ArticlePDF Available

Distinct Differentiation Programs Triggered by IL-6 and LPS in Teleost IgM+ B Cells in the Absence of Germinal Centers

Authors:

Abstract and Figures

Although originally identified as a B cell differentiation factor, it is now known that mammalian interleukin-6 (IL-6) only regulates B cells committed to plasma cells in response to T-dependent (TD) antigens within germinal centers (GCs). Even though adaptive immunity is present in teleost fish, these species lack lymph nodes and GCs. Thus, the aim of the present study was to establish the role of trout IL-6 on B cells, comparing its effects to those induced by bacterial lipopolysaccharide (LPS). We demonstrate that the effects of teleost IL-6 on naïve spleen B cells include proliferation, activation of NF-κB, increased IgM secretion, up-regulation of Blimp1 transcription and decreased MHC-II surface expression that point to trout IL-6 as a differentiation factor for IgM antibody-secreting cells (ASCs). However, LPS induced the secretion of IgM without up-regulating Blimp1, driving the cells towards an intermediate activation state in which antigen presenting mechanisms are elicited together with antibody secretion and expression of pro-inflammatory genes. Our results reveal that, in trout, IL-6 is a differentiation factor for B cells, stimulating IgM responses in the absence of follicular structures, and suggest that it was after follicular structures appeared that this cytokine evolved to modulate TD responses within the GC.
IL-6 and LPS activate IgM secretion in naïve B cells.: (a) ELISPOT analysis of IgM-secreting cells in splenocyte cultures treated with IL-6 (200 ng/ml), LPS (100 μg/ml) or non-stimulated. Splenocytes were cultured for 3 days in ELISPOT plates previously coated with anti-trout IgM mAb (2 μg/ml) in the presence or absence of the different stimuli. After incubation, cells were washed away and a biotinylated anti-trout IgM mAb (1 μg/ml) was used to detect numbers of spot forming cells. Duplicates from a representative experiment (left) and quantification of spot forming cells (right) from 5 independent experiments are shown (mean + standard deviation). (b) Spleen leukocytes were incubated with media containing IL-6, LPS or control media alone for 24 h at 20 °C. After that time, IgM+ B cells were sorted using an anti-trout IgM mAb and RNA was extracted. Relative transcript expression of Blimp-1 and ACKR2 is shown (mean + standard deviation, n = 6). (c) Membrane IgM and total IgM expression of IgM+ cells after incubation with IL-6, LPS or control media for 24, 48 and 72 h. Mean fluorescence intensity (MFI) + standard deviation is shown (n = 9). (d) Dot plots and histograms showing the Forward scatter (FSC) from IgM+ B cells and BrdU+/IgM+ B cells incubated in the presence or absence of LPS or IL-6, from one representative experiment. Graphs showing FSC MFI values from 8 independent experiments (mean + standard deviation) are included next to the histograms for stimulated cultures. *P < 0.05, **P < 0.01, ***P < 0.001.
… 
Content may be subject to copyright.
1
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
www.nature.com/scientificreports
Distinct Dierentiation Programs
Triggered by IL-6 and LPS in Teleost
IgM+ B Cells in The Absence of
Germinal Centers
Beatriz Abós1, Tiehui Wang2, Rosario Castro1, Aitor G. Granja1, Esther Leal1,
Jerey Havixbeck3, Alfonso Luque1, Daniel R. Barreda3, Chris J. Secombes2 & Carolina Tafalla1
Although originally identied as a B cell dierentiation factor, it is now known that mammalian
interleukin-6 (IL-6) only regulates B cells committed to plasma cells in response to T-dependent (TD)
antigens within germinal centers (GCs). Even though adaptive immunity is present in teleost sh,
these species lack lymph nodes and GCs. Thus, the aim of the present study was to establish the role of
trout IL-6 on B cells, comparing its eects to those induced by bacterial lipopolysaccharide (LPS). We
demonstrate that the eects of teleost IL-6 on naïve spleen B cells include proliferation, activation of
NF-κB, increased IgM secretion, up-regulation of Blimp1 transcription and decreased MHC-II surface
expression that point to trout IL-6 as a dierentiation factor for IgM antibody-secreting cells (ASCs).
However, LPS induced the secretion of IgM without up-regulating Blimp1, driving the cells towards
an intermediate activation state in which antigen presenting mechanisms are elicited together with
antibody secretion and expression of pro-inammatory genes. Our results reveal that, in trout, IL-6
is a dierentiation factor for B cells, stimulating IgM responses in the absence of follicular structures,
and suggest that it was after follicular structures appeared that this cytokine evolved to modulate TD
responses within the GC.
e immune system comprises both innate and adaptive immune responses. While the innate immune system
is genetically programmed to detect invariant features of invading microbes, the cells of the adaptive immune
system, such as conventional B cells (B2) and T cells, detect specic epitopes through somatically recombined
receptors. However, it is now recognized that both branches of immunity are highly interconnected and B cells
also possess a certain capacity to directly sense and respond to pathogens though the expression of certain pattern
recognition receptors (PRRs) or through the action of cytokines produced by cells of the innate immune system1.
In general, conventional B cells are activated in response to T-dependent (TD) antigens within the lymphoid
follicles and trigger the formation of germinal centers (GCs). ese sites promote the close collaboration between
proliferating antigen-specic B cells, T follicular helper cells, and the specialized follicular dendritic cells (DCs)
that constitutively occupy the central follicular zones of secondary lymphoid organs. In this environment,
B cells divide in response to antigens and acquire the capacity to dierentiate into antibody-secreting cells (ASCs),
reaching a terminal state of plasma cells or memory B cells, both of them with the capacity to secrete high anity
antibodies. is TD pathway provides a strong long-lived immunological memory, but is relative slow to occur.
us, it must be integrated with additional T-independent (TI) pathways that mainly involve other B cell subsets
such as B1 cells or marginal zone (MZ) B cells. ese TI responses do not require cooperation from T cells, but
instead are much more responsive to products secreted by cells of the innate immune system and have a greater
capacity to directly recognize pathogens1.
Although evolutionarily jawed sh constitute the rst group of animals in which adaptive immunity based on
Ig receptors is present2, many structural immune peculiarities predict important functional dierences between
sh and mammalian B cells. e teleost spleen constitutes the main secondary immune organ in the absence of
lymph nodes. However, the splenic white pulp is poorly developed in teleosts in comparison to mammals and
1Centro de Investigación en Sanidad Animal (CISA-INIA), Madrid, Spain. 2Scottish Fish Immunology Research Centre,
University of Aberdeen, Aberdeen, UK. 3Department of Biological Sciences, University of Alberta, Alberta, Canada.
Correspondence and requests for materials should be addressed to C.T. (email: tafalla@inia.es)
Received: 27 April 2016
Accepted: 28 June 2016
Published: 02 August 2016
OPEN
www.nature.com/scientificreports/
2
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
no GCs are apparent3. Regarding mucosal immunity, although sh B cells have been reported in surfaces such as
gills, skin, digestive tract and nasal cavities4,5, they are scattered throughout the mucosa in disorganized lymphoid
structures6. Additionally, sh contain only three immunoglobulin classes IgM, IgD and IgT (designated as IgZ
in some species). IgT is a teleost sh-specic Ig that seems specialized in mucosal immunity7,8 and IgT+ B cells
constitute a distinct linage7, thus no class switch recombination has ever been reported in sh. As a result, the
lack of teleost follicular structures already anticipates that sh B cell responses best resemble mammalian extr-
afollicular responses. Consequently, teleost B cells share many features of mammalian B1 cells, as for example a
high phagocytic capacity9,10, constitutive expression of many PRRs4,11 or expression of B1-specic cell markers12.
Interleukin 6 (IL-6) is a multi-functional cytokine produced by a wide range of cell types in the early stages
of infection. IL-6 modulates a plethora of immune functions through a receptor composed of the restricted IL-6
receptor chain (IL-6R) and a common signal transducer, gp13013. Although initially described as a B cell dif-
ferentiation factor14, it was later demonstrated that IL-6 is a potent growth and maturation factor only for cells
that have already initiated a dierentiation process towards plasma cells, but has minimal capacity to directly
induce plasma cell dierentiation15. Besides, IL-6 enhances antibody production of ASCs but only those that are
antigen-specic, whereas non-specic ASCs are unresponsive to IL-616. Interestingly, the normal development of
GCs is signicantly altered in the absence of IL-617. Consequently, IL-6 deciency signicantly impairs early TD
IgG production, but has no eect on IgM TI responses17,18. us, it has been postulated that IL-6 is predominantly
involved in the maturation of TD plasma cells in the early stages of GC formation19. At a mucosal level, IL-6 also
seems implicated in promoting IgA TD responses20. Strikingly, the number of B1-derived IgA-secreting cells
signicantly increases in the absence of IL-620, demonstrating that B1 cells are not regulated by this cytokine and
can even be negatively aected by it. Despite this, IL-6 in combination with an anti-IgM antibody induces the
expression of surface CD5 on conventional B2 cells that acquire phenotypic characteristics of B1 cells21.
Taking into account the main role of IL-6 on TD responses in mammals and the unique attributes of sh
B cells, it is important to determine the eect of IL-6 on sh B cells, which we do here using the rainbow trout
(Oncorhynchus mykiss) as a model. We compare the eects elicited by IL-6 to those triggered by lipopolysaccha-
ride (LPS). LPS, a protein-free endotoxin from the cell wall of Gram-negative bacteria, is the most extensively
studied TI antigen in mammals and is directly recognized by B cells through low anity B cell receptors (BCRs)
and PRRs such as Toll like receptors (TLRs)22. LPS is not a mitogen for human B cells, since human B cells express
neither TLR4 nor CD14, the two canonical ligands for Gram ( ) bacterial LPS23,24. In contrast, mice B cells
express TLR4 and are polyclonally activated by it25. Interestingly, although TLR4 is thought to be absent from the
genome of salmonid sh26, some eects of LPS on salmonid B cells have been reported27.
Our ndings demonstrate that, in contrast to what occurs in mammals, B cells obtained from healthy unstim-
ulated trout respond to IL-6. e eects that IL-6 exerted on these IgM+ B cells included proliferation, activa-
tion of NF-κ B, increased IgM secretion, up-regulation of Blimp1 transcription, increased size and decreased
MHC-II surface expression, all pointing to trout IL-6 as a dierentiation factor for naïve IgM B cells towards
ASCs. Interestingly, LPS also increased IgM secretion but the activation prole was very dierent to that elicited
by IL-6, driving the IgM+ B cells towards a pro-inammatory state. Additionally, IL-6 stimulated B cells mobilized
more intracellular calcium in response to BCR cross-linking, demonstrating that IgM ASCs retain a functional
BCR on the cell surface. us, our results demonstrate that in trout, where follicular structures have not yet been
developed, IL-6 regulates IgM B cell responses. is suggests that the emergence of follicular structures marked a
critical point in IL-6 evolution, acquiring a novel capacity to specically regulate TD antibody responses.
Results
The IL-6 receptor complex is transcribed in IgM+ B cells from naïve sh that are activated by
IL-6 stimulation. IL-6 exerts its biological activities through two molecules: the IL-6 receptor, IL-6Rα (also
known as gp80 and CD130) and the associated signal transducer glycoprotein gp130. While IL-6Rα is important
for ligand binding, gp130 is required for adequate intracellular signaling28. Prior to studying the eects of IL-6
on naïve B cells in sh, we determined if these two molecules were constitutively transcribed in sorted naïve
IgM+ B cells from spleen, blood and kidney, being this last organ the main hematopoietic tissue in sh. We found
that IgM+ B cells from all three tissues constitutively transcribed IL-6Rα and gp130 at similar expression levels
(Supplemental Figure S1A), suggesting that naïve B cells have the capacity to respond to IL-6. ese results con-
trast those of mammals, where only activated B cells express IL-6R16,28.
To conrm that unstimulated B cells from trout are responsive to IL-6, we rst examined the expression of
SOCS3 and STAT3, two genes involved in the signaling of IL-6, in sorted IL-6-stimulated trout IgM+ B cells. Both
SOCS3 and STAT3 were signicantly up-regulated in IgM+ B cells in response to IL-6 stimulation (Supplemental
Figure S1B), as previously reported in other IL-6-responsive cell types in trout29. LPS, on the other hand, induced
the transcription of STAT3 but had no eect on SOCS3 mRNA levels (Supplemental Figure S1B). To further
conrm these results and demonstrate a direct eect of IL-6 on IgM+ B cells, we sorted IgM+ cells and aerwards
incubated them with IL-6 or LPS. Aer 1 h of incubation, the phosphorylation of STAT3 was conrmed through
Western blot in response to IL-6 and LPS (Supplemental Figure S1C). ese results conrm a direct eect of IL-6
on IgM+ B cells.
IL-6 induces IgM+ cell proliferation. As already mentioned, mammalian IL-6 has no eect on naïve B
cells, but has been shown to induce the proliferation of pre-B cells30 as well as plasmablasts31. In contrast, LPS is
a potent inducer of B cell proliferation in mice and sh27,32. us, we compared the lymphoproliferative eects
of LPS to those of IL-6 in trout splenocytes. Our results show that trout spleen IgM+ B cells signicantly prolif-
erated in response to IL-6 (Fig.1a,c), although at levels signicantly lower than those elicited by LPS (Fig.1a,c).
When the percentage of IgM+ B cells was evaluated in these cultures without taking into account BrdU uptake,
the percentage of IgM+ cells was quite similar in LPS- and IL-6-treated cultures, suggesting an additional positive
www.nature.com/scientificreports/
3
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
eect of IL-6 on B cell survival (Fig.1b). e proliferative and survival eects observed were specic to IL-6 since
another cytokine produced and tested in parallel, IL-4/13A, was unable to increase the number of trout IgM+ cells
during the in vitro culture33.
Since in mammals IL-6 only activates B cells previously stimulated with TD antigens17,18, we also evaluated
the potential synergistic eect of IL-6 on IgM+ B cell proliferation induced by a TD antigen (TNP-KLH) or a
TI antigen (TNP-LPS). We performed this experiment because despite their lack of follicular structures, sh
can orchestrate responses to TD antigens34, possibly through a mechanism that resembles rare extrafollicular
TD responses reported in mammals35–37. For this, we incubated splenocytes with TNP-KLH or TNP-LPS in the
presence or absence of IL-6 and evaluated the percentage of BrdU+/IgM+ B cells aer 4 days. We observed that
the addition of IL-6 could further enhance the proliferative eects provoked by TNP-LPS or TNP-KLH (Fig.1d),
indicating that, in trout, IL-6 can have regulatory eects on both TD and TI responses.
Figure 1. IL-6 and LPS elicit B cell proliferation and increased survival. Spleen leukocytes were incubated
with IL-6 (200 ng/ml) or LPS (100 μ g/ml) for 3 days at 20 °C. Aer this time, BrdU was added to the cultures and
incubated for a further 24 h. e percentage of proliferating (BrdU+) IgM+ B cells was determined as described in
Material and Methods. (a) Percentage (mean + standard deviation) of proliferating IgM+ B cells (BrdU+/IgM+)
aer treatment with IL-6 and/or LPS (n = 6). (b) IgM+ B cell survival estimated as percentage of IgM+ cells
(proliferating and non-proliferating cells) in cultures (mean + standard deviation) (n = 6). (c) Representative
dot plots are shown. (d) Spleen leukocytes were stimulated with TNP-KLH (5 μ g/ml) or TNP-LPS (5 μ g/ml) in
the presence or absence of IL-6 (200 ng/ml). e percentage of proliferating IgM+ cells was assessed as described
above. Data are shown as the mean fold change relative to the control value for unstimulated controls + standard
deviation (n = 5). Asterisks denote signicant dierences between cells treated with IL-6 or LPS and their
corresponding controls and between IL-6 and LPS treated cells when indicated. * P < 0.05, * * P < 0.01.
www.nature.com/scientificreports/
4
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
IL-6 up-regulates IgM secretion in naïve B cells. In mammals, IL-6 is unable to induce dierentiation
of naïve B cells towards plasmablasts on its own15. In contrast, the in vitro incubation of naïve splenic B cells with
IL-6 was sucient to induce the secretion of IgM, as veried in an ELISPOT assay (Fig.2a,b). When this experi-
ment was carried out in previously immunized sh, IL-6 also increased the secretion of IgM, at levels signicantly
higher than those observed in non-immunized healthy trout (Supplemental Figure S2). As previously reported27,
LPS also induced the secretion of IgM by trout B cells, however, our results suggest that the mechanisms through
which IgM secretion is induced dier between IL-6 and LPS-stimulated B cells. At the transcript level, only IL-6
up-regulated the transcription of Blimp1, a protein required for the development of ASCs and the maintenance of
long-lived plasma cells38, while LPS had no eect (Fig.2b). Interestingly, IL-6 also up-regulated the transcription
of ACKR2, an atypical chemokine receptor specic for innate B cells39, while LPS showed no eect (Fig.2b). us,
Figure 2. IL-6 and LPS activate IgM secretion in naïve B cells. (a) ELISPOT analysis of IgM-secreting cells
in splenocyte cultures treated with IL-6 (200 ng/ml), LPS (100 μ g/ml) or non-stimulated. Splenocytes were
cultured for 3 days in ELISPOT plates previously coated with anti-trout IgM mAb (2 μ g/ml) in the presence or
absence of the dierent stimuli. Aer incubation, cells were washed away and a biotinylated anti-trout IgM mAb
(1 μ g/ml) was used to detect numbers of spot forming cells. Duplicates from a representative experiment (le)
and quantication of spot forming cells (right) from 5 independent experiments are shown (mean + standard
deviation). (b) Spleen leukocytes were incubated with media containing IL-6, LPS or control media alone for 24 h
at 20 °C. Aer that time, IgM+ B cells were sorted using an anti-trout IgM mAb and RNA was extracted. Relative
transcript expression of Blimp-1 and ACKR2 is shown (mean + standard deviation, n = 6). (c) Membrane IgM
and total IgM expression of IgM+ cells aer incubation with IL-6, LPS or control media for 24, 48 and 72 h. Mean
uorescence intensity (MFI) + standard deviation is shown (n = 9). (d) Dot plots and histograms showing the
Forward scatter (FSC) from IgM+ B cells and BrdU+/IgM+ B cells incubated in the presence or absence of LPS
or IL-6, from one representative experiment. Graphs showing FSC MFI values from 8 independent experiments
(mean + standard deviation) are included next to the histograms for stimulated cultures. * P < 0.05, * * P < 0.01,
* * * P < 0.001.
www.nature.com/scientificreports/
5
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
our results demonstrate that, unlike mammalian IL-6, sh IL-6 is a dierentiation factor for naïve B cells towards
an ASC prole. On the other hand, LPS seems to induce the secretion of IgM in a Blimp1-independent fashion.
In mammals, the dierentiation of B cells to plasma cells provokes a down-regulation of membrane Igs that
completely loose the BCR when terminally dierentiated40. However, recent reports have demonstrated that this
seems to only be true for IgG plasma cells, since fully dierentiated IgM or IgA-secreting plasma cells retain a
functional BCR in the cell membrane41. In our studies, we observed that both IL-6 and LPS increased the levels of
expression of total IgM in B cells (Fig.2c), along with increased membrane IgM levels (Fig.2c). erefore, these
IL-6 or LPS-induced plasmablasts retain and even up-regulate IgM levels in the cell surface while increasing their
secretion of IgM, similar to mammalian IgM plasma cells. e secretion of IgM in response to LPS or IL-6 was
associated with an increase in the size of IgM+ cells, as shown by increased forward side scatter (FSC) in stimu-
lated cells in comparison to control cells. is increase in size was signicant when proliferating cells were ana-
lyzed (Fig.2d) demonstrating that LPS and IL-6 induce the proliferation of IgM+ B cells and part of the progeny
dierentiate into ASCs that retain IgM on the cell surface.
IL-6 activates NF-κB in spleen IgM+ B cells. Aer binding to its receptor, IL-6 activates gene expression
mainly through the STAT3 pathway28,42 and, in line with its pro-inammatory nature, it has been shown to acti-
vate NF-κ B in intestinal epithelial cells43. Despite this, whether IL-6 activates NF-κ B in mammalian B cells has
not been established. In mice, LPS induces a dual BCR/TLR-signaling by engaging TLR4 through the lipid A moi-
ety and the BCR through the polysaccharidic moiety44. As a result, both the canonical and non-canonical NF-κ B
signaling pathways are activated in B cells in response to LPS. To determine whether IL-6 and LPS activate the
NF-κ B signaling pathway in our system, we analyzed the translocation of the NF-κ B p65 subunit to the nucleus
of IgM+ B cells using a quantitative imaging ow cytometry-based approach which we have used previously11. We
observed a signicant increase in the percentage of total splenocytes and IgM+ B cells in which p65 translocated
to the nucleus in response to IL-6 and LPS in comparison with control cells, indicating that both stimuli activate
the canonical NF-κ B pathway (Fig.3a).
However, when the transcriptional activity of NF-κ B-dependent genes such as IL-1β 1, IL-8 or TNF-α 3 was
studied, only LPS was capable of inducing a signicant up-regulation of their mRNA levels (Fig.3b). Similarly,
dierential eects of LPS and IL-6 in the transcriptional activation of antimicrobial peptides was also observed.
While LPS up-regulated cathelicidin 1 (CATH1) mRNA levels (Fig.3b) CATH2 was signicantly up-regulated
by both LPS and IL-6, although the eect of IL-6 was much stronger (Fig.3b). Taken together, these results show
that although IL-6 and LPS activate NF-κ B in IgM+ B cells, the downstream eects of this activation are dierent.
IL-6 and LPS-stimulated IgM+ B cells are predisposed for BCR-mediated calcium mobili-
zation. Anti-IgM stimulation of B cells mimics the recognition of a high affinity antigen by the BCR,
Figure 3. Eect of IL-6 and LPS on NF-κB activation and transcription of pro-inammatory and
antimicrobial genes in B cells. (a) Translocation of NF-κ B to the nucleus in IgM+ splenocytes following
stimulation with LPS (100 μ g/ml) or IL-6 (200 ng/ml) for 24 h. e mean percentages of NF-κ B translocation in
total cells (upper panel) and IgM+ B cells (lower panel) from six independent experiments are shown. * * P < 0.01,
* * * P < 0.001. (b) Spleen leukocytes were incubated with IL-6, LPS or control media alone for 24 h at 20 °C. e
eect of IL-6 and LPS on the transcription of IL-1β 1, IL-8, TNF-α 3, cathelicidin-1 (CATH1), CATH2 and hepcidin
was then studied in sorted IgM+ B cells from these cultures. e relative transcript expression (mean + standard
deviation) of 6 independent experiments is shown. * P < 0.05, * * P < 0.01, * * * P < 0.001.
www.nature.com/scientificreports/
6
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
consequently leading to a rapid increase of intracellular calcium45. is calcium mobilization plays an important
role in B cell activation and is required for correct downstream signaling of the BCR45. us, we also studied the
eect of anti-IgM cross-linking of the BCR in splenocyte cultures that had been previously stimulated with IL-6
or LPS and compared them to those in non-stimulated control cells, given the fact that IL-6 or LPS-stimulated
cells seem to retain their BCR. As expected, anti-IgM induced a rapid mobilization of intracellular calcium, and
furthermore, the levels of calcium released upon anti-IgM stimulation were signicantly higher in cells that had
been previously stimulated with IL-6 or LPS (Fig.4a,b). is stimulatory eect was observed aer 24, 48 or 72 h of
stimulation. us, our results reveal that IL-6 renders the cells more responsive to BCR engagement.
IL-6 down-regulates the surface expression of MHC-II in naïve trout B cells. e response of
B cells against an antigen not only requires antigen binding to and signaling through the BCR but also the
processing and presentation of the BCR-bound antigen to helper T cells in the context of MHC-II46. us, we
also determined the eect of IL-6 or LPS stimulation on the levels of expression of surface MHC-II of naïve
B cells using a specic anti-trout MHC-II antibody. We observed that IL-6 provoked a signicant decrease of
surface MHC-II expression that was detectable from 24 h post-stimulation up to 72 h post-stimulation (Fig.5a).
In contrast, LPS-stimulated B cells showed increased surface MHC-II levels at all time points studied (Fig.5a).
Along with this increase in MHC II levels, LPS also up-regulated the levels of transcription of two co-stimulatory
molecules: CD80/86, a molecule with similar homologies to mammalian CD80 and CD8647, and CD83 (Fig.5b).
IL-6, on the other hand, had no eect on their transcription levels (Fig.5b).
IL-6 has no eect on the phagocytic activity of IgM+ B cells. Since trout B cells have a potent phago-
cytic activity9, we also investigated whether IL-6 or LPS pre-stimulation of naïve IgM+ B cells could have an
eect on their phagocytic activity. For this, splenocytes were incubated with IL-6 or LPS for dierent incubation
periods, or le-unstimulated in the same conditions. ereaer, 1 μ m polystyrene-based uorescent beads were
added to the cultures and aer 3 h of incubation the phagocytic activity of IgM+ cells was determined in ow
cytometry. We observed that while pre-stimulation of B cells with LPS signicantly increased the percentage of
phagocytic IgM+ B cells in spleen, pre-stimulation with IL-6 produced no eect (Fig.6a,b).
IL-6 has a synergistic eect on antigen-specic IgM responses in vivo. Given the stimulatory
eects observed for IL-6 on IgM+ B cells in vitro, we next investigated whether this stimulatory eect on naïve
IgM+ B cells could also condition IgM responses elicited in vivo in response to a specic antigen. For this, we
immunized sh with inactivated infectious pancreatic necrosis virus (IPNV) either alone or in combination with
IL-6. Control groups treated with IL-6 alone or PBS were also included. e administration of IL-6 alone sig-
nicantly increased the number of IgM-secreting cells in the spleen at day 2, but no eect was observed in the
following days (Fig.7a). IPNV on its own was unable to increase the number of IgM-secreting cells in spleen or
kidney when compared to controls, however when IPNV was combined with IL-6, the number of IgM-secreting
cells signicantly increased in spleen at day 6 post-immunization and in the kidney at day 2 post-immunization,
when compared to controls or sh injected with IPNV alone (Fig.7a). ese positive eects of IL-6 on the num-
ber of ASCs elicited by IPNV, correlated with signicantly increased IPNV-specic IgM titers in sera of sh
injected with IPNV and IL-6 when compared to titers in sh injected with IPNV alone (Fig.7b). Surprisingly,
this increase in IPNV-specic IgM had no eect on total IgM titers (Fig.7b) at the time point sampled (day 15
post-immunization).
To get additional information on the antibodies present in the sera we also analyzed the titers of natural anti-
bodies against four classic antigens, BSA, galactosidase, phosphorylcholine and dsRNA (poly I:C) in the same
Figure 4. IL-6 and LPS predispose IgM+ B cells to calcium mobilization aer BCR cross-liking. Spleen
leukocytes were incubated with IL-6 (200 ng/ml), LPS (100 μ g/ml) or control media alone for 24, 48 and 72 h
at 20 °C. Aer the dierent incubation periods, cells were loaded with Fluo-3 AM (5 μ M nal concentration),
and its baseline emission was measured by ow cytometry for 30 s, and then stimulated with 0.5 ug/ml
of Alexa 647 conjugated anti-IgM mAb. Fluorescence was then measured for a further 180 s. (a) Mean
uorescence intensity (MFI) plus standard deviation of intracellular Ca2+ levels (Fluo-3) in IgM+ B cells is
shown (n = 6). * P < 0.05, * * P < 0.01, * * * P < 0.001. (b) Scatter plot showing Fluo-3 MFI levels in spleen
IgM+ B cells aer 48 h of incubation with the appropriate stimuli.
www.nature.com/scientificreports/
7
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
groups. We observed a signicant decrease in the titers of antibodies that bind these four antigens in all the groups
treated with IL-6 or/and IPNV. Interestingly, at the same time that IL-6 signicantly increased IPNV-specic IgM
titers when compared to those elicited by IPNV alone (Fig.7b), it decreased the amount of natural antibodies
reactive against BSA or galactosidase in serum (Fig.7b). Taken together, our results strongly suggest that when
combined with an antigen in vivo, IL-6 preferentially activates antigen-specic B cells as occurs in mammals.
Discussion
Although mammalian IL-6 exclusively signals in antigen-experienced cells15, thus only aecting the outcome
of switched antibody isotypes such as IgG or IgA18,19,48; a recent study reported that fugu (Takifugu rubripes)
unstimulated B cells transcribe both IL-6Rα and gp130 and that the in vitro stimulation of blood leukocytes with
IL-6 up-regulates IgM transcription levels42. ese results suggest that sh B cells from healthy unstimulated sh
could be responsive to IL-6. Hence, we decided to investigate further the capacity of IL-6 to regulate B cell activity
in sh, using the rainbow trout as a model. Once we had veried that rainbow trout unstimulated IgM+ B cells
also transcribe IL-6Rα and gp130, we examined the eects of IL-6 on dierent relevant functions of sh B cells.
We compared the eects provoked by IL-6 to those elicited by LPS, since there is still some controversy about the
degree to which trout cells respond to LPS. Although LPS stimulation was reported to increase IgM secretion and
lymphocyte proliferation in trout27, TLR4 seems absent from salmonid genomes26. In this context, our studies
would help to establish how, in the absence of follicular structures, sh IL-6 regulates the activity of IgM+ B cells.
Furthermore, our results would provide additional evidence as to how teleost B cells respond to LPS.
LPS is a potent mitogen of B cells and the in vitro responses of these cells to LPS are very robust both in mice32
and in sh49. In our experiments, LPS and IL-6 were both mitogenic for trout IgM+ cells. Although the prolifera-
tion levels in response to IL-6 were signicantly lower than those observed for LPS, the percentage of viable IgM+
B cells in the cultures was similar in response to the dierent stimuli and signicantly higher than the percentage
observed in control cultures, suggesting that non-proliferating B cells have an increased survival in IL-6-treated
Figure 5. Dierential eects of IL-6 and LPS on antigen presenting properties of IgM+ B cells. Spleen
leukocytes were incubated with IL-6 (200 ng/ml), LPS (100 μ g/ml) or control media alone for 24, 48 and 72 h at
20 °C. e levels of MHC-II expression on the surface of IgM+ B cells were then measured via ow cytometry
using a specic mAb against trout MHC-II. (a) MFI + standard deviation from 6 independent experiments.
* P < 0.05, * * P < 0.01, * * * P < 0.001. Representative histogram from one experiment shown below. (b) Spleen
leukocytes were incubated with IL-6, LPS or control media alone for 24 h at 20 °C. e eect of IL-6 and LPS on
the transcription of CD80/86 and CD83 co-stimulatory molecules was then studied in sorted IgM+ cells from
these cultures. e relative transcript expression (mean + standard deviation) of 5 independent experiments is
shown. * P < 0.05, * * P < 0.01.
www.nature.com/scientificreports/
8
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
cultures, even higher than that observed in LPS-treated cultures. In mammals, while some authors have reported
a proliferative eect of IL-6 on pre-activated B cells50, other researchers have described stimulation of Ig secretion
in the absence of proliferative responses51,52. However, even in those cases where proliferating eects have been
reported, they seem to be associated only with previously activated cells and are never seen in unstimulated B
cells. Although it could be possible that some of the B cells in our cultures have been previously exposed to an
antigen, it seems unlikely that all the B cells responding in our experiments are pre-activated cells given the fact
that these cells express IgD on the cell membrane and transcribe surface IgM and IgD at high levels (data not
shown). On the other hand, even though we stimulated B cells in cultures in which other cells were present dur-
ing the incubation period, we have demonstrated that STAT3 is phosphorylated in IgM+ B cells incubated with
IL-6 in the absence of other cell types. Likewise, it should be noted that there was no signicant proliferation of
IgM cells in these cultures (Fig.1c) and that even if IgM+ B cells already accounted for approximately 30% of the
leukocyte population in the spleen, this percentage was increased to approximately 50% in IL-6-treated cultures
aer 4 days. All the evidence points to a preferential eect of IL-6 on a quite abundant population of B cells within
the spleen of unstimulated sh. In addition to these experiments in which leukocytes were treated with IL-6
alone, we also determined the eect of IL-6 on the B cell proliferation induced by antigen encounter, and again
observed stimulatory eects. ese synergistic eects were visualized in response to either TD (TNP-KLH) or TI
(TNP-LPS) antigens, unlike the situation in mammals where IL-6 only seems to promote TD responses17,18. us,
trout IL-6 has positive eects on proliferation and survival of B cells stimulated with TD antigens, and in contrast
to mammalian IL-6, it also has mitogenic eects on trout naïve B cells and cells activated with TI antigens.
IL-6 was originally reported as a cytokine capable of inducing antibody production in B cell lines, augmenting
the secretion of both IgM and IgG on Epstein-Barr virus transformed B cells51,52. However, later reports demon-
strated a preferential eect of IL-6 on switched antibody isotypes, given the fact that IL-6 only plays a role in
Figure 6. IL-6 has no eect on the phagocytic capacity of IgM+ B cells. Splenocytes were cultured in the
presence of IL-6 (200 ng/ml) or LPS (100 μ g/ml) for 24, 48 or 72 h at 20 °C. Non-stimulated controls were
also included. Aer the dierent incubation periods, cells were exposed to uorescent beads for a further
3 h at 20 °C. Non-ingested beads were removed by centrifugation over a cushion of 3% (w/v) BSA in PBS
supplemented with 4.5 (w/v) D-glucose (Sigma). (a) Data are shown as mean percentage of phagocytic IgM+
B cells (le) or Mean uorescence intensity (MFI) (right) + standard deviation from six independent sh.
* P < 0.05, * * P < 0.01. (b) Representative dot plots for each condition.
www.nature.com/scientificreports/
9
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
the terminal dierentiation of antigen-experienced B cells15. us, IL-6 over-expression in mice provokes IgG
plasmacytosis, but has no eects on plasma cells of other Ig isotypes53,54. Likewise, IL-6-decient mice showed
reduced antigen-specic IgG1, IgG2a and IgG3 levels aer immunization with a TD antigen, but IgM levels were
unaected17. IL-6 also supports the dierentiation of IgA-secreting plasma cells in dierent mucosal surfaces55,56.
Teleost sh do not express IgG, IgA or IgE and rely on non-switched IgM, IgD and IgT responses to ght infec-
tions. In this context, trout IL-6 was capable of inducing IgM secretion in spleen B cells, at a level comparable
Figure 7. Synergistic eects of IL-6 on antigen-induced in vivo IgM production. Rainbow trout were
injected i.p. with 100 μ l of PBS, 100 μ l of PBS with IL-6 (100 ng/sh), 100 μ l of PBS with 2 × 1010 TCID50/ml
inactivated IPNV, or 100 μ l of PBS containing the same amount of IPNV and IL-6. (a) e number of IgM
secreting cells was evaluated at days 2, 6 and 15 post-injection in spleen and kidney leukocyte cultures by
ELISPOT as described in the Materials and methods section (mean + standard deviation; n = 6). Asterisks
indicate signicant dierences between groups as indicated. * P < 0.05. (b) Total IgM, IPNV-specic and natural
(BSA; galactosidase; PC, phosphorylcholine; and poly I:C-specic) IgM titers were measured in the sera of
sh killed at day 15 post-immunization by ELISA. Results are shown as absorbance at 405 nm (for total IgM)
or absorbance at 492 nm (for specic IgMs) for individual sh. Bars indicate mean values in each group and
asterisks denote signicant dierences between groups as indicated. * P < 0.05.
www.nature.com/scientificreports/
10
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
to that induced by LPS. is increased IgM secretion in response to IL-6 was evidenced in ELISPOT and ow
cytometry and the fact that these IL-6 stimulated cells increased in size and up-regulated Blimp1 further supports
our observations indicating that trout IL-6 has the capacity to induce the dierentiation of unstimulated B cells to
IgM ASCs. Interestingly, LPS also provoked a size increase and augmented IgM secretion but without induction of
Blimp1, suggesting that either LPS exerts its activity on a dierent B cell population to IL-6 or alternatively it acts
on the same population but drives them towards a dierent activation state. In mammals, B2 cells dierentiate
into ASCs in response to LPS along with Blimp1 up-regulation, however, there is some controversy as to whether
B1 cells require Blimp1. While some studies revealed that mammalian B1 cells secrete IgM independently of
Blimp157, the IgM production by B1 cells in Blimp1-decient animals was inhibited, suggesting the requirement
of Blimp1 for normal IgM production58.
To further study the eects of IL-6 on IgM secretion, we performed additional experiments in v ivo. Here again,
the in vivo administration of IL-6 alone signicantly increased the number of IgM-secreting cells in the spleen
at day 2 post-injection. is eect was not observed in the kidney and was lost in the spleen at later sampling
points, revealing a tissue-specic transitional eect. In mammals, some of the positive eects that IL-6 has on
IgG expression are also transitional, since IL-6 is required for the normal induction, but not for the maintenance
of plasma cell responses in vivo because the eects of dierent survival factors are redundant59. Additionally,
we studied how IL-6 aected the antibody response elicited by the injection of an IPNV vaccine, observing that
although IPNV by itself was unable to induce a signicant IgM response, when combined with IL-6, the number
of IgM-secreting cells in the spleen or head kidney signicantly increased in comparison to sh immunized
with IPNV alone. Although the stimulatory eect on the number of IgM-secreting cells was no longer visible at
day 15 post-injection, at this point we could still see a synergistic eect of IL-6 on the amount of IPNV-specic
antibodies in serum. While total IgM levels were not signicantly higher in sh injected with IPNV and IL-6,
the increase in the amount of IPNV-specic IgM went along with a signicant decrease in the quantity of IgM
with alternative specicities, such as BSA- or galactosidase-specic IgM. Increases in antigen-specic IgG titers
with constant total IgG titers have also been reported in humans and the authors postulated that specic IgG
levels were increased without having an eect on total IgG titers because the ratio of antigen-specic cells was
very low in relation to the total pool of B cells60. Similarly, it seems that trout IL-6, when combined with IPNV,
increases IgM secretion of a discrete number of B cells with no eect on overall IgM production. Furthermore,
it seems that the negative eect of IL-6 on the production of natural antibodies is a conserved eect, as antibod-
ies against phosphorylcholine and LPS are increased in IL-6 gene knockout mice20. Taken together, our results
clearly demonstrate that IL-6 is able to enhance antigen-specic IgM responses in sh as observed in mammals
for switched Ig isotypes17,53,55,56.
When mammalian B cells start their dierentiation towards plasma cells, a secretory switch in the mRNA pro-
vokes the down-regulation of surface Ig and an increase of secreted Ig, resulting in the lack of BCR once the cells
are fully dierentiated40. However, recent studies have demonstrated that whilst this phenomenon occurs in IgG
plasma cells it is not seen in IgM or IgA-secreting plasma cells, as the latter preserve a functional BCR in the cell
surface even if fully dierentiated41. Similarly, we observed that the IL-6- or LPS-induced dierentiation of trout
B cells to IgM ASCs was not associated with a decrease in surface IgM levels. As hypothesized in mammals41, the
presence of a functional BCR on IgM ASCs would allow them to respond directly to their specic antigen upon
secondary encounters, unlike IgG ASCs. e fact that IL-6 and LPS-activated B cells retain a functional BCR,
despite their dierentiation to ASCs was conrmed by the mobilization of intracellular calcium in response to
anti-IgM cross-linking. Interestingly, these stimulated cells mobilized intracellular calcium at levels signicantly
higher than control B cells, demonstrating that IL-6 and LPS predispose B cells to a posterior signaling through
the BCR despite their dierentiation to ASCs. is is the rst time such an eect is reported for IL-6 in B cells and
consequently it would be interesting to determine the eect of mammalian IL-6 on BCR signaling in IgM- and
IgA-secreting plasma cells.
e transcription factor NF-κ B is critically involved in many cellular processes such as inammation, immune
response, proliferation or apoptosis61. In our experiments, we have established that IL-6 and LPS canonically
activate NF-κ B in IgM+ B cells from unstimulated sh. In mammals, BCR cross-linking, CD40 ligation62 or stim-
ulation with LPS or phorbol ester63 have been shown to activate NF-κ B in B cells. However, to our knowledge,
this is the rst report of IL-6-mediated NF-κ B activation in B cells, although IL-6 has been shown to activate this
transcription factor in epithelial cells43. As a consequence of NF-κ B activation, dierent immune genes should
be transcriptionally up-regulated. However, only LPS induced the transcription of typically NF-κ B-regulated
genes such as IL-1β , IL-8 and TNF-α , suggesting that although IL-6 activated cells translocate p65 to the nucleus,
additional cellular mechanisms modify the downstream eects. Of course, the up-regulation of IgM synthesis
observed in IL-6-stimulated B cells could be regulated by NF-κ B as the promoter of the IgM light chain is one of
the main targets for NF-κ B in mammals64. Since it is known that dierent NF-κ B components can be activated
through a variety of mechanisms with dierent downstream eects61, it seems possible that IL-6 and LPS are
modulating distinct NF-κ B activation pathways, and this should be studied further.
As trout IL-6 induced the transcription of several antimicrobial peptides in trout macrophages29 and taking
into account that B cells in sh have antimicrobial properties associated to their phagocytic activity9, we also
studied the eect of IL-6 and LPS on CATH-1, CATH-2 and hepcidin transcription. While CATH-1 and CATH-2
were up-regulated by LPS, IL-6 only up-regulated CATH-2, as occurred in macrophages29. Despite the fact that
IL-6 modulates hepcidin transcription in macrophages29, IL-6 had no eects on hepcidin expression in IgM+ B
cells. Nevertheless, these results highlight the potent antimicrobial properties of sh B cells. Similar to other anti-
gen presenting cells, B cells express MHC II on the cell surface. us, we determined the eects of IL-6 and LPS
on MHC II surface expression levels and the phagocytic capacity of trout B cells. Again the eects exerted by IL-6
and LPS were quite dierent, all of them summarized in Table1. While IL-6 signicantly down-regulated MHC-II
surface expression on B cells, as expected in an ASC65, LPS provoked a signicant up-regulation of MHC-II
www.nature.com/scientificreports/
11
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
expression and increased mRNA levels of the co-stimulatory molecules CD80/86 and CD83. e activation of
IL-6/STAT3 also induces the suppression of antigen presentation in human dendritic cells66. In contrast, and as
seen in our studies, mammalian B cells stimulated with LPS up-regulate MHC-II surface expression together
with co-stimulatory molecules (CD86, CD40)67 and plasma cells dierentiated in response to TI antigens retain
MHC-II expression and a functional antigen presenting machinery68. us, it seems that plasma cells generated
in response to LPS or other TI antigens, are not exclusively specialized in antibody secretion and still play a role
in other immune functions such as antigen presentation and pathogen clearance, and in line with this hypothesis,
LPS provoked a slight but signicant increase in the number of phagocytic B cells in the spleen cultures. is
increase was observed from 24 to 72 h post-treatment, whereas IL-6 had no eect. Since mammalian B1 cells are
also phagocytic, it would be interesting to determine if LPS is capable of increasing their phagocytic capacity.
Overall our results demonstrate that in contrast to mammals, sh IL-6 can trigger unstimulated B cells to
initiate their dierentiation towards ASCs. Consequently, trout IL-6 has mitogenic eects on IgM+ cells and these
proliferating cells increase in size, augment their secretion of IgM, up-regulate Blimp1 and decrease their surface
MHC-II expression. On the other hand, LPS shows a potent mitogenic activity and increases the secretion of IgM,
while driving the cells towards a quite dierent prole that predicts additional functions such as increased anti-
gen presentation (given the increased MHC-II surface expression, up-regulated transcription of co-stimulatory
molecules and higher phagocytic activity) and a pro-inammatory role (through the up-regulation of IL-1ß, IL-8
and TNF-α ). ese results highlight that IgM secretion can be induced through quite distinct dierentiation
pathways in sh.
Methods
Animals. Healthy specimens of female rainbow trout (Oncorhynchus mykiss) of approximately 50–70 g were
obtained from Centro de Acuicultura El Molino (Madrid, Spain). Fish were maintained at the Animal Health
Research Center (CISA-INIA) laboratory at 14 °C with a re-circulating water system and 12:12 h light:dark photo-
period. Fish were fed twice a day with a commercial diet (Skretting, Spain). Prior to any experimental procedure,
sh were acclimatized to laboratory conditions for 2 weeks and during this period no clinical signs were ever
observed. e experiments described comply with the Guidelines of the European Union Council (2010/63/
EU) for the use of laboratory animals and were previously approved by the Ethics committee from the Instituto
Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA; Code CEEA 2011/044).
Leukocyte isolation. Rainbow trout were killed via benzocaine (Sigma) overdose and blood was extracted
with a heparinized needle from the caudal vein and diluted 10 times with Leibovitz medium (L-15, Life
Technologies) supplemented with 100 I.U./ml penicillin, 100 ug/ml streptomycin (P/S), 10 units/ml heparin and
5% fetal calf serum (FCS) (all supplements also obtained from Life Technologies). Spleen and kidney were col-
lected and single cell suspensions generated using 100 μ m nylon cell strainers (BD Biosciences). Blood cell sus-
pensions were placed onto 51% Percoll (GE Healthcare) cushions whereas kidney and spleen suspensions were
placed onto 30/51% discontinuous density gradients. All suspensions were then centrifuged at 500 × g for 30 min
at 4 °C. e interface cells were collected and washed twice with L-15 containing 5% FCS.
Immune function IL-6 LPS
STAT3 transcription Up-regulated Up-regulated
SOCS3 transcription Up-regulated No eect
STAT3 phosphorylation Activated Activated
Mitogenic eects Signicant Very signicant
IgM secretion Increased Increased
mIgM expression Increased Increased
Total IgM expression Increased Increased
Blimp1 transcription Up-regulated No eect
ACKR2 transcription Up-regulated No eect
p65 translocation to nucleus Ye s Ye s
IL-1β transcription No eect Up-regulated
IL-8 transcription No eect Up-regulated
TNF-α transcription No eect Up-regulated
CATH1 transcription No eect Up-regulated
CATH2 transcription Up-regulated Up-regulated
Hepcidin transcription No eect No eect
BCR signaling to anti-IgM Increased Increased
MHC-II expression Decreased Increased
Phagocytic activity No eect Increased
CD80/86 transcription No eect Up-regulated
CD83 transcription No eect Up-regulated
Table 1. Summary table comparing the eects of IL-6 and LPS on trout spleen IgM+ B cells.
www.nature.com/scientificreports/
12
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
Production of rainbow trout recombinant IL-6. e sequence encoding the mature peptide of trout IL-669
was amplied from spleen cDNA prepared from Aeromonas salmonicida infected sh70 and cloned to the pET/
Duet-1 vector (Novagen, UK). e N-terminal ATG (M) codon and a C-terminal His tag (GSGHHHHHHHHHH)
were incorporated from the vector for translation initiation and for purication of the recombinant protein. us,
the rtIL-6 had 211 amino acids, with a predicted molecular weight of 23.8 kDa and pI of 7.27. Sequence analysis
of three clones (pET/IL-6a, b, c) revealed that they were identical but diered by four nucleotides, of which two
resulted in two amino acid changes from the published IL-669, presumably due to polymorphism. e sequence has
been submitted to EMBL/GenBank/DDBJ databases under the accession number FR715329. A plasmid was used to
transform BL21 Star (DE3) competent cells (Invitrogen) and protein expression was induced by IPTG and puried
under denaturing conditions as described previously71. e puried, denatured rtIL-6 was refolded in refolding
buer containing 50 mM Tris–HCl, pH8.0, 0.5 M arginine, 0.5% Triton-100 and 5 mM β -mercaptoethanol at 4 °C for
2 days. e refolded rtIL-6 was repuried under native conditions and eluted at concentrations of up to 0.5 mg/ml.
LPS contamination was checked by examining the expression of a number of LPS-responsive genes, including
IL-1β , TNF-α , IL-8, IL-10 and IL-11, and by undertaking the Limulus amoebocyte lysate assay (Sigma) as per the
manufacturer’s instructions.
Cell stimulation. Total leukocyte populations were dispensed in 24-well plates at a density of 2 × 106 cells per ml
and incubated with the appropriate stimulus: recombinant trout IL-6 (200 ng/ml), LPS (100 μ g/ml; > 95% purity)
(Sigma), TNP-KLH (5 μ g/ml) (Biotools), TNP-LPS (5 μ g/ml) (Biotools) or anti-IgM Ab (2 μ g/μ l) (clone 1.14)72 at
concentrations previously optimized. Non-stimulated controls were always included. Cells were always incubated
at 20 °C for dierent periods of time depending on specic experiments.
Cell sorting. IgM+ B cells were sorted from spleen, blood or kidney leukocyte suspensions using a BD FACSAria
III (BD Biosciences) cell sorter. For this, leukocytes were incubated for 30 min on ice with an anti-trout IgM mAb
(1.14) coupled to phycoerythrin (PE) in Staining buer (PBS containing 1% FCS and 0.5% sodium azide) that
prevents cell activation. Following two washing steps, cells were resuspended in FACS buer and IgM+ B cells iso-
lated based on their FSC/SSC proles (to exclude the granulocyte gate) and then on the basis of the uorescence
emitted by the anti-trout IgM antibody. IgM+ and IgM cells were then collected in Trizol for subsequent RNA
isolation.
Real time PCR analysis of sorted cells. Total RNA was isolated from IgM+ sorted populations using
Tri-reagent (Life Technologies). e cDNA synthesis and real-time PCR analysis were performed as described
previously33,71. e primers (Supplementary Table S1) for real-time-PCR were designed so that at least one primer
crossed an intron, to ensure that genomic DNA could not be amplied under the PCR conditions used. e
expression level of each gene was rst normalized against the expression level of EF-1α and then expressed as a
fold change that was calculated as the average expression of the IL-6/LPS stimulated samples divided by that of
the controls.
Detection of phosphorylated STAT3. To conrm the phosphorylation of STAT3 in IL-6 and LPS-treated
B cells and demonstrate direct eects on IgM+ B cells, we rst sorted IgM+ cells from splenocyte cultures as
described above and then incubated them with the dierent stimuli in the absence of other cell types. For this,
sorted cells adjusted to 2 × 106 cells per ml were disposed in 96 well plates and incubated with IL-6 (200 ng/ml),
LPS (100 μ g/ml) or media alone for 1 h at 20 °C. Aer the incubation, cells were lysed using RIPA buer contain-
ing protease inhibitors (Roche). Proteins were fractionated onto a denaturing 12% SDS-PAGE gel and transferred
onto a polyvinylidene diuoride (PVDF) membrane (Immobilon-P; Millipore Merck). Aer blocking in PBS with
5% skim milk for 1 h, the membrane was incubated with a mAb against phospho-STAT3 (Ser727) (Santa Cruz
Biotechnology) in blocking solution at 4 °C overnight. Aer three washing steps, the membrane was incubated
for 1 h with the secondary antibody, a goat anti-mouse IgG-HRP conjugate (GE Healthcare Life Sciences). e
reactive bands were visualized with the ECL system (GE Healthcare Life Sciences).
B cell proliferation. e BrdU Flow Kit (Becton Dickinson) was used to measure the proliferation of IgM+
cells following manufacturers instructions. Splenocytes at a concentration of 2 × 106 cells per ml were incubated
for 3 days at 20 °C with the dierent stimuli as described above. Bromodeoxyuridine (BrdU, 10 μ M) was then
added to the cultures and the cells were incubated for an additional 24 h. Aer that time, trout cells were collected
and stained with anti-IgM-PE (1.14) antibody and then xed and permeabilized with Cytox/Cytoperm Buer
for 15 min on ice. Aerwards, cells were incubated with Cytoperm Permeabilization Buer Plus for 10 min on ice
and re-xed with Cytox/Cytoperm Buer for 5 min at RT. Cells were then incubated with DNase (30 μ g/106 cells)
for 1 h at 37 °C to expose the incorporated BrdU. Finally, cells were stained with FITC anti-BrdU antibody for
20 min at RT and analysed by ow cytometry (BD FACSCalibur, BD Biosciences).
ELISPOT analysis. ELISPOT was used to quantify the number of IgM-secreting B cells. For this, ELISPOT
plates containing Inmobilon-P membranes (Millipore) were activated with 70% ethanol for 30 s, coated with
anti-trout IgM mAb (clone 4C10) at 2 μ g/ml in PBS and incubated overnight at 4 °C. To block non-specic bind-
ing to the membrane, plates were then incubated with 2% bovine serum albumin (BSA) in PBS for 2 h at RT.
Stimulated (with IL-6 or LPS as a positive control) or unstimulated splenocytes from individual sh were added
to the wells in triplicate at a concentration of 1 × 105 cells per well. Aer 72 h of incubation at 20 °C, cells were
washed away 5 times with PBS and plates were blocked again with 2% BSA in PBS for 1 h at room temperature.
Aer blocking, biotinylated anti-trout IgM mAb (clone 4C10) was added to the plates and incubated at 1 μ g/ml fo r
1 h at RT. Following additional washing steps (5 times in PBS) the plates were developed using streptavidin-HRP
(ermo Scientic) at RT for 1 h, washed again with PBS and incubated with 3-amino 9-ethylcarbazole (Sigma
www.nature.com/scientificreports/
13
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
Aldrich) for 30 min at RT in the dark. Substrate reaction was stopped by washing the plates with tap water. Once
the membranes had dried, they were digitally scanned and spot counts determined by the ImmunoSpot Series 45
Micro ELISPOT Analyzer.
Determination of total IgM levels. To determine total (both intracellular and extracellular) IgM levels,
cells were xed for 5 min with 4% paraformaldehyde in PBS, then permeabilized for 30 min in permeabilizitation
buer (staining buer containing 0.1% saponin) and thereaer incubated with an anti-IgM-PE antibody in per-
meabilization buer for another 30 min. Aer incubation, cells were washed three times with staining buer and
analyzed by ow cytometry.
NF-kB activation. Trout splenocytes were isolated and seeded in complete MGFL-15 medium (MGFL-15
supplemented with P/S, 100 μ g/ml gentamicin, 10% newborn calf serum (Gibco) and 5% carp serum). Cells were
incubated for 24 h with complete media containing IL-6, LPS or control media alone. Following stimulation,
cells were xed in 1% formaldehyde, and washed twice in PBS with 2% calf serum and 0.1% saponin (perme-
abilization buer). To determine nuclear translocation in IgM+ cells, splenocytes were stained with anti-p65
(Santa Cruz Biotechnology) and anti-trout IgM (1.14) for 30 min at 4 °C followed by 20 min at RT. Following the
primary staining, cells were washed and stained with goat anti-rabbit APC (Jackson ImmunoResearch) and rab-
bit anti-mouse FITC (Jackson ImmunoResearch). Prior to acquisition, Hoechst33342 nuclear stain (Molecular
Probes) was added as per the manufacturer’s recommendations. Data were collected on an ImageStream MKII
and analyzed using IDEAS soware (Amnis), as described previously11.
Analysis of MHC-II expression. e levels of MHC-II expression on the surface of IgM+ B cells were
measured via ow cytometry using a mAb against trout MHC-II73. Stimulated or unstimulated splenocytes were
washed in Staining Buer and co-incubated with PE-conjugated anti-trout IgM and the Alexa 647-conjugated
anti-MHC-II antibody for 30 min at 4 °C protected from light. Finally cells were washed twice with the same
buer and analysed by ow cytometry.
Phagocytic activity. Puried splenocytes (2 × 106) were cultured in 1 ml/well in 24-well plates in the pres-
ence or absence of IL-6 or LPS for 24, 48 or 72 h. Aer each time point, cells were incubated for 16 h at 20 °C
with uorescent beads (FluoSpheres Microspheres, 1.0 μ m, Crimson Red Fluorescent 625/645, 2% solids; Life
Technologies) at a cell/bead ratio of 1:10 or without beads as negative controls. Non-ingested beads were removed
by centrifugation (100 × g for 10 min at 4 °C) over a cushion of 3% (w/v) BSA (Fraction V; Fisher Scientic) in
PBS supplemented with 4.5 (w/v) D-glucose (Sigma). Aerwards, cells were resuspended in staining buer, labe-
led with PE-anti-IgM mAb (30 min at 4 °C), washed with the same buer and analyzed by ow cytometry.
Calcium ux. For calcium ux analysis, the calcium indicator Fluo-3 AM (Life Technologies) was used, fol-
lowing the manufacturers instructions. Briey, Fluo-3 was dissolved in DMSO and further diluted in an equal
volume of 20% (w/v) Pluronic F-127 (Life Technologies). Splenocytes were cultured in the presence or absence
of LPS or IL-6 during 24, 48 or 72 h. Aer each time point, cells were diluted in L-15 medium without FCS and
incubated with Fluo-3 AM at a nal concentration of 5 μ M for 1 h. Cells were then collected and washed, and a
baseline reading for 30 s acquired in a FACSCalibur ow cytometer. en 0.5 μ g/ml of anti-IgM were added to the
tube, and the emission of uorescence (525 nm) determined for 180 s in each sample.
Bioactivity of IL-6 in vivo. To assess the bioactivity of rtIL-6 in vivo, rainbow trout of approximately 5–7 g
were divided into three groups of 24 sh and injected intraperitoneally (i.p.) with 100 μ l of PBS, 100 μ l of PBS with
IL-6 (100 ng/sh), 100 μ l of PBS containing 2 × 1010 TCID50/ml inactivated IPNV (kindly donated by Professor
Øystein Evensen), or 100 μ l of PBS with a combination of IPNV and IL-6 at the same concentration as above. At
days 2, 6 and 15, six trout from each group were killed by benzocaine overdose. Blood was collected from the
caudal vein to determine antibody concentration and thereaer splenocytes and kidney leukocytes were isolated
to determine the number of IgM-secreting cells in ELISPOT assays as described above.
Antibody production. Serum samples were obtained aer blood clotting at RT for 1–2 h followed by incu-
bation overnight at 4 °C. Aerwards, the clot was centrifuged at 4000 rpm for 10 min and serum samples were
collected in a new tube that was centrifuged again at 10000 rpm for 10 min. Supernatants were nally collected in
dierent tubes and stored at 20 °C until use. e production of total IgM, IPNV-specic IgM and natural IgM
antibodies (with reactivity to TNP-BSA, galactosidase, phosphorylcholine and poly I:C) was then established by
capture ELISA.
To assess total IgM levels, 96-well ELISA plates were coated overnight with 100 μ l of 2 μ g/ml mouse anti-trout
Ig mAb 4C10. Wells were then blocked with 100 ul of 1% BSA in 1% Tween-20 PBS for 1 h at RT. Plates were
washed 3 times with PBS-1% Tween-20 and serum samples were diluted 1:100 in PBS-1% BSA and added to the
wells. Samples were incubated 1 h at RT and washed 3 times in PBS-1% Tween-20. en, 50 μ l of biotinylated
4C10 mAb (1 μ g/ml) diluted in blocking buer were added to the wells and samples incubated for 1 h at RT. Aer
three washing steps, plates were incubated with 50 μ l of Streptavidin-HRP (1:2000 in PBS-1% BSA) for 1 h at RT.
Wells were washed again 3 times and then 50 μ l of TMB substrate (Sigma) were added. Absorbance at OD405 was
measured in a FLUO Star Omega Microplate Reader.
An ELISA method was also used to measure IPNV-specic antibodies. Plates were coated with 100 μ l of poly-
clonal anti-IPNV antibody diluted at 1:5000 in diluent buer (1% fat free dry milk-PBS) and incubated overnight
at 4 °C. Plates were washed 3 times in PBS-0.05% Tween-20 and blocked with 5% fat free dry milk-PBS for 2 h
at RT. Aer 3 washing steps, serum samples were diluted at 1:100 in diluent buer and incubated overnight at
www.nature.com/scientificreports/
14
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
4 °C. Aer that, biotinylated 4C10 mAb and Streptavidin-HRP were used as described before. OPD substrate was
added to the wells and samples were measured at OD492 nm.
In order to measure the levels of natural IgM antibodies, plates were coated with 100 μ l of 15 μ g/ml of
TNP-BSA, 10 μ g/ml galactosidase, 20 μ g/ml phosphorylcholine or 20  g/ml poly I:C diluted in PBS and incu-
bated overnight at 4 °C. Plates were then blocked with 100 μ l of 1% BSA in 1% Tween-20 in PBS for 1 h at RT. Aer
washing steps in PBS-1% Tween-20, the serum samples were diluted 1:100 in PBS-1% BSA and added to the wells.
Biotinylated 4C10 mAb and Streptavidin-HRP were also used as described in total anti-IgM detection ELISA.
Finally, OPD substrate was added to the wells and samples were measured at OD492 nm.
Statistical analysis. Statistical analyses were performed using a two-tailed Student’s t test with Welch’s cor-
rection when the F test indicated that the variances of both groups diered signicantly. e dierences between
the mean values were considered signicant on dierent degrees, where * means P 0.05, * * means P 0.01 and
* * * means P 0.005 (GraphPad Prism 4 soware).
References
1. Cerutti, A., Puga, I. & Cols, M. New helping friends for B cells. Eur. J. Immunol. 42, 1956–1968 (2012).
2. Herrin, B. . & Cooper, M. D. Alternative adaptive immunity in jawless vertebrates. J. Immunol. 185, 1367–1374 (2010).
3. Zapata, A. G., Torroba, M., Vicente, A., Varas, A., Sacedon, . & Jimenez, E. e relevance of cell microenvironments for the
appearance of lympho-haemopoietic tissues in primitive vertebrates. Histol. Histopathol. 10, 761–778 (1995).
4. Abos, B., Castro, ., Pignatelli, J., Luque, A., Gonzalez, L. & Tafalla, C. Transcriptional heterogeneity of IgM(+ ) cells in rainbow
trout (Oncorhynchus myiss) tissues. PLoS One 8, e82737 (2013).
5. Tacchi, L. et al. Nasal immunity is an ancient arm of the mucosal immune system of vertebrates. Nat. Commun. 5, 5205 (2014).
6. Salinas, I. & Parra, D. Fish mucosal immunity: intestine. In: Mucosal Health in Aquaculture (ed. Bec, B. H., Peatman, E.). Elsevier
(2015).
7. Zhang, Y. A. et al. IgT, a primitive immunoglobulin class specialized in mucosal immunity. Nat. Immunol. 11, 827–835 (2010).
8. Xu, Z. et al. Teleost sin, an ancient mucosal surface that elicits gut-lie immune responses. Proc. Natl. Acad. Sci. USA 110,
13097–13102 (2013).
9. Li, J. et al. B lymphocytes from early vertebrates have potent phagocytic and microbicidal abilities. Nat. Immunol. 7, 1116–1124
(2006).
10. Parra, D. et al. Pivotal advance: peritoneal cavity B-1 B cells have phagocytic and microbicidal capacities and present phagocytosed
antigen to CD4+ T cells. J. Leuoc. Biol. 91, 525–536 (2012).
11. Abos, B., Castro, ., Gonzalez Granja, A., Havixbec, J. J., Barreda, D. . & Tafalla, C. Early activation of teleost B cells in response
to rhabdovirus infection. J. Virol. 89, 1768–1780 (2015).
12. Castro, ., Abos, B., Gonzalez, L., Aquilino, C., Pignatelli, J. & Tafalla, C. Molecular characterization of CD9 and CD63, two
tetraspanin family members expressed in trout B lymphocytes. Dev. Comp. Immunol. 51, 116–125 (2015).
13. Taga, T. et al. Interleuin-6 triggers the association of its receptor with a possible signal transducer, gp130. Cell 58, 573–581 (1989).
14. Hirano, T., Aira, S., Taga, T. & ishimoto, T. Biological and clinical aspects of interleuin 6. Immunol. Today 11, 443–449 (1990).
15. Jego, G., Bataille, . & Pellat-Deceunync, C. Interleuin-6 is a growth factor for nonmalignant human plasmablasts. Blood 97,
1817–1822 (2001).
16. Gonzalez-Garcia, I., Ocana, E., Jimenez-Gomez, G., Campos-Caro, A. & Brieva, J. A. Immunization-induced perturbation of human
blood plasma cell pool: progressive maturation, IL-6 responsiveness, and high PDI-BF1/BLIMP1 expression are critical
distinctions between antigen-specic and nonspecic plasma cells. J. Immunol. 176, 4042–4050 (2006).
17. opf, M., Herren, S., Wiles, M. V., Pepys, M. B. & osco-Vilbois, M. H. Interleuin 6 inuences germinal center development and
antibody production via a contribution of C3 complement component. J. Exp. Med. 188, 1895–1906 (1998).
18. opf, M. et al. Impaired immune and acute-phase responses in interleuin-6-decient mice. Nature 368, 339–342 (1994).
19. amsay, A. J. et al. e role of interleuin-6 in mucosal IgA antibody responses in vivo. Science 264, 561–563 (1994).
20. Bao, S., Husband, A. J. & Beagley, . W. B1 B cell numbers and antibodies against phosphorylcholine and LPS are increased in IL-6
gene nocout mice. Cell. Immunol. 198, 139–142 (1999).
21. Cong, Y. Z., abin, E. & Wortis, H. H. Treatment of murine CD5- B cells with anti-Ig, but not LPS, induces surface CD5: two B-cell
activation pathways. Int. Immun ol. 3, 467–476 (1991).
22. Fagarasan, S. & Honjo, T. T-Independent immune response: new aspects of B cell biology. Science 290, 89–92 (2000).
23. Bernasconi, N. L., Onai, N. & Lanzavecchia, A. A role for Toll-lie receptors in acquired immunity: up-regulation of TL9 by BC
triggering in naive B cells and constitutive expression in memory B cells. Blood 101, 4500–4504 (2003).
24. Garraud, O. et al. evisiting the B-cell compartment in mouse and humans: more than one B-cell subset exists in the marginal zone
and beyond. BMC Immunol. 13, 63 (2012).
25. Chiron, D., Beeredjian-Ding, I., Pellat-Deceunync, C., Bataille, . & Jego, G. Toll-lie receptors: lessons to learn from normal and
malignant human B cells. Blood 112, 2205–2213 (2008).
26. Pietretti, D. & Wiegertjes, G. F. Ligand specicities of Toll-lie receptors in sh: Indications from infection studies. Dev. Comp.
Immunol. 43, 205–222 (2014).
27. Zwollo, P., Cole, S., Bromage, E. & aattari, S. B cell heterogeneity in the teleost idney: evidence for a maturation gradient from
anterior to posterior idney. J. Immunol. 174, 6608–6616 (2005).
28. Mihara, M., Hashizume, M., Yoshida, H., Suzui, M. & Shiina, M. IL-6/IL-6 receptor system and its role in physiological and
pathological conditions. Clin. Sci. (Lond) 122, 143–159 (2012).
29. Costa, M. M., Maehr, T., Diaz-osales, P., Secombes, C. J. & Wang, T. Bioactivity studies of rainbow trout (Oncorhynchus myiss)
interleuin-6: eects on macrophage growth and antimicrobial peptide gene expression. Mol. Immunol. 48, 1903–1916 (2011).
30. Friederichs, ., Schmitz, J., Weissenbach, M., Heinrich, P. C. & Schaper, F. Interleuin-6-induced proliferation of pre-B cells
mediated by receptor complexes lacing the SHP2/SOCS3 recruitment sites revisited. Eur. J. Biochem. 268, 6401–6407 (2001).
31. Jego, G. et al. eactive plasmacytoses are expansions of plasmablasts retaining the capacity to dierentiate into plasma cells. Blood
94, 701–712 (1999).
32. Andersson, J., Sjoberg, O. & Moller, G. Induction of immunoglobulin and antibody synthesis in vitro by lipopolysaccharides. Eur. J.
Immunol. 2, 349–353 (1972).
33. Wan g, T. et al. First in-depth analysis of the novel 2-type cytoines in salmonid sh reveals distinct patterns of expression and
modulation but overlapping bioactivities. Oncotarget 7, 10917–10946 (2016).
34. Cain, . D., Jones, D. . & aison, . L. Antibody-antigen inetics following immunization of rainbow trout (Oncorhynchus myiss)
with a T-cell dependent antigen. Dev. Comp. Immunol. 26, 181–190 (2002).
35. Lee, S. . et al. B cell priming for extrafollicular antibody responses requires Bcl-6 expression by T cells. J. Exp. Med. 208, 1377–1388
(2011).
36. Bortnic, A., Chernova, I., Quinn, W. J. 3rd, Mugnier, M., Cancro, M. P. & Allman, D. Long-lived bone marrow plasma cells are
induced early in response to T cell-independent or T cell-dependent antigens. J. Immunol. 188, 5389–5396 (2012).
www.nature.com/scientificreports/
15
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
37. Chappell, C. P., Draves, . E., Giltiay, N. V. & Clar, E. A. Extrafollicular B cell activation by marginal zone dendritic cells drives T
cell-dependent antibody responses. J. Exp. Med. 209, 1825–1840 (2012).
38. Martins, G. & Calame, . egulation and functions of Blimp-1 in T and B lymphocytes. Annu. ev. Immunol. 26, 133–169 (2008).
39. Hansell, C. A. et al. Universal expression and dual function of the atypical chemoine receptor D6 on innate-lie B cells in mice.
Blood 117, 5413–5424 (2011).
40. Yoshida, T. et al. Memory B and memory plasma cells. Immunol ev 237, 117–139 (2010).
41. Pinto, D. et al. A functional BC in human IgA and IgM plasma cells. Blood 121, 4110–4114 (2013).
42. aneda, M., Odaa, T., Suetae, H., Tahara, D. & Miyadai, T. Teleost IL-6 promotes antibody production through STAT3 signaling
via IL-6 and gp130. Dev. Comp. Immunol. 38, 224–231 (2012).
43. Wang, L., Walia, B., Evans, J., Gewirtz, A. T., Merlin, D. & Sitaraman, S. V. IL-6 induces NF-appa B activation in the intestinal
epithelia. J. Immunol. 171, 3194–3201 (2003).
44. Pone, E. J. et al. BC-signalling synergizes with TL-signalling for induction of AID and immunoglobulin class-switching through
the non-canonical NF-appaB pathway. Nat. Commun. 3, 767 (2012).
45. Wilson, H. A., Greenblatt, D., Po enie, M., Finelman, F. D. & Tsien, . Y. Crosslinage of B lymphocyte surface immunoglobulin by
anti-Ig or antigen induces prolonged oscillation of intracellular ionized calcium. J. Exp. Med. 166, 601–606 (1987).
46. Yuse, M. I. & Lennon-Dumenil, A. M. Studying MHC class II presentation of immobilized antigen by B lymphocytes. Method s Mol.
Biol. 960, 529–543 (2013).
47. Zhang, Y. A., Hiima, J., Li, J., LaPatra, S. E., Luo, Y. P. & Sunyer, J. O. Conservation of structural and functional features in a
primordial CD80/86 molecule from rainbow trout (Oncorhynchus myiss), a primitive teleost sh. J. Immunol. 183, 83–96 (2009).
48. Eto, D. et al. IL-21 and IL-6 are critical for dierent aspects of B cell immunity and redundantly induce optimal follicular helper CD4
T cell (T) dierentiation. PLoS One 6, e17739 (2011).
49. Zwollo, P. Dissecting teleost B cell dierentiation using transcription factors. Dev. Comp. Immunol. 35, 898–905 (2011).
50. Levy, Y., Fermand, J. P. & Brouet, J. C. Dierential eects of low and high concentrations of interleuin 6 on human B cells. Eur. J.
Immunol. 20, 2389–2393 (1990).
51. Hirano, T. et al. Purication to homogeneity and characterization of human B-cell dierentiation factor (BCDF or BSFp-2). Proc.
Natl. Acad. Sci. USA 82, 5490–5494 (1985).
52. Muraguchi, A. et al. e essential role of B cell stimulatory factor 2 (BSF-2/IL-6) for the terminal dierentiation of B cells. J. Exp.
Med. 167, 332–344 (1988).
53. Suematsu, S. et al. IgG1 plasmacytosis in interleuin 6 transgenic mice. Proc. Natl. Acad. Sci. USA 86, 7547–7551 (1989).
54. Maeda, ., Mehta, H., Drevets, D. A. & Coggeshall, . M. IL-6 increases B-cell IgG production in a feed-forward proinammatory
mechanism to sew hematopoiesis and elevate myeloid production. Blood 115, 4699–4706 (2010).
55. oll, P. et al. In viv o eects of the anti-interleuin-6 receptor inhibitor tocilizumab on the B cell compartment. Arthritis heum. 63,
1255–1264 (2011).
56. Ladjemi, M. Z. et al. Increased IgA production by B-cells in COPD via lung epithelial interleuin-6 and TACI pathways. Eur.  esp ir.
J 45, 980–993 (2015).
57. Tumang, J. ., Frances, ., Yeo, S. G. & othstein, T. L. Spontaneously Ig-secreting B-1 cells violate the accepted paradigm for
expression of dierentiation-associated transcription factors. J. Immunol. 174, 3173–3177 (2005).
58. Savitsy, D. & Calame, . B-1 B lymphocytes require Blimp-1 for immunoglobulin secretion. J. Exp. Med. 203, 2305–2314 (2006).
59. Cassese, G. et al. Plasma cell survival is mediated by synergistic eects of cytoines and adhesion-dependent signals. J. Immunol.
171, 1684–1690 (2003).
60. Dumont, N., Aubin, E., Proulx, D. P., Lemieux, . & Bazin, . Increased secretion of hyperimmune antibodies following
lipopolysaccharide stimulation of CD40-activated human B cells in vitro. Immunology 126, 588–595 (2009).
61. Perins, N. D. & Gilmore, T. D. Good cop, bad cop: the dierent faces of NF-appaB. Cell Death Dier. 13, 759–772 (2006).
62. Francis, D. A., Sen, ., ice, N. & othstein, T. L. eceptor-specic induction of NF-appaB components in primary B cells. Int.
Immunol. 10, 285–293 (1998).
63. Liu, J. L., Chiles, T. C., Sen, . J. & othstein, T. L. Inducible nuclear expression of NF-appa B in primary B cells stimulated through
the surface Ig receptor. J. Immunol . 146, 1685–1691 (1991).
64. Sen, . & Baltimore, D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell 46, 705–716 (1986).
65. Calame, . L. Plasma cells: nding new light at the end of B cell development. Nat. Immunol. 2, 1103–1108 (2001).
66. itamura, H. et al. Activation of IL-6/STAT3-signaling cascade induces suppression of antigen presentation by human dendritic
cells (TUM4P.917). J. Immunol. 192, 138 (2014).
67. Xu, H., Liew, L. N., uo, I. C., Huang, C. H., Goh, D. L. & Chua, . Y. e modulatory eects of lipopolysaccharide-stimulated B cells
on dierential T-cell polarization. Immunology 125, 218–228 (2008).
68. Pelletier, N., McHeyzer-Williams, L. J., Wong, . A., Urich, E., Fazilleau, N. & McHeyzer-Williams, M. G. Plasma cells negatively
regulate the follicular helper T cell program. Nat. Immunol. 11, 1110–1118 (2010).
69. Iliev, D. B., Castellana, B., Macenzie, S., Planas, J. V. & G oetz, F. W. Cloning and expression analysis of an IL-6 homolog in rainbow
trout (Oncorhynchus myiss). Mol. Immunol. 44, 1803–1807 (2007).
70. Wang, T. & Secombes, C. J. Complete sequencing and expression of three complement components, C1r, C4 and C1 inhibitor, of the
classical activation pathway of the complement system in rainbow trout Oncorhynchus myiss. Immunogenetics 55, 615–628 (2003).
71. Wan g, T. et al. Functional characterization of a nonmammalian IL-21: rainbow trout Oncorhynchus myiss IL-21 upregulates the
expression of the  cell signature cytoines IFN-gamma, IL-10, and IL-22. J. Immunol. 186, 708–721 (2011).
72. DeLuca, D., Wilson, M. & Warr, G. W. Lymphocyte heterogeneity in the trout, Salmo gairdneri, dened with monoclonal antibodies
to IgM. Eur. J. Immunol. 13, 546–551 (1983).
73. Granja, A. G. et al. Identication of teleost sin CD8alpha+ dendritic-lie cells, representing a potential common ancestor for
mammalian cross-presenting dendritic cells. J. Immunol. 195, 1825–1837 (2015).
Acknowledgements
We would like to thank Lucia Gonzalez and Maria Sanz for technical assistance. Professor Øystein Evensen
is also acknowledged for providing us with the inactivated IPNV. is work was supported by the European
Research Council (ERC Starting Grant 2011 280469) and by the European Commission under the 7th Framework
Programme for Research and Technological Development (FP7) of the European Union (Grant Agreement
311993 TARGETFISH). T.W. received funding from the MASTS pooling initiative (e Marine Alliance for
Science and Technology for Scotland). MASTS is funded by the Scottish Funding Council (grant reference
HR09011).
Author Contributions
B.A. performed most of the experimental work. T.W. and C.J.S. produced the recombinant IL-6 and performed
all transcriptional analysis. R.C., E.L., A.G.G. and A.L assisted B.A. in tissue sampling and processing, especially
www.nature.com/scientificreports/
16
Scientific RepoRts | 6:30004 | DOI: 10.1038/srep30004
during in vivo experiments. J.H. and D.B. analyzed NF-κ B activation in stimulated cells. C.T. designed the
experiments and wrote the main body of the paper, with contributions from all other authors.
Additional Information
Supplementary information accompanies this paper at http://www.nature.com/srep
Competing nancial interests: e authors declare no competing nancial interests.
How to cite this article: Abós, B. et al. Distinct Dierentiation Programs Triggered by IL-6 and LPS in Teleost
IgM+ B Cells in e Absence of Germinal Centers. Sci. Rep. 6, 30004; doi: 10.1038/srep30004 (2016).
is work is licensed under a Creative Commons Attribution 4.0 International License. e images
or other third party material in this article are included in the article’s Creative Commons license,
unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license,
users will need to obtain permission from the license holder to reproduce the material. To view a copy of this
license, visit http://creativecommons.org/licenses/by/4.0/
... At a more functional level, we have determined their response to a wide range of immune stimuli known to regulate splenic B cells in rainbow trout, such as bacterial lipopolysaccharide (LPS), inactivated viral hemorrhagic septicemia virus (iVHSV), B cell activating factor (BAFF), or a combination of interleukin 10 (IL10) and CD40 ligand (CD40L). [25][26][27] The results obtained highlight the differences among IgM + IgD À and IgD + IgM À B cells in these two mucosal surfaces, suggesting unique roles for IgD in teleost mucosal immunity. ...
... Finally, we studied how these B cell subsets in gills and skin leukocyte cultures responded to a wide range of immune stimuli known to have strong effects on systemic B cells. [25][26][27] In gills, LPS stimulation significantly increased the percentages of IgM + IgD À B cells in cultures, whereas it decreased the percentage of IgD + IgM À B cells ( Figure 6). Stimulation with iVHSV also decreased the percentage of IgD + IgM À B cells in gill leukocyte cultures, but had no effect on the IgM + IgD À B cell subset ( Figure 6). ...
... 39 Nevertheless, this is not a general rule for teleost, given that rainbow trout B cells differentiated in response to IL6 decreased surface MHC II levels. 25 Interestingly, IgD + IgM À plasmablasts had significantly lower surface MHC II levels than mucosal IgM + IgD À B cells and had a significantly lower capacity to process antigens, strongly suggesting that rainbow trout IgD + IgM À plasmablasts no longer participate in antigen presentation or cognate T cell activation, while IgM-secreting plasmablasts still do. ...
Article
Full-text available
Although most B cells in teleost systemic compartments co-express IgM and IgD on the surface, cells exclusively expressing either of the two Igs are common in fish mucosal tissues, providing us with a unique opportunity to further characterize IgD+IgM− B cells, an intriguing B cell subset. Hence, we compared the phenotype of IgD+IgM− cells to that of IgM+IgD− B cells in rainbow trout gills and skin, also establishing the response of these subsets to immune stimulation. The transcriptional profile and secreting capacity of IgD+IgM− B cells corresponded to that of cells that have started a differentiation program toward plasmablasts, similarly to IgM+IgD− B cells. Yet, IgM+IgD− B cells retained high levels of surface MHC II and antigen-processing abilities, while these were much lower in IgD+IgM− cells, suggesting important differences in their antigen-presenting capacities. Our findings contribute to a deeper understanding of the enigmatic role of IgD in mucosal surfaces.
... LPS is a potent B cell mitogen that in iScience Article rainbow trout has been shown to increase the survival, IgM secretion, and MHC II surface expression of IgM + B cells and to promote their proliferation. 21 Thus, to study whether ER or mitochondria content of B cells were affected by LPS stimulation, we incubated isolated peripheral blood leukocytes (PBLs) with or without 100 mg/mL of LPS for 3 days in standard culture conditions and then used MitoTracker and ER Tracker to quantify the amount of mitochondria and ER in IgM + B cells by flow cytometry. This time point was selected after conducting a preliminary time course experiment in which we established that this was the point where LPS stimulatory effects were the highest ( Figure S1). ...
... For example, previous studies from our group demonstrated that when rainbow trout splenic B cells are stimulated with IL-6, a reduction of MHC II levels is observed. 21 Thus, it would be interesting to investigate in future studies ER and mitochondria levels in these IL-6-stimulated B cells. Similarly, in the peritoneal cavity, a subpopulation of B cells with a plasmablast phenotype expressed lower surface MHC II levels than peritoneal naive B cells (Granja and Tafalla, 2019). ...
Article
Full-text available
The differentiation of B cells into antibody-secreting cells is fundamental for the generation of humoral immunity. In mammals, this process involves a series of metabolic and intracellular changes, not studied to date in teleost fish, where a clear distinction between naïve B cells and plasmablasts/ plasma cells (PCs) is still missing. Thus, in the current study, we have established that upon activation, teleost B cells undergo an expansion of the endoplasmic reticulum (ER) but experience no significant changes in mitochondria content. In parallel, the transcription of genes implicated in B cell differentiation increase, while that of mitochondrial genes decrease. In this context, ER monitoring has allowed us to distinguish between small cells with low amounts of ER (FSCloERlo B cells), that correspond to undifferentiated cells, and large cells with expanded ER (FSChiERhi B cells), characterized as plasmablasts. The results shed new light on the B cell differentiation process in teleosts and provide us with novel tools to study B cell function in these species.
... It has been previously established that as trout B cells differentiate towards a plasmablast/plasma cell profile, they lose surface IgD and they increase their size (20,33). In the same way, an attenuated MHC II expression is viewed as a hallmark of plasmablast/plasma cell differentiation (33,34). For this reason, we decided to analyze by flow cytometry both the size and the surface MHC II expression levels of the two major B cell populations in the trout AT, IgM + IgD + and IgM + IgDcells. ...
... Thus, it was postulated that such plasma cells are not exclusively specialized in antibody secretion but retain roles in antigen presentation and pathogen clearance through a functional antigen presenting machinery. Likewise, when rainbow trout splenic B cells are stimulated with LPS, they initiate a differentiation process that goes along with an upregulation of MHC II surface levels, while that induced by IL-6 implies a reduction of MHC II levels (34). On the other hand, and taking into account that the mammalian AT is a site were numerous B1 cells can be found, it should also be noted that, in fact, mammalian B1 and plasmablasts also share many features. ...
Article
Full-text available
The immune response of the adipose tissue (AT) has been neglected in most animal models until investigations in human and mice linked obesity to chronic inflammation, highlighting the immune nature of this tissue. Despite this, in teleost fish, only a few studies have addressed the immune role of the AT. These studies have mostly focused on reporting transcriptional changes in the AT in response to diverse intraperitoneally delivered stimuli. Although the presence of B cells within the AT was also previously revealed, these cells have never been phenotypically or functionally characterized and this is what we have addressed in the current study. Initially, the B cell populations present in the rainbow trout ( Oncorhynchus mykiss ) AT were characterized in comparison to B cells from other sources. As occurs in other rainbow trout tissues, IgM ⁺ IgD ⁺ , IgM ⁺ IgD ⁻ and IgD ⁺ IgM ⁻ B cell subsets were identified in the AT. Interestingly, AT IgM ⁺ IgD ⁻ B cells showed a transcriptional profile that agrees with that of cells that have committed to plasmablasts/plasma cells, being this profile much more pronounced towards a differentiation state than that of blood IgM ⁺ IgD ⁻ B cells. Accordingly, the IgM-secreting capacity of AT B cells is significantly higher than that of blood B cells. Additionally, AT IgM ⁺ IgD ⁺ B cells also showed specific phenotypic traits when compared to their counterparts in other tissues. Finally, we established how these B cell subsets responded when rainbow trout were intraperitoneally injected with a model antigen. Our results demonstrate that the AT hosts plasmablasts/plasma cells that secrete specific IgMs, as happens in the peritoneal cavity and systemic immune tissues. Although the presence of these antigen-specific IgM-secreting cells was more abundant in the peritoneal cavity, these specific differentiated B cells were detected in the AT for long time periods at levels similar to those of spleen and head kidney. Our results provide new evidence regarding the immune role of the teleost AT, indicating that it functions as a secondary lymphoid organ that promotes immunity to peritoneal antigens.
... Stimulants such as lipopolysaccharide (LPS), 12-Otetradecanoylphorbol-13-acetate (TPA or phorbol 12-myristate 13-acetate), and cytokines (interleukin-6 and granulocyte-macrophage colony-stimulating are used to increase the yield of metaphases, their role in IgM-negative B cells being limited. 21,22 Oligonucleotides containing CpG motif, such as synthetic DSP30, can stimulate cells of the immune system in vitro and hence can be used to increase the yield of metaphases in vitro. 23 Various cytokines such as IL-10, IL-2, and TNF-alpha can also be used as mitogens for increasing the yield of metaphases. ...
Article
Full-text available
Plasma cell dyscrasias are a heterogeneous group of neoplasms characterized by abnormal proliferation of plasma cells with or without over production of monoclonal immunoglobulins. Chromosomal abnormalities are acquired either early in the course of the disease or during disease progression. Plasma cell dyscrasias are categorized into multiple cytogenetic subtypes that form an integral component of risk-stratified treatment protocols. The primary genetic events are IgH gene translocations and non-random gains of chromosomes 3/5/7/9/11/15/19 and or 21. The secondary genetic events consist of chromosome 1 abnormalities (1p deletion and 1q gain or amplification), deletion 17p/TP53, deletion 13q, and MYC gene rearrangements. Plasma cells being at the end of differentiation spectrum of B cells, have low proliferative potential precluding the use of karyotyping in identification of chromosomal abnormalities. Analysis of enriched plasma cells using interphase fluorescent in situ hybridization (FISH) is the technique of choice for identifying these abnormalities. It is essential to enrich plasma cells before the FISH analysis, and numerous plasma cell enrichment techniques have been described. In the paper, we review the cytogenetic approach to identify clinically significant genetic aberrations including the effective use of FISH panels and plasma cell enrichment techniques.
... These phenotypic characteristics develop when naive B cells are activated and differentiate into plasmablast/plasma cells (33,37,38). In addition, surface expression of major (39). Our recent study of a single-cell transcriptome of AK leukocytes from Nile tilapia (Oreochromis niloticus) found that AK leukocytes comprise myeloid cells and lymphocytes, and that B cells can divided into subpopulations based on their differentiation stage (40). ...
Article
Full-text available
Plasma cells are terminally differentiated antibody-secreting B lymphocytes that contribute to humoral immunity by producing large numbers of antibodies. Increasing evidence suggests that teleost fish B cells share certain characteristics with mammalian B1 B cells, including antibody-secreting, phagocytic, and antigen-presenting capacities. However, the difference between mature B cells and plasma cells remains unclear. In this study, we found that, based on their light-scattering characteristics, tilapia anterior kidney (AK) leukocytes can be categorized into two IgM ⁺ B-cell subsets: the lymphoid (L) gate and granulocyte–monocyte/macrophage (G-M) subsets. G-M gate cells are more numerous than L-gate cells and have higher mean fluorescence, but lower forward scatter and side scatter. We analyzed the morphological and ultrastructural features of sorted IgM ⁺ cells and found that L-gate IgM ⁺ cells have a high nucleus–cytoplasm ratio and lymphocyte-like morphology, whereas G-M gate IgM ⁺ cells have a small nucleus, more abundant endoplasmic reticulum, and a larger number of mitochondria, and have a plasma cell-like or macrophage-like morphology. To further characterize the cell types, we examined the specific patterns of expression of B-cell- and T-cell-related genes. We found that B-cell-specific genes were expressed by both L-gate and G-M gate IgM ⁺ cells, and that G-M gate IgM ⁺ cells secreted extremely high levels of IgM. However, T-cell-related genes were highly expressed only in L-gate IgM – cells. These results suggest that G-M gate IgM ⁺ cells are similar to plasma-like cells, with high antibody-secreting capacity. Given that G-M gate cells include the granulocyte, monocyte, and macrophage cell types, but not B cells, monocyte/macrophage markers were used to investigate the cell types further. A macrophage receptor with a collagenous structure was frequently observed, and macrophage-expressed gene-1 was highly expressed, in the G-M gate IgM ⁺ cells. Phagocytic capacity, as determined by ingestion of beads or bacteria, was significantly higher in G-M gate IgM ⁺ cells than in L-gate IgM ⁺ cells, as was antigen-processing capacity. Our findings show that tilapia AK leukocytes can be divided into two IgM ⁺ B-cell subsets and that G-M gate IgM ⁺ cells resemble plasma-like cells, having high antibody-secreting, phagocytic, and antigen-presenting capacities. Thus, this study increases our understanding of the functions of teleost fish plasma-like cells.
... Therefore, these memory cells can react rapidly to a second antigen encounter and are essential for successful vaccination. Here, we observed that MIF was upregulated at D3 and the MHC II b chain was upregulated at D7. Interestingly, it has been proven that the MHC II b chain could induce the expression of tnf-a and il6 in fish and regulate and coordinate the interaction between cytokines and their receptors (40,41). It may suggest that the change of MIF and the MHC II b chain on the surface of antigen-presenting cells could stimulate the secretion of cytokines and activation of Th1type responses in spleen of rainbow trout post F. psychrophilum infection. ...
Article
Full-text available
Flavobacterium psychrophilum , the etiological agent of bacterial coldwater disease and rainbow trout fry syndrome, causes considerable losses in salmonid aquaculture globally. Systemic F. psychrophilum infections in rainbow trout ( Oncorhynchus mykiss ) lead to a range of clinical signs, including ulcerative lesions in the skin and muscle and splenitis. Previous studies offered an integrative analysis of the skeletal muscle response to F. psychrophilum infection in rainbow trout. However, little is known about the molecular mechanism of immune response in the spleen, which is an important immune organ of rainbow trout. Here, we investigated the time-course splenic transcriptome profiles in uninfected rainbow trout (CK) and F. psychrophilum –infected rainbow trout at day 3 and day 7 (D3, D7) by RNA-seq analyses. Among the 7,170 differentially expressed genes (DEGs) in the three comparisons (D3 vs. CK, D7 vs. CK, D3 vs. D7), 1,286 DEGs showed consistent upregulation or downregulation at D3 and D7 and were associated with pattern recognition, acute-phase response, complement cascade, chemokine and cytokine signaling, and apoptosis. The Real time quantitative PCR (RT-qPCR) analysis of eight DEGs confirmed the accuracy of the RNA-Sequencing (RNA-seq) data. Our results reflected a general process from pathogen recognition to inflammatory cytokine generation and delineated a putative Toll-like receptor signaling pathway in rainbow trout spleen, following F. psychrophilum infection. Taken together, these results provide new insights into the molecular mechanism of the immune response to F. psychrophilum infection and are a valuable resource for future research on the prevention and control of bacterial coldwater disease during salmon culture.
... Modifications of VEGF signaling were reported in studies with mammals exposed to LPS, an effect linked to the known effects of LPS on angiogenesis [111,112]. Interestingly, in vitro studies reported modifications in IL-1β expression in lumpfish leukocytes [113] and in gilthead seabream macrophages after 24 h bacterial exposure [114] and in IL-6 expression in LPS treated trout IgM+ B cells [115]. ...
Article
Full-text available
The environmental conditions and isolation in the Antarctic have driven the evolution of a unique biodiversity at a macro to microorganism scale. Here, we investigated the possible adaptation of the teleost Notothenia coriiceps immune system to the cold environment and unique microbial community of the Southern Ocean. The fish immune system was stimulated through an intraperitoneal injection of lipopolysaccharide (LPS 0111:B4 from E. coli) and the tissue transcriptomic response and plasma biochemistry were analyzed 7 days later and compared to a sham injected control. Gene transcription in the head-kidney, intestine and skin was significantly modified by LPS, although tissues showed different responsiveness, with the duodenum most modified and the skin the least modified. The most modified processes in head-kidney, duodenum and skin were related to cell metabolism (up-regulated) and the immune system (comprising 30% of differentially expressed genes). The immune processes identified were mostly down-regulated, particularly interleukins and pattern recognition receptors (PRRs), nucleotide-binding oligomerization domain-like receptors and mannose receptors, unlike the toll-like receptors response commonly described in other teleost fish. The modified transcriptional response was not mirrored by a modified systemic response, as the circulating levels of enzymes of innate immunity, lysozyme and antiproteases, were not significantly different from the untreated and sham control fish. In conclusion, while the N. coriiceps immune system shares many features with other teleosts there are also some specificities. Further studies should better characterize the PRRs and their role in Antarctic teleosts, as well as the importance of the LPS source and its consequences for immune activation in teleosts.
... A number of investigations have been performed to elucidate the regulation of fish immune system by cytokines. Previous researches have shown that interleukin-10 (IL-10) [18], chemokine 9 (CK9) [19], and type I interferon-3 (IFN I-3) [20] could enhance the phagocytosis of teleost IgM + B cells, while interleukin-6 (IL-6) [21] and B cell-activating factor (BAFF) [22] have no significant effect on phagocytic ability of IgM + B cells. Although a number of molecules participating the phagocytosis have been identified, yet little is known about the signaling pathway involving the phagocytosis in mIgM + lymphocytes. ...
Article
The phagocytic actives of B cells in fish have been proven in recent years. In this study, five positive hybridomas secreting monoclonal antibodies (MAbs) against largemouth bass IgM were produced. Indirect immunofluores- cence assay (IFA) demonstrated that five MAbs could specifically recognize membrane-bound IgM (mIgM) molecule of largemouth bass. Indirect ELISA and Western blotting analysis showed that all the five MAbs had no cross-reactions with the other two teleost IgMs. Flow cytometry analysis (FCM) revealed that the percentages of largemouth bass mIgM+ lymphocytes in head kidney, peripheral blood and spleen were 51.66 ± 0.608%, 16.5 ± 1.235% and 42.92 ± 1.091%, respectively. In addition, the phagocytosis rates of mIgM + lymphocytes ingesting Nocardia seriolae from head kidney, peripheral blood and spleen were calculated to be 5.413 ± 0.274%, 16.6 ± 0.289% and 26.3 ± 0.296%, respectively. The qPCR results of sorted cells indicated that most inflammatory cytokines (IFNγ, IL-1β, IL-2, IL-12β, IL-34, IL-10), chemokine (CXCL12), chemokines receptors (CXCR2, CXCR4) and genes (FcγRIa, NCF1, CFL, ARP2/3, CD45, Syk, MARCKS) related to FcγR-mediated phagocytic signaling pathway in phagocytic mIgM+ lymphocytes were up-regulated significantly (P < 0.05). Taken together, the results suggested that the MAb (MM06H) produced in this paper could be used as a tool to study mIgM+ lym- phocytes of largemouth bass, and FcγR may participate in the phagocytosis of mIgM+ lymphocytes, which is helpful to further study the role of mIgM+ lymphocytes in innate immunit
Article
T and B lymphocytes (T and B cells) are immune effector cells that play critical roles in adaptive immunity and defend against external pathogens in most vertebrates, including teleost fish. In mammals, the development and immune response of T and B cells is associated with cytokines including chemokines, interferons, interleukins, lymphokines, and tumor necrosis factors during pathogenic invasion or immunization. Given that teleost fish have evolved a similar adaptive immune system to mammals with T and B cells bearing unique receptors (B-cell receptors (BCRs) and T-cell receptors (TCRs)) and that cytokines in general have been identified, whether the regulatory roles of cytokines in T and B cell-mediated immunity are evolutionarily conserved between mammalians and teleost fish is a fascinating question. Thus, the purpose of this review is to summarize the current knowledge of teleost cytokines and T and B cells as well as the regulatory roles of cytokines on these two types of lymphocytes. This may provide important information on the parallelisms and dissimilarities of the functions of cytokines in bony fish versus higher vertebrates, which may aid in the evaluation and development of adaptive immunity-based vaccines or immunostimulants.
Article
Full-text available
IL-4 and IL-13 are closely related canonical type-2 cytokines in mammals and have overlapping bioactivities via shared receptors. They are frequently activated together as part of the same immune response and are the signature cytokines produced by T-helper (Th)2 cells and type-2 innate lymphoid cells (ILC2), mediating immunity against extracellular pathogens. Little is known about the origin of type-2 responses, and whether they were an essential component of the early adaptive immune system that gave a fitness advantage by limiting collateral damage caused by metazoan parasites. Two evolutionary related type-2 cytokines, IL-4/13A and IL-4/13B, have been identified recently in several teleost fish that likely arose by duplication of an ancestral IL-4/13 gene as a consequence of a whole genome duplication event that occurred at the base of this lineage. However, studies of their comparative expression levels are largely missing and bioactivity analysis has been limited to IL-4/13A in zebrafish. Through interrogation of the recently released salmonid genomes, species in which an additional whole genome duplication event has occurred, four genomic IL-4/13 loci have been identified leading to the cloning of three active genes, IL-4/13A, IL-4/13B1 and IL-4/13B2, in both rainbow trout and Atlantic salmon. Comparative expression analysis by real-time PCR in rainbow trout revealed that the IL-4/13A expression is broad and high constitutively but less responsive to pathogen-associated molecular patterns (PAMPs) and pathogen challenge. In contrast, the expression of IL-4/13B1 and IL-4/13B2 is low constitutively but is highly induced by viral haemorrhagic septicaemia virus (VHSH) infection and during proliferative kidney disease (PKD) in vivo, and by formalin-killed bacteria, PAMPs, the T cell mitogen PHA, and the T-cell cytokines IL-2 and IL-21 in vitro. Moreover, bioactive recombinant cytokines of both IL-4/13A and B were produced and found to have shared but also distinct bioactivities. Both cytokines rapidly induce the gene expression of antimicrobial peptides and acute phase proteins, providing an effector mechanism of fish type-2 cytokines in immunity. They are anti-inflammatory via up-regulation of IL-10 and down-regulation of IL-1β and IFN-γ. They modulate the expression of cellular markers of T cells, macrophages and B cells, the receptors of IFN-γ, the IL-6 cytokine family and their own potential receptors, suggesting multiple target cells and important roles of fish type-2 cytokines in the piscine cytokine network. Furthermore both cytokines increased the number of IgM secreting B cells but had no effects on the proliferation of IgM+ B cells in vitro. Taken as a whole, fish IL-4/13A may provide a basal level of type-2 immunity whilst IL-4/13B, when activated, provides an enhanced type-2 immunity, which may have an important role in specific cell-mediated immunity. To our knowledge this is the first in-depth analysis of the expression, modulation and bioactivities of type-2 cytokines in the same fish species, and in any early vertebrate. It contributes to a broader understanding of the evolution of type-2 immunity in vertebrates, and establishes a framework for further studies and manipulation of type-2 cytokines in fish.
Article
Immunosuppression in tumor microenvironments is one of the critical issues for cancer immunotherapy. We have been demonstrated that IL-6, a pleiotropic cytokine that regulate the growth, differentiation and survival of a variety of cells, significantly inhibited maturation of murine dendritic cells (DCs) via STAT3 activation both in vitro and in vivo. In this study, we focused on IL-6/STAT3-signaling cascade in human dendritic cells (DCs), and investigated the effects of IL-6 on antigen-presenting ability of DCs. Surface HLA class I, HLA class II, and costimulatory molecules of human DCs were evaluated after the stimulation with IL-6 in vitro. As a result, HLA-DR and CD86 expressions were significantly reduced by IL-6 treatment of human DCs. The reduction was remarkably blocked in the presence of STAT3 inhibitor. In addition, IFN-γ production by T cells after TCR-stimulation was significantly suppressed in the presence of IL-6-treated DCs compared with control DCs. Moreover, we found that activation of antigen-specific CD4+ T cells by DCs was remarkably reduced by the IL-6-treatment. In this study, we demonstrated that IL-6/STAT3-signaling cascade was one of the regulating factors for antigen presentation of DCs. From these findings suggest that IL-6/STAT3 signaling cascade regulates immunosuppressive function of human DCs, which would be a promising target for improving the effects of cancer immunotherapy.
Article
Circulating plasma cells in 10 cases of reactive plasmacytosis had a shared phenotype with early plasma cell (CD19+CD38+ CD138+ CD40+CD45+ CD11a+ CD49e−CD56−). In most cases, a minor subpopulation of CD28+ plasma cells was also detected. Reactive plasma cells were highly proliferative, suggesting the presence of circulating progenitors (plasmablasts). After CD138+ plasma cell removal, highly proliferative CD138− plasmablasts differentiated into CD138+ plasma cells within a few days. This differentiation, which was associated with increased CD38 and decreased HLA-DR expression, was further confirmed by a large increase in intracellular Ig content (associated with Ig secretion) and was concomitant with extensive secretion of interleukin-6 (IL-6). The addition of neutralizing anti–IL-6 and anti-CD126 (IL-6 receptor) monoclonal antibodies totally prevented Ig secretion and cell differentiation by inducing apoptosis of plasmablasts, which indicates that IL-6 is an essential survival factor for plasmablasts. This report provides the first characterization of normal plasmablasts and shows that their phenotype is not exactly that of multiple myeloma cells.
Article
Anti-lg stimulated murine B cells express high levels of surface CD5 (ly-1) and increased CD44 while maintaining surface IgD, CD23 and J11d. Sorting of CD5- and CD5+ cells demonstrates that anti-lg induces CD5 expression rather than the selective expansion of CD5+ cells. Anti lg plus interleukin-6 (IL-6) induces the CD23, IgD, low ly-5 (B220) (CD45low), J11d(high) phenotype of typical CD5+ peritoneal B cells. In contrast, lipopolysaccharide (LPS)-stimulated B cells have high levels of CD44 but decreased surface IgD, CD23 and J11d and no CD5. Thus LPS and anti-lg generate activated cells with differing phenotypes. Induced CD5+ cells have increased viability, even in the absence of added exogenous factors, while the viability of CD5- B cells is dependent on factors such as IL-4. We conclude that conventional CD5- B cells can be activated by either of two pathways: one generating CD5+ B cells; the other yielding conventional activated cells. We hypothesize that the first path requires slg cross-linking and corresponds to T-independent (type 2) stimulation, while cognate interaction with helper T cells in the absence of slg cross-linking induces B cells to enter the second path.
Article
Interleukin 6(IL-6) is a multi-functional cytokine that is produced by a range of cells and plays a central role in host defense mechanisms. Abnormal production of IL-6 has been suggested to be involved in glomerulonephritis, plasmacytomagenesis and in the pathogenesis of autoimmune diseases. In this review, Toshio Hirano and colleagues discuss the possible involvement of IL-6 in a variety of diseases, the regulatory mechanism(s) of expression of the IL-6 gene and the structure and function of the IL-6 receptor.
Article
Although fish constitute the most ancient animal group in which an acquired immune system is present, the presence of dendritic cells (DCs) in teleosts has been addressed only briefly, and the identification of a specific DC subset in teleosts remained elusive because of the lack of specific Abs. In mice, DCs expressing CD8α(+) in lymphoid tissues have the capacity to cross-present extracellular Ags to T cells through MHC I, similarly to tissue-derived CD103(+) DCs and the human CD141(+) DC population. In the current study, we identified a large and highly complex subpopulation of leukocytes coexpressing MHC class II and CD8α. This CD8α(+) MHC II(+) DC-like subpopulation constituted ∼1.2% of the total leukocyte population in the skin, showing phenotypical and functional characteristics of semimature DCs that seem to locally regulate mucosal immunity and tolerance in a species lacking lymph nodes. Furthermore, we identified trout homologs for CD141 and CD103 and demonstrated that, in trout, this skin CD8(+) DC-like subpopulation expresses both markers. To our knowledge, these results provide the first evidence of a specific DC-like subtype in nonimmune tissue in teleosts and support the hypothesis of a common origin for all mammalian cross-presenting DCs. Copyright © 2015 by The American Association of Immunologists, Inc.
Chapter
Mucosal Health in Aquaculture is an essential reference on mucosal health for the diverse aquaculture community. Rich in explanatory figures and schematics, the book includes important concepts such as structural and cellular composition of mucosal surfaces in fish and shellfish, known functional roles of molecular and cellular actors during pathogen invasion, impacts of nutrition on the mucosal barriers, impacts of chemical treatments on mucosal surfaces, mucosal vaccines and vaccination strategies, and more. The health of cultured aquaculture species is critical in establishing the sustainable growth of the aquaculture industry worldwide, and mucosal health is of particular interest to those working in aquaculture because mucosal surfaces (skin, gill, intestine, reproductive tissues) constitute the first line of defense against pathogen invasion. Mucosal Health in Aquaculture captures the latest research on mucosal barriers in aquaculture species and their impacts on nutrition and immunity to ensure sustainable aquaculture development.
Article
Our results indicate that B lymphocytes stimulated with anti-Ig or antigen exhibit repetitive [Ca2+]i transients which persist for hours. The magnitude of these transients favors an important and ongoing role for [Ca2+]i elevation in antigen driven B cell activation. Repetitive Ca2+ transients may prove to be a prevalent mechanism of Ca2+ signaling. In preliminary experiments (with L. E. Samelson and R. D. Klausner), we have observed Ca2+ transients in cloned T cells stimulated with antigen. Woods et al. have described repetitive free Ca2+ transients in hepatocytes stimulated with extracellular ligands promoting glycogenolysis, and suggest that the intervals of base-line [Ca2+]i levels explain the absence of mitochondrial overload in chronically stimulated cells. These considerations apply equally to B lymphocytes and recommend caution in delineating the range of Ca2+-mediated functions by prolonged coculture of cells with Ca2+ ionophores. Our experiments were done in a simple recording chamber with one cell type. No cell interactions were observed. Given the variety of indicator dyes now available, the technical approach we present, augmented by a more sophisticated recording chamber, is a potentially powerful tool for examining the intrinsic, and T- or accessory cell-dependent, physiology of B cell differentiation.