ArticlePDF Available

Two macrophage migration inhibitory factors regulate starfish larval immune cell chemotaxis

Authors:

Abstract and Figures

Immune cell recruitment is critical step in the inflammatory response and associated diseases. However, the underlying regulatory mechanisms are poorly understood in invertebrates. Mesenchyme cells of the starfish larvae, which allowed Metchnikoff to complete his landmark experiments, are important model for analysis of immune cell migration. The present study investigated the role of macrophage migration inhibitory factor (MIF)-an evolutionarily conserved cytokine that is functionally similar to chemokines-in the larvae of the starfish Patiria (Asterina) pectinifera, which were found to possess two orthologs, ApMIF1 and ApMIF2. ApMIF1 and ApMIF2 clustered with mammalian MIF and its homolog D-dopachrome tautomerase (DDT), respectively, in the phylogenetic analysis. In contrast to the functional similarity between mammalian MIF and DDT, ApMIF1 knockdown resulted in the excessive recruitment of mesenchyme cells in vivo, whereas ApMIF2 deficiency inhibited the recruitment of these cells to foreign bodies. Mesenchyme cells migrated along a gradient of recombinant ApMIF2 in vitro, whereas recombinant ApMIF1 completely blocked ApMIF2-induced directed migration. Moreover, the expression patterns of ApMIF1 and ApMIF2 messenger RNA in bacteria-challenged mesenchyme cells were consistent with in vivo observations of cell behaviors. These results indicate that ApMIF1 and ApMIF2 act as chemotactic inhibitory and stimulatory factors, respectively, and coordinately regulate mesenchyme cell recruitment during the immune response in starfish larvae. This is the first report describing opposing functions for MIF- and DDT-like molecules. Our findings provide novel insight into the mechanisms underlying immune regulation in invertebrates.Immunology and Cell Biology advance online publication, 2 February 2016; doi:10.1038/icb.2016.6.
Content may be subject to copyright.
OUTSTANDING OBSERVATION
Two macrophage migration inhibitory factors regulate
starsh larval immune cell chemotaxis
Ryohei Furukawa1, Kana Tamaki2and Hiroyuki Kaneko1
Immune cell recruitment is critical step in the inammatory response and associated diseases. However, the underlying
regulatory mechanisms are poorly understood in invertebrates. Mesenchyme cells of the starsh larvae, which allowed
Metchnikoff to complete his landmark experiments, are important model for analysis of immune cell migration. The present
study investigated the role of macrophage migration inhibitory factor (MIF)an evolutionarily conserved cytokine that is
functionally similar to chemokinesin the larvae of the starsh Patiria (Asterina)pectinifera, which were found to possess two
orthologs, ApMIF1 and ApMIF2. ApMIF1 and ApMIF2 clustered with mammalian MIF and its homolog D-dopachrome
tautomerase (DDT), respectively, in the phylogenetic analysis. In contrast to the functional similarity between mammalian MIF
and DDT, ApMIF1 knockdown resulted in the excessive recruitment of mesenchyme cells in vivo, whereas ApMIF2 deciency
inhibited the recruitment of these cells to foreign bodies. Mesenchyme cells migrated along a gradient of recombinant ApMIF2
in vitro, whereas recombinant ApMIF1 completely blocked ApMIF2-induced directed migration. Moreover, the expression patterns
of ApMIF1 and ApMIF2 messenger RNA in bacteria-challenged mesenchyme cells were consistent with in vivo observations of
cell behaviors. These results indicate that ApMIF1 and ApMIF2 act as chemotactic inhibitory and stimulatory factors,
respectively, and coordinately regulate mesenchyme cell recruitment during the immune response in starsh larvae. This is the
rst report describing opposing functions for MIF- and DDT-like molecules. Our ndings provide novel insight into the
mechanisms underlying immune regulation in invertebrates.
Immunology and Cell Biology (2016) 94, 315321; doi:10.1038/icb.2016.6; published online 2 February 2016
The starsh is an echinoderm that is phylogenetically close to the
ancestral chordate. The transparent larvae possess a simple body
structure, which allowed Metchnikoff to complete his landmark
experiments that led to his theories of phagocytosis and
chemotaxis.1We previously described in detail the characteristics of
Patiria (Asterina)pectinifera mesenchyme cells, such as their distribu-
tion patterns and role in host defense.2,3 Most mesenchyme cells are
distributed beneath the larval body wall and form a dynamic network
structure while moving randomly. When a foreign substance is
injected into the larval blastocoel, it is completely encapsulated and/
or phagocytosed by mesenchyme cells converging from various
directions within 2 h. Based on these characteristics, mesenchyme
cells are functionally equivalent to blastocoelar cells in larval echinoids,
such as those arising from a subset of secondary mesenchyme cells in
sea urchin.4,5 Signicantly, the number of recruited mesenchyme cells
is dependent on the amount and size of the foreign body,2suggesting
that this process is strictly regulated by the starsh larval immune
system.
Chemokines are essential in the mammalian immune system as
leukocyte recruitment and arrest are critical steps in the inamma-
tory response and associated diseases.6On the other hand, little is
known about the mechanisms of chemotactic migration during
the invertebrate immune response. A recent comparative whole-
genome analysis failed to identify putative chemokine or chemokine
receptors in invertebrates,7implying that invertebrate chemotaxis is
regulated by chemokine-like function chemokines, which have func-
tions similar to those of classic chemokines but lack their typical
structure.8
Macrophage migration inhibitory factor (MIF) is a chemokine-like
function chemokine that is evolutionarily ancient and highly
conserved.911 In mammals, MIF directly exerts chemokine-like
functions via the CXC chemokine receptors CXCR2 and
CXCR4,12,13 and indirectly induces leukocyte arrest.1416 Thus, MIF
acts as a pleiotropic inammatory cytokine with critical roles in
physiological immunity as well as inammatory diseases and cancer.8
MIF activity is shared by D-dopachrome tautomerase (DDT); this
includes binding the same receptor complex and induction of similar
cell signaling and effector functions.1719 In contrast, the function of
invertebrate MIF and DDTespecially in the immune systemis not
well understood.
We predicted that the starsh MIF ortholog regulates the recruit-
ment of larval mesenchyme cells in the immune response. In the
1
Department of Biology, Research and Education Center for Natural Sciences, Keio University, Kanagawa, Japan and
2
School of Fundamental Science and Technology, Graduate
School of Science and Technology, Keio University, Kanagawa, Japan
Correspondence: Dr R Furukawa, Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 2-1-1 Nishitokuda,
Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan.
E-mail: rfuru@iwate-med.ac.jp or furyohei@gmail.com
Received 27 October 2015; revised 7 January 2016; accepted 7 January 2016; published online 2 February 2016
Immunology and Cell Biology (2016) 94, 315321
&
2016 Australasian Society for Immunology Inc. All rights reserved 0818-9641/16
www.nature.com/icb
present study, we identied two MIF orthologs, designated as ApMIF1
and ApMIF2, and characterized their functions in the context of
immunoreactive migration of mesenchyme cells in vivo and in vitro.
We also propose a regulatory model for mesenchyme cell recruitment
by ApMIF1 and ApMIF2 in the starsh larval immune response.
RESULTS
Structural features of ApMIF1 and ApMIF2
Over 56 000 P. pectinifera expressed sequence tags have been sub-
mitted to the DNA Data Bank of Japan (http://www.ddbj.nig.ac.jp)
without annotations. Using the protein sequences of human
macrophage MIF and its homolog DDT to search this database using
the TBLASTN program, we identied two MIF orthologs in
P. pectinifera, hereafter referred to as ApMIF1 and ApMIF2 (GenBank
accession nos. LC062600 and LC062601, respectively). Based on the
obtained expressed sequence tag sequences, we isolated full-length
complementary DNA (cDNA)-encoding ApMIF1 and ApMIF2 from
the cDNA library of mesenchyme cells, and determined that they
encode 116- and 120-amino acid polypeptides, respectively. The
deduced amino acid sequences revealed two α-helices and six β-
strands, which are characteristic of other MIF family members
(Figure 1a). To assess the evolutionary relationships within the
deuterostome MIF family, we performed phylogenetic analysis of
12 MIF orthologs by applying the widely used neighbor-joining
and maximum likelihood methods (Figure 1b). Two distinct types
of deuterostome MIFs were observed with both methods: ApMIF1
clustered with vertebrate MIF but was distantly related to ApMIF2 and
vertebrate DDT.
Human MIF is a non-cognate ligand of CXCR2 and CXCR4.12 The
molecular details of the MIF/CXCR4 interaction are not well under-
stood; however, the pseudo-(E)LR motif and N-like loop are required
for the functioning of human MIF as a non-canonical CXCR2
ligand.13,20 The pseudo-(E)LR motif in MIF is characterized by Asp-
45 and Arg-12 (Figure 1a, arrowheads), and forms a three-
dimensional structure resembling the N-terminal GluLeuArg
(ELR) motif present in a subgroup of CXC chemokines. The N-like
loop in MIF spans 10 amino acids from positions 47 to 56, but is
structurally distinct from that of CXC chemokines (Figure 1a, black
bar). Interestingly, the N-like loop of ApMIF1 was 80% identical to
that of human MIF, whereas the loop in ApMIF2 was 40 and 30%
identical to those of human MIF and DDT, respectively. In addition, a
pseudo-(E)LR motif was found in ApMIF2 but not in ApMIF1.
ApMIF1 or ApMIF2 knockdown affects the recruitment of
mesenchyme cells during the immune response
We tested the efcacy of ApMIF1 and ApMIF2 knockdown using
morpholino antisense oligonucleotides (MO) for loss-of-function
experiments. Eggs injected with each of the MOs showed normal
morphogenesis, including distribution and migration of mesenchyme
cells in the blastocoel, formation of the mouth and
Figure 1 ApMIF1 and ApMIF2 sequences and phylogenetic relationships. (a) Sequence alignment of ApMIF1 and ApMIF2 with human MIF and DDT. The
locations of α-helices and β-strands are indicated; the N-like loop is indicated by a black bar; and the pseudo-(E)LR motif in ApMIF2 is indicated by
arrowheads. Alignment of the N-like loop is also shown in a box, and hyphens denoting identical residues. (b) Phylogenetic trees of deuterostome MIFs using
either the neighbor-joining method (left) or the maximum likelihood method (right). All ambiguous positions were removed for each sequence pair. The
percentage of trees in which associated taxa clustered together in the bootstrap test (1000 replicates) is shown next to the branches. The collapsed branch
in the Maximum Likelihood Tree has a bootstrap value o50. Scale bars: left, evolutionary distance; right, substitutions per site.
Regulation of chemotaxis by starsh MIF
RFurukawaet al
316
Immunology and Cell Biology
compartmentalization of the digestive tract (Supplementary Figure
S1a). However, compared with larvae injected with control MO,
ApMIF1-MO- and ApMIF2-MO-injected larvae were smaller and had
fewer cells (Supplementary Figures S1a and c). This decrease in size
was rescued by co-injecting the corresponding messenger RNA
(mRNA) (Supplementary Figure S1b), conrming the specicity of
knockdown by each MO. In subsequent experiments, ApMIF1 and
ApMIF2 loss of function was determined as a larval size of o700 μm
in length.
To examine the roles of ApMIF1 and ApMIF2 in the immune
responseespecially the migration of mesenchyme cellswe injected
a large oil droplet (diameter =20 μm) into the center of the head
blastocoel of morphants, which is the area that is furthest from the
body wall (~5080 μm from the body wall; Supplementary Figure
S1a). In vivo time-lapse observation revealed an exaggerated immune
response in ApMIF1-MO larvae, with excessive recruitment
of mesenchyme cells as compared with control-MO larvae
(Supplementary Movies S1 and S2). In contrast, fewer mesenchyme
cells were present near the oil droplet in ApMIF2-MO than in control-
MO larvae (Supplementary Movies S1 and S3). A quantitative analysis
of mesenchyme cells recruited to the oil droplet revealed that the total
number of cells attached to or surrounding the oil droplet increased
over time in control-MO larvae (Figures 2a and b); 2 h after injection,
68 cells were attached to the droplet (Figure 2a), consistent with
previous observations.2By comparison, the number of mesenchyme
cells surrounding the oil droplet was higher in ApMIF1-MO larvae
and reached a plateau 45 min after oil droplet injection, with similar
numbers observed up to 2 h after injection. In contrast, in ApMIF2-
MO larvae, fewer cells contacted the oil droplet as compared with
control-MO larvae 45 min after injection, and this did not change
until the end of the observation period.
Recombinant ApMIF1 and ApMIF2 affect mesenchyme cell
migration
To assess the contribution of ApMIF1 and ApMIF2 to the migration of
mesenchyme cells, we prepared the recombinant proteins rMIF1 and
rMIF2 (Supplementary Figure S2) and carried out a chemotactic
migration assay with a Zigmond chamber. To eliminate the inuence
of endogenous MIFs in the assay, we prepared mesenchyme cells from
larvae in which both ApMIF1 and ApMIF2 were knocked down. Cell
migration was evaluated by tracking the movements of individual cells
in the presence of recombinant rMIF1 and rMIF2 (Figure 3). As the
fusogenic nature and network-forming ability of mesenchyme cells
made it difcult to accurately track their migration, we excluded cells
that fused to each other during the observation period.
Exposure to rMIF2 but not rMIF1 increased cell migration speed
and cell directionality within 20 min (Figures 3a and b). When rMIF2
was applied as a gradient, cells migrated toward the source (Figure 3c,
top). These migration patterns can be seen in Rose plots that show the
frequency distribution of cell orientations (Figure 3c, bottom).
Rayleigh tests conrmed that mesenchyme cells migrated randomly
when rMIF2 concentration was uniform (P40.05), similar to cells in
articial seawater (ASW). However, when rMIF2 was applied as a
gradient, the direction of migration was strongly biased toward the
region of high-rMIF2 concentration (Figure 3c), Po0.05. On the
other hand, when an rMIF2 gradient was formed under a homo-
geneous concentration of rMIF1, mesenchyme cells maintained a
random migration pattern (P40.05, Rayleigh test), and velocity and
directionality were similar to those of migrating cells in ASW.
Figure 4 shows the change in migration speed every 10 min during
the chemotaxis assay. In the presence of an rMIF2 gradient, the
migration speed of mesenchyme cells gradually increased and attained
a maximum after 3040 min. However, when both rMIF1 and rMIF2
were applied, no signicant difference was noted in migration speed
relative to cells in ASW at any time point.
ApMIF1 and ApMIF2 mRNA expression levels are differentially
upregulated by bacterial challenge
To ascertain whether gene expression under immune stimulation can
explain the mesenchyme cell behaviors regulated by the recombinant
protein, we examined changes in temporal mRNA expression proles
of ApMIF1 and ApMIF2 in response to bacterial challenge. ApMIF1
mRNA levels were increased about 4.3-fold at 5 min as compared with
that observed in naive mesenchyme cells at 0 min and remained
constant until 90 min, when the levels became extremely high
(41700-fold higher than baseline) (Figure 5a). ApMIF2 mRNA
expression increased ~ 100-fold within 15 min, and decreased after
90 min (Figure 5a). The mRNA expression of ApMIF2 was higher
than that of ApMIF1 until 60 min; however, after 90 min, the level
of ApMIF1 mRNA was ~ 10-fold higher than that of ApMIF2
(Figure 5b).
Figure 2 Recruitment of mesenchyme cells to injected oil droplets. (a) Number of mesenchyme cells attached to oil droplets. (b) Number of mesenchyme
cells within 20 μm of the surface of the oil droplet without contact. **Po0.01, ***Po0.001 (ttest; n=27 (control-MO), 37 (ApMIF1-MO) and 37
(ApMIF2-MO)).
Regulation of chemotaxis by starsh MIF
R Furukawa et al
317
Immunology and Cell Biology
DISCUSSION
In this study, we investigated the functional properties of two starsh
MIF orthologs, ApMIF1 and ApMIF2, in the context of immune cell
recruitment in larvae. ApMIF1 and ApMIF2 clustered with MIF and
DDT, respectively, in the phylogenetic analysis. In contrast to the
functional similarity between mammalian MIF and DDT, the in vitro
chemotactic migration assay revealed that rMIF1 and rMIF2 had
antagonistic functions, with the former inhibiting and the latter
inducing directed mesenchyme cell migration. This result was con-
sistent with in vivo observations that ApMIF2 knockdown abrogated
mesenchyme cell migration toward a foreign body, whereas ApMIF1
deciency led to excessive cell recruitment. Moreover, the expression
levels of ApMIF1 and ApMIF2 mRNA, which were upregulated by
bacterial stimulation, mirrored mesenchyme cell behaviors in vivo.
These results indicate that ApMIF1 and ApMIF2 act as chemotaxis
Figure 3 Regulation of chemotactic mesenchyme cell migration by ApMIF1 and ApMIF2. (ac) Cells seeded in the Zigmond chamber were tracked by time-
lapse microscopy for 1 h in ASW, a homogeneous concentration of rMIF1 (rMIF1 constant), a homogeneous concentration of rMIF2 (rMIF2 constant), a
gradient of rMIF2 (rMIF2 gradient) or a gradient of rMIF2 in a homogeneous concentration of rMIF1 (rMIF1 constant+rMIF2 gradient). Cell velocity (that is,
accumulated distance/observation time) (a) and directionality (that is, Euclidean distance/accumulated distance) (c) were calculated from tracks o f individual
migrating cells (plotted in the upper row of c). **Po0.01 (ttest). Rose diagrams (bottom row of c) were also plotted to visualize the distribution of migration
angles. The Rayleigh test was used to evaluate unimodal distribution of cell direction at end points. P40.05 was considered as a uniform distribution
(random migration).
Figure 4 Changes in mesenchyme cell migration speed. Migration speed per
10 min was calculated from tracks of individual migrating cells shown in
Figure 3c. **Po0.01 vs ASW control (ttest).
Regulation of chemotaxis by starsh MIF
RFurukawaet al
318
Immunology and Cell Biology
inhibitory and stimulatory factors in the larval immune system,
respectively.Thisistherst report describing opposing functions
for MIF- and DDT-like molecules in the immune response.
Based on these ndings, we generated a model of mesenchyme
cell recruitment by ApMIF1 and ApMIF2 (Figure 6). The rst
contact between randomly migrating cells and a foreign body triggers
a dramatic upregulation of ApMIF2 gene expression and
ApMIF2 secretion, creating an ApMIF2-rich environment around
the foreign substance. In ApMIF1 morphants, the number of recruited
mesenchyme cells reached a plateau 45 min after the oil droplet
injection, suggesting that all cells within the range of ApMIF2 activity
had been recruited by that time. This activity range was estimated to
be 70 μm based on the migration speed of rMIF2-treated mesenchyme
cells (1.42.2 μmmin1; Figure 4), which corresponds to the distance
from the body wall to the central area of the blastocoel where the oil
droplet was injected (5080 μm, Supplementary Figure S1a). This
indicates that ApMIF2 can recruit a sufcient number of mesenchyme
cells from the body wall where they are mainly distributed irrespective
of where the foreign body is located. On the other hand, ApMIF1 is
secreted by cells undergoing chemotactic migration to inhibit the
activity of ApMIF2 and prevent the recruitment of more distant cells.
Indeed, the upregulation of ApMIF1 and ApMIF2 mRNA expression
followed similar trends. Finally, when an appropriate number of
mesenchyme cells had been recruited, ApMIF1 completely inhibited
ApMIF2 activity. According to our model, the activities of the two
MIFs can regulate the recruitment of a sufcient number of
mesenchyme cells for the amount and size of the foreign body.2
Intriguingly, the cooperative regulation of the chemotaxis by
ApMIF1 and ApMIF2 implies a trade-off between efciency and risk
avoidance. The requirement for inhibition by ApMIF1 indicates that
the immune system of starsh larvae is at risk for excessive or
insufcient accumulation of mesenchyme cells if there is an imbalance
in the levels of ApMIF1 and ApMIF2. Risks of this nature are a feature
of various cytokine systems; for example, sepsis development is closely
related to an imbalance between the levels of pro- and anti-
inammatory cytokine including MIF.21 On the other hand, we
previously reported that foreign materials can easily invade the
blastocoel of starsh larvae through the body wall;2given this
susceptibility, the inhibitory effect of ApMIF1 might contribute to
preserving part of the structural network, providing protection against
sequential invasion of multiple foreign bodies. Although the antag-
onistic activities of cytokines can be very effective for a subset of
immune cells to localize immune responses, there is a constant risk of
immunological disequilibrium even in a relatively primitive animal.
We recently identied the P. pectinifera ortholog of mammalian
dedicator of cytokinesis (DOCK)2, an intracellular regulator of
mesenchyme cell migration in the immune response.3DOCK2 is
activated downstream of the non-cognate MIF receptors CXCR2 and
CXCR412 to regulate immune cell chemotaxis.22,23 Our previous work
revealed that loss of ApDOCK function also resulted in the failure of
mesenchyme cell recruitment,3similar to ApMIF2-MO larvae
Figure 5 Temporal changes in ApMIF1 and ApMIF2 mRNA expression in cultured mesenchyme cells following bacterial challenge. (a) Relative expression
levels of ApMIF1 and ApMIF2 mRNA in bacteria-challenged and naive mesenchyme cells (0 min). (b) Temporal changes in the ratio of ApMIF2 to ApMIF1
mRNA expression (n=3).
Figure 6 Model of the control of chemotaxis by ApMIF1 and ApMIF2.
Mesenchyme cells move randomly before an immune response. Within
20 min of invasion of a foreign body (that is, an oil droplet) (a), a nearby
mesenchyme cell makes initial contact with the foreign body and secretes
ApMIF2 (b). Adjacent cells are activated by ApMIF2 binding and move along
the ApMIF2 gradient toward the source (c). Migrating cells secrete ApMIF1,
which inhibits chemotaxis in other more distal mesenchyme cells (c).
Consequently, an immune response is initiated by an appropriate number of
mesenchyme cells (d).
Regulation of chemotaxis by starsh MIF
R Furukawa et al
319
Immunology and Cell Biology
(Figure 2 and Supplementary Movie S3), implying that ApMIF2
triggers while ApMIF1 inhibits ApDOCK-mediated signaling. Despite
the evolutionarily conserved roles of MIF and DOCK family members
in immune cell recruitment, to date there have been no orthologs of
mammalian MIF receptors identied in invertebrates. This raises the
possibility of conserved canonical MIF receptor(s); given that ApMIF1
and ApMIF2 have motifs for chemokine receptor binding, other types
of G protein-coupled receptor superfamily members may be con-
sidered as candidates.
In summary, the identication of starsh MIF receptor(s) not only
provides insight into the evolutionary conservation and diversity of
MIF function, but it also claries the mechanism by which ApMIF1
and ApMIF2 exert their opposing functions. In addition, the starsh
MIF receptor(s) can provide a useful tool for investigating treatment
strategies for MIF-related diseases.
METHODS
Animals
Adult P. pectinifera were collected along the coast of Japan and maintained at
15 °C in a glass aquarium lled with ASW (MarineArt SF-1; Tomita
Pharmaceutical, Tokushima, Japan). Developing larvae were obtained as
previously described.2In brief, mature eggs prepared by treatment with 1-
methyladenine (Sigma-Aldrich, Tokyo, Japan) were fertilized with diluted
sperm, and were allowed to develop in ASW at 20 °C.
Cloning and identication of ApMIF1 and ApMIF2
To identify starsh orthologs of MIF family members, we carried out
TBLASTN searches of456 000 expressed sequence tags of P. pectinifera eggs
and gastrulae deposited in the DNA Data Bank of Japan (GenBank accession
no. DB384948DB441778) using amino acid sequences of human MIF
(NP_002406) and DDT (NP_001346) as queries with e-value scoreso1e 5.
In total, 57 candidate sequences were obtained from the TBLASTN hits that
encoded two different sequences. Based on these sequences, we designed the
following two primer pairs: 5-CAG CGA ATC TTC CTG AAT TGT CAA
G-3/5-CAG TTT TCA CGG CTA CAC TAA CTT G-3and 5-GTG
GAG AAA TTG CTT GTC GTA-3/5-GGG AGT TGG GGT GGT GCA
G-3.Amplied cDNAs were subcloned from a cDNA library of cultured
mesenchyme cells into the pGEM-T vector (Promega, Tokyo, Japan) and
sequenced as previously described.24 Deduced amino acid sequences were
analyzed with the BLAST algorithm, and we identied one ortholog each of
MIF and DDT. Phylogenetic trees were reconstructed with the neighbor-joining
and maximum likelihood methods based on CLUSTALW-aligned MIF/DDT
sequences using MEGA6 software.25 All positions containing gaps and missing
data were eliminated. For the neighbor-joining tree, evolutionary distances were
computed with the p-distance amino acid substitution model using uniform
rates among sites and pairwise deletion. For the maximum likelihood tree, we
applied the LG+G model, which was the best t substitution model estimated
by the lowest Bayesian Information Criterion scores, and an initial tree for the
heuristic search was generated automatically by applying Neighbor-Joining and
BioNJ algorithms to a matrix of estimated pairwise distances. Both methods use
the bootstrap method with 1000 replicates for phylogeny testing.
Morpholino oligonucleotides
MOs were purchased from Gene Tools (Philomath, OR, USA). ApMIF1- and
ApMIF2-MO sequences were 5-TCTACGATCGGCATCTTGACAATTC-3
and 5-TCG CAC AGAGGCATGTTGATTCTTG-3, respectively, and were
complementary to mRNA sequences containing the translation start site
(underlined). Standard Control Morpholino Oligo (Gene Tools) was used as
the control-MO. MOs were dissolved in sterile water at 1 mMto obtain a stock
solution, which was diluted to 240 μMbefore injection. For simultaneous
knockdown of ApMIF1 and ApMIF2, equal volumes of each MO at 480 μM
were mixed. In brief, follicle cells were removed from immature eggs using
calcium-free seawater (Jamarin, Osaka, Japan), and the eggs were injected with
each MO in a microinjection chamber.26 The detailed injection procedure has
been described elsewhere.27
Microinjection and in vivo analysis of mesenchyme cell migration
Silicone oil (Shin-Etsu Chemical Co., Tokyo, Japan) was injected into larvae as
previously described.2Injected larvae were immobilized and observed at 20 °C
by time-lapse using an IX71 light microscope (Olympus, Tokyo, Japan)
controlled with DP controller software (Olympus).3Images were acquired
every 2 min for 2 h. The larvae were then labeled with a monoclonal antibody
against MC5a membranous metalloproteinase of the astacin family27 and
mesenchyme cell markercombined with propidium iodide nuclear staining
(0.1 μgml
1; Wako Pure Chemical Industries, Osaka, Japan).2Specimens were
visualized by laser confocal microscopy with FluoView software (Olympus).
Preparation of recombinant proteins
DNA fragments encoding ApMIF1 or ApMIF2 were amplied using the
following forward and reverse primer pairs: for rMIF1, 5-CAC CAT
GCC GAT CGT AGA-3and 5-TCA TAT TTT GCC AGC AAG TTT
GTC-3and for rMIF2, 5-CAC CAT GCC TCT GTG CGA GCT GAA-3and
5-TTA TTT CAG TTG GCT GGC GAG TTT GCC-3. PCR products were
subcloned into the pET100/D-TOPO vector (Invitrogen, Tokyo, Japan), and
the recombinant proteins (rMIF1 and rMIF2) were expressed in Escherichia coli
strain BL21 and puried by metal-chelate afnity chromatography using the
His60 Ni Gravity Column Purication kit (Clontech, Shiga, Japan) according to
the manufacturers instructions.
Chemotaxis assay
Mesenchyme cells were obtained from morphant or control larvae as previously
described.3The chemotaxis assay was performed using a Zigmond chamber
(Funakoshi, Tokyo, Japan). The assembled chamber contained a gap of exactly
5μm between the surface of the bridge and coverslip on which cells were
cultured. An rMIF2 gradient was formed by placing ASW and 10 nMrMIF2-
containing ASW in separate wells. In the case of no gradient, both wells were
lled with ASW or 10 nMrMIF-containing ASW. To obtain a stable gradient,
the chamber was allowed to stand for 3060 min at 20 °C under humid
conditions before observation. Time-lapse images were acquired every 2 min
for 1 h. Mesenchyme cells were tracked in two independent experiments using
the Manual Tracking plugin of ImageJ software (National Institutes of Health,
Bethesda, MD, USA). Tracks were analyzed using the Chemotaxis and
Migration Tool (Image J) to obtain cell speed and directionality, which
represents a ratio of Euclidean distance to accumulated distance and shows
the direction of migration. The Rayleigh test for unimodal direction clustering
was used to determine whether the cell population showed directionality along
the chemotactic gradient;28 the distribution of directions was considered
uniform for P40.05.
Bacterial challenge
Cultured E. coli were adjusted to a concentration of OD600=0.5 in ASW and
boiled for 5 min. A 100-μl volume of E. coli-ASW was added to cultured
mesenchyme cells in a plastic culture dish (diameter, 6 cm; BD, Tokyo, Japan)
with 5 ml ASW.
Real-time PCR
Total RNA isolation and a cDNA synthesis were performed as previously
described.24 A140-bpfragmentoftheApMIF1 gene and a 137-bp fragment of
the ApMIF2 gene were amplied using the primer pairs 5-TGC CCA
AGT CAT CAC AGA AA-3/5-TTT TGC CAG CAA GTT TGT CA-3and
5-CAG CGG ATT CAG CAG TAA ACA C-3/5-TTG CCT CAA TGA
GAT GAA AGG A-3, respectively. As a reference gene, a 114-bp fragment of
the P. pectinifera 18 S rRNA gene was amplied using the previously reported
18 S-3-f and 18 S-3-r primers.24 Real-time PCR was carried out using the
KAPA SYBR FAST qPCR master mix (Kapa Biosystems, Woburn, MA, USA)
and a StepOne Real-Time PCR system (Applied Biosystems, Foster City,
CA, USA).
Regulation of chemotaxis by starsh MIF
RFurukawaet al
320
Immunology and Cell Biology
CONFLICT OF INTEREST
The authors declare no conict of interest.
ACKNOWLEDGEMENTS
We thank members of the Asamushi Marine Biological Station of Tohoku
University for supplying the starsh. We also thank members of the Tateyama
Marine Laboratory, Marine Coastal Research Center, Ochanomizu University
for maintaining the adult starsh. We are particularly grateful to Drs Hiroshi
Takeda, Motonori Hoshi, Midori Matsumoto, Ritsu Kuraishi and Kazuya
Kobayashi for critical discussions. This work was supported by Keio Gijuku
Academic Development Funds (to RF and HK).
1MetchnikoffE.Lectures on the Comparative Pathology of Inammation, Delivered at
Pasteur Institute: Dover: New York, 1891 (reprinted in 1968).
2 Furukawa R, Takahashi Y, Nakajima Y, Dan-Sohkawa M, Kaneko H. Defense system by
mesenchyme cells in bipinnaria larvae of the starsh, Asterina pectinifera. Dev Comp
Immunol 2009; 33:205215.
3 Furukawa R, Funabashi H, Matsumoto M, Kaneko H. Starsh ApDOCK protein
essentially functions in larval defense system operated by mesenchyme cells. Immunol
Cell Biol 2012; 90:955965.
4 Tamboline CR, Burke RD. Secondary mesenchyme of the sea urchin embryo: ontogeny
of blastocoelar cells. J Exp Zool 1992; 262:5160.
5 Silva JR. The onset of phagocytosis and identity in the embryo of Lytechinus variegatus.
Dev Comp Immunol 2000; 24:733739.
6 Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inammation: the
leukocyte adhesion cascade updated. Nat Rev Immunol 2007; 7:678689.
7 DeVries ME, Kelvin AA, Xu L, Ran L, Robinson J, Kelvin DJ. Dening the origins and
evolution of the chemokine/chemokine receptor system. J Immunol 2006; 176:
401415.
8 Tillmann S, Bernhagen J, Noels H. Arrest functions of the MIF ligand/receptor axes in
atherogenesis. Front Immunol 2013; 4:115.
9 Sato A, Uinuk-ool TS, Kuroda N, Mayer WE, Takezaki N, Dongak R et al. Macrophage
migration inhibitory factor (MIF) of jawed and jawless shes: Implications for its
evolutionary origin. Dev Comp Immunol 2003; 27:401412.
10 Vermeire JJ, Cho Y, Lolis E, Bucala R, Cappello M. Orthologs of macrophage migration
inhibitory factor from parasitic nematodes. Trends Parasitol 2008; 24:355363.
11 Li F, Huang S, Wang L, Yang J, Zhang H, Qiu L et al. A macrophage migration inhibitory
factor like gene from scallop Chlamys farreri: involvement in immune response and
wound healing. Dev Comp Immunol 2011; 35:6271.
12 Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR et al. MIF is a
noncognate ligand of CXC chemokine receptors in inammatory and atherogenic cell
recruitment. Nat Med 2007; 13:587596.
13 Kraemer S, Lue H, Zernecke A, Kapurniotu A, Andreetto E, Frank R et al. MIF-
chemokine receptor interactions in atherogenesis are dependent on an n-loop-based 2-
site binding mechanism. FASEB J 2011; 25:894906.
14 Gregory JL, Leech MT, David JR, Yang YH, Dacumos A, Hickey MJ. Reduced leukocyte-
endothelial cell interactions in the inamed microcirculation of macrophage migration
inhibitory factor-decient mice. Arthritis Rheum 2004; 50: 30233034.
15 Fan H, Hall P, Santos LL, Gregory JL, Fingerle-Rowson G, Bucala R et al. Macrophage
migration inhibitory factor and CD74 regulate macrophage chemotactic responses via
MAPK and rho GTPase. JImmunol2011; 186:49154924.
16 Santos LL, Fan H, Hall P, Ngo D, MacKay CR, Fingerle-Rowson G et al. Macrophage
migration inhibitory factor regulates neutrophil chemotactic responses in inammatory
arthritis in mice. Arthritis Rheum 2011; 63:960970.
17 Coleman AM, Rendon BE, Zhao M, Qian M-WW, Bucala R, Xin D et al. Cooperative
regulation of non-small cell lung carcinoma angiogenic potential by macrophage
migration inhibitory factor and its homolog, D-dopachrome tautomerase. JImmunol
2008; 181: 23302337.
18 Xin D, Rendon BE, Zhao M, Winner M, Coleman AM, Mitchell RA. The MIF homologue
D-dopachrome tautomerase promotes COX-2 expression through beta-catenin-
dependent and - independent mechan isms. Mol Cancer Res 2010; 8:160116 09.
19 Merk M, Zierow S, Leng L, Das R, Du X, Schulte W et al. The D-dopachrome
tautomerase (DDT) gene product is a cytokine and functional homolog of macrophage
migration inhibitory factor (MIF)..Proc Natl Acad Sci USA 2011; 108:E577E585.
20 Weber C, Kraemer S, Drechsler M, Lue H, Koenen RR, Kapurniotu A et al. Structural
determinants of MIF functions in CXCR2-mediated inammatory and atherogenic
leukocyte recruitment. Proc Natl Acad Sci USA 2008; 105: 1627816283.
21 Schulte W, Bernhagen J, Bucala R. Cytokines in sepsis: potent immunoregulators
and potential therapeutic targetsan updated view. Mediators Inamm 2013; 2013:
165974.
22 Reif K, Cyster J. The CDM protein DOCK2 in lymphocyte migration. Trends Cell Biol
2002; 12:368373.
23 Sai J, Raman D, Liu Y, Wikswo J, Richmond A. Parallel phosphatidylinositol 3-kinase
(PI3K)-dependent and Src-dependent pathways lead to CXCL8-mediated Rac2 activa-
tion and chemotaxis. JBiolChem2008; 283:2653826547.
24 Furukawa R, Matsumoto M, Kaneko H. Characterization of a scavenger receptor
cysteine-rich-domain-containing protein of the starsh, Asterina pectinifera:ApSRCR1
acts as an opsonin in the larval and adult innate immune systems. Dev Comp Immunol
2012; 36:5161.
25 Tamura K, Stecher G, Peterson D, Filipski A, Kumar S. MEGA6: Molecular evolutionary
genetics analysis version 6.0. MolBiolEvol2013 ; 30:27252739.
26 Keihart O. Microinjection of echinoderm eggs: apparatus and procedures. Methods Cell
Biol 1982; 25:1331.
27 Hamanaka G, Matsumoto M, Hoshi M, Kaneko H. Studies on function of MC5
molecule that is a novel membrane-type metalloproteinase of astacin family
during morphogenesis of the starsh, Asterina pectinifera. In: Harris LG, Bottger SA,
Walker CW, Lesser MP (eds). Echinoderms. Taylor & Francis Group: London. 2 010,
pp 457462.
28 Zicha D, Dunn GA. Are growth factors chemotactic agents? Exp Cell Res 1995; 221:
526529.
The Supplementary Information that accompanies this paper is available on the Immunology and Cell Biology website (http://www.nature.com/icb)
Regulation of chemotaxis by starsh MIF
R Furukawa et al
321
Immunology and Cell Biology
... Ainsi, une première étude fonctionnelle a démontré que BgMIF1 du mollusque Biomphalaria glabrata présente certaines activités biologiques majeures du MIF humain, et participe à la réponse immunitaire après infection du mollusque par le parasite Schistosoma mansoni [16]. Différentes études ont ensuite identifié des MIFs chez d'autres invertébrés libres comme d'autres espèces de mollusques, des crustacés ou des échinodermes [17][18][19]. De façon inattendue, un travail de notre équipe a récemment montré que des parasites de plantes comme les pucerons du pois et du pêcher (Acyrthosiphon pisum et Myzus persicae) injectent également des MIFs dans leur plante hôte. Ces MIFs de pucerons modulent la réponse immunitaire de la plante et sont indispensables à l'établissement de l'infection [20]. ...
... Depuis, différentes études ont été réalisées sur d'autres espèces d'invertébrés comme des échinodermes (Patiria pectinifera [17] ), des arthropodes, des mollusques et des helminthes [11,18,19,35,36]. ...
... Chez l'ensemble des espèces étudiées, les MIFs jouent un rôle dans l'immunité de l'organisme. Elles contribuent notamment au recrutement de cellules nécessaires à la mise en place d'une réponse immunitaire chez Haliotis diversicolor [111] (couramment appelé l'ormeau) et chez Patiria pectinifera [17], une étoile de mer. ...
Thesis
Les cytokines MIFs (Macrophage Migration Inhibitory Factor) sont des protéines multifonctionnelles qui, chez les mammifères, interviennent dans plusieurs processus majeurs tels que le contrôle du cycle et de la mobilité cellulaire, l’activation de la réponse immunitaire et l’inhibition de l’apoptose. Des travaux récents montrent que les protéines MIFs peuvent également jouer un rôle majeur dans l’immunité des invertébrés, et être utilisées par des organismes parasites d’animaux ou de végétaux pour inhiber les défenses de leurs hôtes respectifs, ce qui soulève la question de leur diversité, de leur histoire évolutive et des potentielles différences fonctionnelles. L’objectif général de ce travail de thèse était d’explorer la diversité et l’histoire évolutive des protéines MIFs à une échelle trans-règne, puis de rechercher leurs éventuelles différences fonctionnelles, en se focalisant sur les systèmes plantes-pathogènes. Nous avons tout d’abord identifié les MIFs chez 803 espèces de plantes, champignons, protistes, et métazoaires, et analysé leur présence/absence et histoire évolutive en fonction des taxa, de l’écologie et du mode de vie (libre ou parasitaire) des espèces. Nous avons montré que l’histoire évolutive des MIFs, chez les eucaryotes, est complexe et implique des duplications ancestrales ainsi que des pertes multiples ou des re-duplications récentes. Les plantes (espèces libres autotrophes) et les parasites de plantes (autres que champignons) possèdent un nombre médian de trois MIFs, alors que les espèces hétérotrophes et les parasites d’animaux ont un nombre de MIF plus faible et/ou plus variable. De plus, les protéines MIFs semblent essentielles et fortement conservées, avec de nombreux résidus sous sélection purifiante, chez certains groupes comme les plantes, alors que dans d’autres groupes, elles semblent facultatives (e.g. champignons) ou présentes en plusieurs copies divergentes (e.g. nématodes, insectes), ce qui suggère de potentielles néofonctionalisations. Nous avons ensuite analysé l’effet des protéines MIFs de plusieurs espèces sur la mort cellulaire en système végétal. Tous les organismes testés (plantes oomycètes, protozoaires, insectes et nématodes), y compris ceux n’ayant pas d’interaction avec les plantes, possèdent au moins une protéine MIF capable d’inhiber cette mort cellulaire. Cela suggère que l’inhibition de la mort cellulaire en plante ne correspond pas à une néofonctionalisation des MIFs de parasites de plantes, mais serait liée à des propriétés structurales et conservées des MIFs. Toutefois, aucun des paramètres étudiés (localisation subcellulaire) ou prédits in silico (présence de motifs, structures 3D, oligomérisation, modifications post-traductionnelles) ne semble lié à cette activité d’inhibition de la mort cellulaire. De futures études fonctionnelles poussées sont nécessaires à l’élucidation des relations structure/fonction de ces protéines complexes.
... Candidate genes involved in this regulation appear to be two macrophage migration inhibitory factors (MIFs): ApMIF2 and ApMIF1 (Furukawa et al., 2016). These immune effectors are evolutionarily ancient and highly conserved (Furukawa et al., 2016). ...
... Candidate genes involved in this regulation appear to be two macrophage migration inhibitory factors (MIFs): ApMIF2 and ApMIF1 (Furukawa et al., 2016). These immune effectors are evolutionarily ancient and highly conserved (Furukawa et al., 2016). Therefore, the immune cell behaviors of chemotaxis, cytoskeletal modifications, and syncytia formation provide excellent nodes to investigate how the innate immune system functions generally, in a larva, and likely elements that are shared in an adult. ...
Article
Full-text available
Sea star wasting—marked in a variety of sea star species as varying degrees of skin lesions followed by disintegration— recently caused one of the largest marine die-offs ever recorded on the west coast of North America, killing billions of sea stars. Despite the important ramifications this mortality had for coastal benthic ecosystems, such as increased abundance of prey, little is known about the causes of the disease or the mechanisms of its progression. Although there have been studies indicating a range of causal mechanisms, including viruses and environmental effects, the broad spatial and depth range of affected populations leaves many questions remaining about either infectious or non-infectious mechanisms. Wasting appears to start with degradation of mutable connective tissue in the body wall, leading to disintegration of the epidermis. Here, we briefly review basic sea star biology in the context of sea star wasting and present our current knowledge and hypotheses related to the symptoms, the microbiome, the viruses, and the associated environmental stressors. We also highlight throughout the article knowledge gaps and the data needed to better understand sea star wasting mechanistically, its causes, and potential management.
... In vertebrates, MIF is a pleiotropic cytokine, involved in both tissue damage and repair upon noxious stimuli , promoting platelet survival, chemotaxis of immune cells, and release of other inflammatory mediators, such as tumor necrosis factor (TNF) and IL-1β, and growth factors. In invertebrates, like starfish (an echinoderm), MIF is at the center stage of phagocyte's chemotaxis (Furukawa et al. 2016). IL-14 has been described as an important growth factor for B cells in vertebrates. ...
Article
Full-text available
Innate immunity is present in all animals. In this review, we explore the main conserved mechanisms of recognition and innate immune responses among animals. In this sense, we discuss the receptors, critical for binding to pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs); the downstream signaling proteins; and transcription factors that govern immune responses. We also highlight conserved inflammatory mediators that are induced after the recognition of DAMPs and PAMPs. At last, we discuss the mechanisms that are involved in the regulation and/or generation of reactive oxygen species (ROS), influencing immune responses, like heme-oxygenases (HOs).
... Box 1.2 and Figure 1.6 in Chapter 1) that function through pathogen recognition receptors (PRRs) and associated cell signalling pathways, anti-microbial peptides and proteins (AMPs) in the coelomic fluid, and a melanisation pathway (Smith and Söderhall 1991;Smith et al. 2010;Franco et al. 2011;Smith et al. 2018). Many of the sea urchin PRRs are conserved among many animals (Bulgakov et al. 2013;Gowda et al. 2013) and include Toll-like receptors (TLRs), NOD-like receptors (NLRs), scavenger receptors that are cysteine rich (SRCRs), peptidoglycan recognition proteins (PGRPs), Gram-negative binding proteins (GNBPs), cytokine signalling molecules, plus proteins that are involved in the signalling pathways for PRRs (Pancer 2000;Hibino et al. 2006;Buckley and Rast 2012;Furukawa et al. 2016;Buckley et al. 2017). Anti-pathogen effector mechanisms include multiple AMPs, the diverse SpTransformer proteins, a wide variety of lectins, a complement pathway, a prophenoloxidase cascade, and other molecules (Smith et al. 2001;Hibino et al. 2006;Roch et al. 1992;Li et al. 2015;Smith et al. 2010;Buckley and Rast 2012;Dheilly et al. 2013;Smith and Lun 2017). ...
Chapter
Echinoderms are basal deuterostomes and the sister phylum to the chordates. All are marine animals with effective and robust innate immunity. Echinoderms are host to diseases in both larvae and adults, which include bald sea urchin disease, sea star wasting disease, among others that are defined by a range of different and often overlapping symptoms The diseases infect either individual animals or entire populations depending on the virulence and rate of spread and can lead to death in individuals or to mass die-offs. Pathogens include marine bacteria, typically Gammaproteobacteria and members of the Vibrio splendidus clade, as well as protists and viruses. Identify the initiating pathogen has been challenging but DNA sequencing approaches provide an avenue for pathogen identification, for differentiating between an initiating pathogen and organisms that cause the secondary infections, and for characterising changes to the normal microbiome of an infected echinoderm. Echinoderm pathologies have direct impacts on marine ecology and on aquaculture for edible species that, in some cases, can be alleviated by optimising the habitat conditions for healthy echinoderm populations.
... Box 1.2 and Figure 1.6 in Chapter 1) that function through pathogen recognition receptors (PRRs) and associated cell signalling pathways, anti-microbial peptides and proteins (AMPs) in the coelomic fluid, and a melanisation pathway (Smith and Söderhall 1991;Smith et al. 2010;Franco et al. 2011;Smith et al. 2018). Many of the sea urchin PRRs are conserved among many animals (Bulgakov et al. 2013;Gowda et al. 2013) and include Toll-like receptors (TLRs), NOD-like receptors (NLRs), scavenger receptors that are cysteine rich (SRCRs), peptidoglycan recognition proteins (PGRPs), Gram-negative binding proteins (GNBPs), cytokine signalling molecules, plus proteins that are involved in the signalling pathways for PRRs (Pancer 2000;Hibino et al. 2006;Buckley and Rast 2012;Furukawa et al. 2016;Buckley et al. 2017). Anti-pathogen effector mechanisms include multiple AMPs, the diverse SpTransformer proteins, a wide variety of lectins, a complement pathway, a prophenoloxidase cascade, and other molecules (Smith et al. 2001;Hibino et al. 2006;Roch et al. 1992;Li et al. 2015;Smith et al. 2010;Buckley and Rast 2012;Dheilly et al. 2013;Smith and Lun 2017). ...
... Aggregates have also been documented in echinoids (sea urchins) in response to foreign particles and serving coagulation purposes [13]. Despite not having been described as independent aggregates in asteroids, some studies showed the clotting of numerous cells at wound sites [14,15]. A similar involvement in wound healing was reported in ophiuroids (brittle stars) and crinoids (feather stars) [16,17], with no description of the presence of aggregates. ...
Article
The innate immunity of echinoderms has been a research focus since the early twentieth century, consistently providing ever deeper knowledge of its complexity and evolutionary aspects. At its core are coelomocytes, which are diverse cells collectively known to respond in a variety of ways, including via movement, phagocytosis, and aggregation. However, features of cellular immunity have never been compared in echinoderms from phylogenetic and distributional perspectives, to provide insight into ecological and evolutionary patterns. The present study catalyzed and characterized the formation of coelomocyte aggregates in members of all five extant classes of echinoderms. The morphological characteristics of these aggregates (including their colour, shape, texture, size) were assessed, as well as the major cells composing them. Coelomocyte diversity (both as free and aggregated forms) was determined to be maximum in class Holothuroidea, followed by Echinoidea, with the other classes showing similar levels of diversity. The colours of coelomocyte aggregates appeared to be more closely linked to phylogeny (classes, orders) rather than geographic range, or external colour of the species themselves. Asteroids and ophiuroids displayed primarily light-coloured aggregates, from transparent to green; while holothuroids, echinoids and crinoids demonstrated more vivid variants, from red to deep purple. The kinetics of aggregate formation and expulsion were monitored in selected species, showing immediate cellular response to foreign particulate matter in the form of encapsulation and various methods of expulsion, including through the dermal papillae of asteroids and the anus (cloaca) of holothuroids. The findings support that coelomocyte aggregate formation is a conserved immune response across all five extant classes of echinoderms with variations in their cell catalysts, complexity, shape, colour, and size.
... However, as in other invertebrates, the EsMIF sequence lacks key residues for certain enzyme activities and receptor binding that are present in vertebrate orthologs (SI Appendix, Fig. S1A), and the E. scolopes genome does not encode the well-described MIF receptors present in vertebrates (36). These data support the idea that MIF proteins have different functions and regulatory mechanisms in vertebrates and invertebrates (63)(64)(65). ...
Article
Full-text available
Significance Daily biological rhythms are fundamental, evolutionarily conserved features of many symbioses. Here we use the binary squid–vibrio association, the simplicity of which has offered the resolution to explore strategies of symbiosis that are shared with more complex systems, such as those of mammals. We demonstrate a pivotal role for an evolutionarily conserved cytokine, macrophage migration inhibitory factor, or MIF, which is abundant in epithelia supporting the symbionts of both the squid light organ and the mammalian gut. In the mature squid association, MIF regulates the cyclic metabolic dialogue that underlies oscillations in host-provided symbiont nutrients. As such, this study integrates the role of MIF across two distinct time scales: the host’s life history and its daily rhythms.
Article
Full-text available
The sea urchin, Strongylocentrotus purpuratus has seven described populations of distinct coelomocytes in the coelomic fluid that are defined by morphology, size, and for some types, by known functions. Of these subtypes, the large phagocytes are thought to be key to the sea urchin cellular innate immune response. The concentration of total coelomocytes in the coelomic fluid increases in response to pathogen challenge. However, there is no quantitative analysis of how the respective coelomocyte populations change over time in response to immune challenge. Accordingly, coelomocytes collected from immunoquiescent, healthy sea urchins were evaluated by flow cytometry for responses to injury and to challenge with either heat-killed Vibrio diazotrophicus, zymosan A, or artificial coelomic fluid, which served as the vehicle control. Responses to the initial injury of coelomic fluid collection or to injection of V. diazotrophicus show significant increases in the concentration of large phagocytes, small phagocytes, and red spherule cells after one day. Responses to zymosan A show decreases in the concentration of large phagocytes and increases in the concentration of small phagocytes. In contrast, responses to injections of vehicle result in decreased concentration of large phagocytes. When these changes in coelomocytes are evaluated based on proportions rather than concentration, the respective coelomocyte proportions are generally maintained in response to injection with V. diazotrophicus and vehicle. However, this is not observed in response to zymosan A and this lack of correspondence between proportions and concentrations may be an outcome of clearing these large particles by the large phagocytes. Variations in coelomocyte populations are also noted for individual sea urchins evaluated at different times for their responses to immune challenge compared to the vehicle. Together, these results demonstrate that the cell populations in sea urchin immune cell populations undergo dynamic changes in vivo in response to distinct immune stimuli and to injury and that these changes are driven by the responses of the large phagocyte populations.
Article
Full-text available
Echinoderms have a large coelomic cavity containing coelomocytes. When the coelomic fluid is removed from the cavity, the cells aggregate immediately. We found that a fraction or an extract of the intestine of the sea cucumber, Apostichopus japonicus, markedly accelerated cellular movement and aggregation on a glass slide, and this effect was clearly inhibited by galactose. We successfully purified the aggregation-promoting factor, a 16 kDa protein, from the intestine. TOF-MS analysis followed by de novo sequencing revealed that the protein is a C-type lectin. RNA-seq data and cDNA cloning demonstrated the factor to be a novel lectin, named AjGBCL, consisting of 158 aa residues in the mature form. Microscopic observation revealed that most of the aggregating cells moved toward aggregates and not to an intestinal fragment, suggesting that AjGBCL is not a chemoattractant but a cellular aggregation-inducing factor that may induce aggregates to release chemoattractant. We report, for the first time, an endogenous molecule that promotes coelomocyte aggregation in echinoderms.
Preprint
Full-text available
Renal interstitial fibrosis is the pathological basis of end-stage renal disease, in which the heterogeneity of macrophages in renal microenvironment plays an important role. However, the molecular mechanisms of macrophage plasticity during renal fibrosis progression remain unclear. In this study, we found for the first time that increased expression of Twist1 in macrophages was significantly associated with the severity of renal fibrosis in IgA nephropathy patients and mice with unilateral ureteral obstruction (UUO). Ablation of Twist1 in macrophages markedly alleviated renal tubular injury and renal fibrosis in UUO mice, accompanied by a lower extent of macrophage infiltration and M2 polarization in the kidney. The knockdown of Twist1 inhibited the chemotaxis and migration of macrophages, at least partially, through the CCL2/CCR2 axis. Twist1 downregulation inhibited M2 macrophage polarization and reduced the secretion of the profibrotic factors Arg1, MR (CD206), IL-10, and TGF-β. Galectin-3 was decreased in the macrophages of the conditional Twist1-deficient mice, and Twist1 was shown to directly activate galectin-3 transcription. Up-regulation of galectin-3 recovered Twist1-mediated M2 macrophage polarization. In conclusion, Twist1/galectin-3 signaling regulates macrophage plasticity (M2 phenotype) and promotes renal fibrosis. This study could suggest new strategies for delaying kidney fibrosis in patients with chronic kidney disease.
Article
Full-text available
We announce the release of an advanced version of the Molecular Evolutionary Genetics Analysis (MEGA) software, which currently contains facilities for building sequence alignments, inferring phylogenetic histories, and conducting molecular evolutionary analysis. In version 6.0, MEGA now enables the inference of timetrees, as it implements our RelTime method for estimating divergence times for all branching points in a phylogeny. A new Timetree Wizard in MEGA6 facilitates this timetree inference by providing a graphical user interface (GUI) to specify the phylogeny and calibration constraints step-by-step. This version also contains enhanced algorithms to search for the optimal trees under evolutionary criteria and implements a more advanced memory management that can double the size of sequence data sets to which MEGA can be applied. Both GUI and command-line versions of MEGA6 can be downloaded from www.megasoftware.net free of charge.
Article
Full-text available
Sepsis and septic shock are among the leading causes of death in intensive care units worldwide. Numerous studies on their pathophysiology have revealed an imbalance in the inflammatory network leading to tissue damage, organ failure, and ultimately, death. Cytokines are important pleiotropic regulators of the immune response, which have a crucial role in the complex pathophysiology underlying sepsis. They have both pro- and anti-inflammatory functions and are capable of coordinating effective defense mechanisms against invading pathogens. On the other hand, cytokines may dysregulate the immune response and promote tissue-damaging inflammation. In this review, we address the current knowledge of the actions of pro- and anti-inflammatory cytokines in sepsis pathophysiology as well as how these cytokines and other important immunomodulating agents may be therapeutically targeted to improve the clinical outcome of sepsis.
Article
Full-text available
Macrophage migration inhibitory factor (MIF) has been defined as an important chemokine-like function (CLF) chemokine with an essential role in monocyte recruitment and arrest. Adhesion of monocytes to the vessel wall and their transendothelial migration are critical in atherogenesis and many other inflammatory diseases. Chemokines carefully control all steps of the monocyte recruitment process. Those chemokines specialized in controlling arrest are typically immobilized on the endothelial surface, mediating the arrest of rolling monocytes by chemokine receptor-triggered pathways. The chemokine receptor CXCR2 functions as an important arrest receptor on monocytes. An arrest function has been revealed for the bona fide CXCR2 ligands CXCL1 and CXCL8, but genetic studies also suggested that additional arrest chemokines are likely to be involved in atherogenic leukocyte recruitment. While CXCR2 is known to interact with numerous CXC chemokine ligands, the CLF chemokine MIF, which structurally does not belong to the CXC chemokine sub-family, was surprisingly identified as a non-cognate ligand of CXCR2, responsible for critical arrest functions during the atherogenic process. MIF was originally identified as macrophage migration inhibitory factor (this function being eponymous), but is now known as a potent inflammatory cytokine with CLFs including chemotaxis and leukocyte arrest. This review will cover the mechanisms underlying these functions, including MIF’s effects on LFA1 integrin activity and signal transduction, and will discuss the structural similarities between MIF and the bona fide CXCR2 ligand CXCL8 while emphasizing the structural differences. As MIF also interacts with CXCR4, a chemokine receptor implicated in CXCL12-elicited lymphocyte arrest, the arrest potential of the MIF/CXCR4 axis will also be scrutinized as well as the recently identified role of pericyte MIF in attracting leukocytes exiting through venules as part of the pericyte “motility instruction program.”
Article
Full-text available
In larvae of the starfish, Asterina pectinifera, mesenchyme cells operate in the defense system through various behaviors. We have investigated mesenchyme cell dynamics during the immune response by identifying ApDOCK, a new member of the DOCK180 superfamily protein. In 4-day-old bipinnaria larvae processed for morpholino oligonucleotide-mediated knockdown of ApDOCK, injection of inorganic foreign substances revealed that (1) mesenchyme cells fail to undergo either directed migration toward a large oil-droplet or persistent spreading on the oil-droplet after contact; (2) neither uptake of micro-beads nor cell-to-cell fusion on the large oil-droplet differed from that of mesenchyme cells from control larvae. Similar behaviors were also recorded in experiments where bacteria were injected. Under culture conditions, the expression level of ApDOCK mRNA was significantly associated with the immunological behavior of mesenchyme cells. Apparently, the mesenchyme cells from ApDOCK loss-of-function larvae exhibited insufficient lamellipodium formation via lack of fibrous form of actin organization at the leading edge. These results suggest that the migratory congregation and persistence of encapsulation of larval mesenchyme cells are intracellularly regulated by ApDOCK protein, and this regulation is associated with organization of cytoskeletal actin.Immunology and Cell Biology advance online publication, 17 July 2012; doi:10.1038/icb.2012.37.
Article
Full-text available
Macrophage migration inhibitory factor (MIF) is a pivotal regulator of the immune response. Neutralization or genetic deletion of MIF does not completely abrogate activation responses, however, and deletion of the MIF receptor, CD74, produces a more pronounced phenotype than MIF deficiency. We hypothesized that these observations may be explained by a second MIF-like ligand, and we considered a probable candidate to be the protein encoded by the homologous, D-dopachrome tautomerase (D-DT) gene. We show that recombinant D-DT protein binds CD74 with high affinity, leading to activation of ERK1/2 MAP kinase and downstream proinflammatory pathways. Circulating D-DT levels correlate with disease severity in sepsis or malignancy, and the specific immunoneutralization of D-DT protects mice from lethal endotoxemia by reducing the expression of downstream effector cytokines. These data indicate that D-DT is a MIF-like cytokine with an overlapping spectrum of activities that are important for our understanding of MIF-dependent physiology and pathology.
Article
Full-text available
Proteins containing a scavenger receptor cysteine-rich (SRCR) domain (SRCR proteins) play an important role in the innate immune system of various metazoan animals. In the starfish Asterina pectinifera, mesenchyme cells and coelomocytes govern the two distinct innate immune systems of the larvae and adults, respectively. Here we identify a cDNA encoding a protein containing nine SRCR domains termed ApSRCR1, and present characterization of the molecular structure, expression, subcellular localization and function of ApSRCR1 protein during ontogenesis of this animal. ApSRCR1 protein is a membrane-type protein with a predicted molecular mass of approximately 120 kDa. During ontogenesis, ApSRCR1 protein is de novo synthesized and localizes to cytoplasmic vesicles in both mesenchyme cells and coelomocytes without translation of maternal mRNA; however, the net production and modification by N-glycosylation of ApSRCR1 protein differs in each cell type. In both types of cell, functional inhibition of ApSRCR1 protein leads to incompetent bacterial clearance and failure of aggregate formation. However, this inhibitory effect is weaker in the mesenchyme cells than in the coelomocytes. In the bacteria-sensitized adult, ApSRCR1 protein is up-regulated and digested to enable its secretion into the coelomic fluid. This secreted form of ApSRCR1 protein can apparently bind to bacteria. Overall, we show that ApSRCR1 protein is finely regulated for expression not only during development but also in a sensitive innate immunological situation, and thereupon acts as an opsonin for bacteria to different extents in the larvae and adults of A. pectinifera.
Article
Full-text available
Macrophage migration inhibitory factor (MIF) promotes leukocyte recruitment to sites of inflammation. However, whether this stems from a direct effect on leukocyte migration is unknown. Furthermore, the role of the MIF-binding protein CD74 in this response has not been investigated. Therefore, the aim of this study was to examine the contributions of MIF and CD74 to chemokine-induced macrophage recruitment. Intravital microscopy studies demonstrated that CCL2-induced leukocyte adhesion and transmigration were reduced in MIF(-/-) and CD74(-/-) mice. MIF(-/-) and CD74(-/-) macrophages also exhibited reduced chemotaxis in vitro, although CD74(-/-) macrophages showed increased chemokinesis. Reduced CCL2-induced migration was associated with attenuated MAPK phosphorylation, RhoA GTPase activity, and actin polymerization in MIF(-/-) and CD74(-/-) macrophages. Furthermore, in MIF(-/-) macrophages, MAPK phosphatase-1 was expressed at elevated levels, providing a potential mechanism for the reduction in MAPK phosphorylation in MIF-deficient cells. No increase in MAPK phosphatase-1 expression was observed in CD74(-/-) macrophages. In in vivo experiments assessing the link between MIF and CD74, combined administration of MIF and CCL2 increased leukocyte adhesion in both MIF(-/-) and CD74(-/-) mice, showing that CD74 was not required for this MIF-induced response. Additionally, although leukocyte recruitment induced by administration of MIF alone was reduced in CD74(-/-) mice, consistent with a role for CD74 in leukocyte recruitment induced by MIF, MIF-treated CD74(-/-) mice displayed residual leukocyte recruitment. These data demonstrate that MIF and CD74 play previously unappreciated roles in CCL2-induced macrophage adhesion and migration, and they indicate that MIF and CD74 mediate this effect via both common and independent mechanisms.
Article
MC5 molecule that exclusively expresses on the surface of embryonic mesenchyme cells (MCs) of starfish, Asterina pectinifera, newly comes within the category of metalloproteinase of astacin family. We studied the problem of how this novel membrane-type metalloproteinase functions during morphogenesis of the starfish. Inhibition experiments of MC5 molecule expression by use of Morpholino antisense oligonucleotides (MC5-MAO) reveals a phenotype as being "dwarfed effect"; those are, (1) Injection of the egg with MC5-MAO causes the larva to be significantly smaller, accompanied by a decrease in cell numbers and suppression of DNA-replication of epithelial cells (ECs) predominating at the body. (2) MC5-MAO injected larva displayed delayed development without inhibiting cell differentiation on nerve, muscle, and stomach cells once reaches the bipinnaria stage. These observations suggest that MC5 molecule positively works for ECs to proliferate. This hypothesis was proved by overexpression of MC5 mRNA and rescue-experiments co-injection with MC5 mRNA and MC5-MAO. Next, we examined the problem whether MCs involve in positive regulation for ECs proliferation. It is observed that although MC5-MAO induces "dwarfed effect" directly on the ECs, the phenotype becomes remarkable after ingression of the MCs into the blastocoel. Moreover, by injecting with MCs into the embryo showing "dwarfed effect", the embryo is shown to gain the DNA-replication meaningfully. Our results suggest that MCs are endowed with inducing activity for the ECs to proliferate during starfish morphogenesis.
Article
The stage at which phagocytosis can first be characterized in the embryos of the sea urchin Lytechinus variegatus was investigated by microinjecting the yeast Saccharomyces cerevisiae into its blastocoele. Secondary mesenchyme cells were first observed phagocytosing during the mid-gastrula stage. Subsequently, as the incubation time increased, the number of yeast per phagocyte rose. Using vital fluorescence dyes, stained free yeast were seen in the blastocoele during late-gastrula stage, indicating cell death and suggesting specific factors, such as proteases, in the extracellular environment. The starting point of phagocytic activity reflects a biological capacity for distinguishing between self and nonself. Thus, the phagocytosis of yeast by mesenchymal cells beginning in the mid-gastrula stage in L. variegatus may indicate the moment of acquisition of ‘identity’ (self) in this organism. Comparative aspects of embryo and adult phagocytes in L. variegatus are also discussed.
Article
Macrophage migration inhibitory factor (MIF) facilitates multiple aspects of inflammatory arthritis, the pathogenesis of which has been significantly linked to the activity of neutrophils. The effects of MIF on neutrophil recruitment are unknown. This study was undertaken to investigate the contribution of MIF to the regulation of neutrophil chemotactic responses. K/BxN serum-transfer arthritis was induced in wild-type (WT), MIF(-/-) , and monocyte chemotactic protein 1 (MCP-1; CCL2)-deficient mice as well as in WT mice treated with monoclonal antibodies to cytokine-induced neutrophil chemoattractant (anti-KC). Leukocyte trafficking in vivo was examined using intravital microscopy, and neutrophil function in vitro was examined using migration chambers and assessment of MAP kinase activation. K/BxN serum-transfer arthritis was markedly attenuated in MIF(-/-) mice, with reductions in the clinical and histologic severity of arthritis and the synovial expression of KC and interleukin-1. Arthritis was also reduced by anti-KC antibody treatment, but not in MCP-1-deficient mice. In vivo, neutrophil recruitment responses to KC were reduced in MIF(-/-) mice. Similarly, MIF(-/-) mouse neutrophils exhibited reduced chemotactic responses to KC in vitro, despite displaying unaltered chemokine receptor expression. Reduced chemotactic responses of MIF(-/-) mouse neutrophils were associated with reduced phosphorylation of p38 and ERK MAP kinases. These findings suggest that MIF promotes neutrophil trafficking in inflammatory arthritis via facilitation of chemokine-induced migratory responses and MAP kinase activation. Therapeutic MIF inhibition could limit synovial neutrophil recruitment.