De- 41. Casey, T. M., Meade, J. L., Hewitt, E. W. (2007) Organelle proteomics:
fective granule exocytosis in Rab27a-deficient lymphocytes from Ashen identification of the exocytic machinery associated with the natural
mice. J. Cell Biol. 152, 835–842. killer cell secretory lysosome. Mol. Cell. Proteomics
21. Stinchcombe, J. C., Barral, D. C., Mules, E. H., Booth, S., Hume, A. N., 42. Munafo, D. B., Johnson, J. L., Ellis, B. A., Rutschmann, S., Beutler, B.,
Machesky, L. M., Seabra, M. C., Griffiths, G. M. (2001) Rab27a is re- Catz, S. D. (2007) Rab27a is a key component of the secretory machin-
quired for regulated secretion in cytotoxic T lymphocytes. J. Cell Biol. ery of azurophilic granules in granulocytes. Biochem. J. 402, 229 –239.
152, 825–834. 43. Mizuno, K., Tolmachova, T., Ushakov, D. S., Romao, M., Abrink, M.,
22. Ochkur, S. I., Jacobsen, E. A., Protheroe, C. A., Biechele, T. L., Pero, Ferenczi, M. A., Raposo, G., Seabra, M. C. (2007) Rab27b regulates mast
R. S., McGarry, M. P., Wang, H., O’Neill, K. R., Colbert, D. C., Colby, cell granule dynamics and secretion. Traffic 8, 883–892.
T. V., Shen, H., Blackburn, M. R., Irvin, C. C., Lee, J. J., Lee, N. A. 44. Fukuda, M. (2008) Regulation of secretory vesicle traffic by Rab small
(2007) Coexpression of IL-5 and eotaxin-2 in mice creates an eosino- GTPases. Cell. Mol. Life Sci. 65, 2801–2813.

www.jleukbio.org Volume 94, December 2013 Journal of Leukocyte Biology 1273


45. McAdara Berkowitz, J. K., Catz, S. D., Johnson, J. L., Ruedi, J. M., Thon, 52. Menager, M. M., Menasche, G., Romao, M., Knapnougel, P., Ho, C. H.,
V., Babior, B. M. (2001) JFC1, a novel tandem C2 domain-containing Garfa, M., Raposo, G., Feldmann, J., Fischer, A., de Saint Basile, G.
protein associated with the leukocyte NADPH oxidase. J. Biol. Chem. 276, (2007) Secretory cytotoxic granule maturation and exocytosis require
18855–18862. the effector protein hMunc13-4. Nat. Immunol.
46. Lacy, P., Logan, M. R., Bablitz, B., Moqbel, R. (2001) Fusion protein 53. Herrero-Turrion, M. J., Calafat, J., Janssen, H., Fukuda, M., Mollinedo,
vesicle-associated membrane protein 2 is implicated in IFN␥-induced F. (2008) Rab27a regulates exocytosis of tertiary and specific granules in
piecemeal degranulation in human eosinophils from atopic individuals. human neutrophils. J. Immunol. 181, 3793–3803.
J. Allergy Clin. Immunol. 107, 671–678. 54. Spencer, L. A., Melo, R. C., Perez, S. A., Bafford, S. P., Dvorak, A. M.,
47. Logan, M. R., Lacy, P., Bablitz, B., Moqbel, R. (2002) Expression of eo- Weller, P. F. (2006) Cytokine receptor-mediated trafficking of pre-
sinophil target SNAREs as potential cognate receptors for vesicle-associ- formed IL-4 in eosinophils identifies an innate immune mechanism of
ated membrane protein-2 in exocytosis. J. Allergy Clin. Immunol. 109, cytokine secretion. Proc. Natl. Acad. Sci. U. S. A. 103, 3333–3338.
299 –306. 55. Adamko, D. J., Wu, Y., Gleich, G. J., Lacy, P., Moqbel, R. (2004) The
48. Logan, M. R., Lacy, P., Odemuyiwa, S. O., Steward, M., Davoine, F., induction of eosinophil peroxidase release: improved methods of mea-
Kita, H., Moqbel, R. (2006) A critical role for vesicle-associated mem- surement and stimulation. J. Immunol. Methods 291, 101–108.
brane protein-7 in exocytosis from human eosinophils and neutrophils. 56. Erjefalt, J. S., Greiff, L., Andersson, M., Adelroth, E., Jeffery, P. K.,
Allergy 61, 777–784. Persson, C. G. (2001) Degranulation patterns of eosinophil granulocytes
49. Logan, M. R., Odemuyiwa, S. O., Moqbel, R. (2003) Understanding exo- as determinants of eosinophil driven disease. Thorax 56, 341–344.
cytosis in immune and inflammatory cells: the molecular basis of media- 57. Stelts, D., Egan, R. W., Falcone, A., Garlisi, C. G., Gleich, G. J., Kreut-
tor secretion. J. Allergy Clin. Immunol. 111, 923–932. ner, W., Kung, T. T., Nahrebne, D. K., Chapman, R. W., Minnicozzi, M.
50. Shirakawa, R., Higashi, T., Tabuchi, A., Yoshioka, A., Nishioka, H., Fu- (1998) Eosinophils retain their granule major basic protein in a murine
kuda, M., Kita, T., Horiuchi, H. (2004) Munc13-4 is a GTP-Rab27-bind- model of allergic pulmonary inflammation. Am. J. Respir. Cell Mol. Biol.
ing protein regulating dense core granule secretion in platelets. J. Biol. 18, 463–470.
Chem. 279, 10730 –10737.
51. Neeft, M., Wieffer, M., de Jong, A. S., Negroiu, G., Metz, C. H., van
Loon, A., Griffith, J., Krijgsveld, J., Wulffraat, N., Koch, H., Heck, A. J.,
Brose, N., Kleijmeer, M., van der Sluijs, P. (2005) Munc13-4 is an effec- KEY WORDS:
tor of rab27a and controls secretion of lysosomes in hematopoietic cells. allergic asthma 䡠 degranulation 䡠 subcellular fractionation 䡠 IL-5 trans-
Mol. Biol. Cell 16, 731–741. genic mice 䡠 hE2 transgenic mice 䡠 Ashen mice

1274 Journal of Leukocyte Biology Volume 94, December 2013 www.jleukbio.org

You might also like