ArticlePDF AvailableLiterature Review

Abstract and Figures

Macrophages are found across all vertebrate species, reside in virtually all animal tissues, and play critical roles in host protection and homeostasis. Various mechanisms determine and regulate the highly plastic functional phenotypes of macrophages, including antimicrobial host defenses (pro-inflammatory, M1-type), and resolution and repair functions (anti-inflammatory/regulatory, M2-type). The study of inflammatory macrophages in immune defense of teleosts has garnered much attention, and antimicrobial mechanisms of these cells have been extensively studied in various fish models. Intriguingly, both similarities and differences have been documented for the regulation of lower vertebrate macrophage antimicrobial defenses, as compared to what has been described in mammals. Advances in our understanding of the teleost macrophage M2 phenotypes likewise suggest functional conservation through similar and distinct regulatory strategies, compared to their mammalian counterparts. In this review, we discuss the current understanding of the molecular mechanisms governing teleost macrophage functional heterogeneity, including monopoetic development, classical macrophage inflammatory and antimicrobial responses as well as alternative macrophage polarization towards tissues repair and resolution of inflammation.
Content may be subject to copyright.
Biology 2015, 4, 881-906; doi:10.3390/biology4040881
biology
ISSN 2079-7737
www.mdpi.com/journal/biology
Review
Biology of Bony Fish Macrophages
Jordan W. Hodgkinson 1,*, Leon Grayfer 2 and Miodrag Belosevic 1
1 Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada;
E-Mail: mbelosev@ualberta.ca
2 Department of Biological Sciences, George Washington University, Washington, DC 20052, USA;
E-Mail: leon_grayfer@email.gwu.edu
* Author to whom correspondence should be addressed; E-Mail: jordanwi@ualberta.ca;
Tel.: +1-780-492-1265; Fax: +1-780-492-9234.
Academic Editor: Brian Dixon
Received: 26 October 2015 / Accepted: 24 November 2015 / Published: 30 November 2015
Abstract: Macrophages are found across all vertebrate species, reside in virtually all
animal tissues, and play critical roles in host protection and homeostasis. Various mechanisms
determine and regulate the highly plastic functional phenotypes of macrophages, including
antimicrobial host defenses (pro-inflammatory, M1-type), and resolution and repair functions
(anti-inflammatory/regulatory, M2-type). The study of inflammatory macrophages in
immune defense of teleosts has garnered much attention, and antimicrobial mechanisms of
these cells have been extensively studied in various fish models. Intriguingly, both similarities
and differences have been documented for the regulation of lower vertebrate macrophage
antimicrobial defenses, as compared to what has been described in mammals. Advances in
our understanding of the teleost macrophage M2 phenotypes likewise suggest functional
conservation through similar and distinct regulatory strategies, compared to their mammalian
counterparts. In this review, we discuss the current understanding of the molecular
mechanisms governing teleost macrophage functional heterogeneity, including monopoetic
development, classical macrophage inflammatory and antimicrobial responses as well as
alternative macrophage polarization towards tissues repair and resolution of inflammation.
Keywords: teleost; macrophages; antimicrobial; cytokine; respiratory burst; nitric oxide;
nutrient depravation
OPEN ACCESS
Biology 2015, 4 882
1. Introduction
Macrophage lineage cells present a remarkably versatile array of functional specializations across
vertebrates. As resident cells in virtually all tissues, macrophages aid in maintaining homeostatic
environments, and upon infection, are typically one of the first cell types to encounter intruding
pathogens, where they orchestrate appropriate immune responses. Our understanding of macrophage
biology has greatly expanded since the first description of starfish larvae phagocytes in 1882 by Élie
Metchnikoff, who was later awarded the Nobel Prize for his contribution to cellular immunity in
1908 [1]. Since then, the macrophage has emerged as an essential cell type in all vertebrates, endowed
with a panoply of functional capabilities.
Much of our understanding of macrophage biology comes from research in mammalian models,
where distinct macrophage subsets of have been characterized, including classically activated cells by
interferons (IFN) and tumor necrosis factor alpha (TNFα) (M1); alternatively activated cells by IL-4
and IL-13 (M2a); macrophages activated by immune complexes or apoptotic cells (M2b); and
regulatory macrophages, deactivated by IL-10, TGF-β, or glucocorticoids (M2c), which culminate in
the various effector subtypes, broadly described as having a “kill” or “heal” response (reviewed in [2]).
Recent transcriptomic and proteomic analyses of macrophage populations derived by these distinct
stimuli have underlined the vast complexities of these mechanisms at the molecular level, controlling
the various physiological responses of macrophages.
In teleosts, the best characterized macrophage phenotype is that comparable to the M1 activation
state, which serves a critical role in host protection. These cells may rapidly kill pathogens by
engulfment and production of toxic reactive intermediates [3], phagolysosomal acidification [4], and
restriction of nutrient availability [5]. Furthermore, M1 macrophages are robust factories of cytokines,
chemokines, and lipid mediators, which act to potentiate and fine-tune the inflammatory and adaptive
immune responses. More recently, efforts to characterize alternative activation states of teleost
macrophages are focusing on the biology of fish IL4 and IL13 homologues (IL4/13A and IL4/13B)
and arginase activity, implying a conserved M2a phenotype [6,7]. Similar deactivating roles of
glucocorticoids (GC), immune complexes, IL-10, and TGF-β have been demonstrated in teleosts,
suggesting conserved functions of these ligands in deactivating or aiding in the tissue repair [810].
Characterization of the regulatory mechanisms responsible for shaping macrophage polarity is a
unique challenge in teleosts, as whole-genome duplication and gene-specific duplication events,
combined with disparate evolutionary pressures, have endowed distinct teleosts with multiple gene
copies, the product of some of which may have adopted respectively divergent roles [11].
This review discusses bony fish macrophage development, polarization, and functional responses,
thus comprehensively coalescing the current understanding of teleost macrophage biology.
2. Macrophage Development
2.1. Teleost Embryonic Monopoiesis
Akin to most other vertebrates studied to date, teleost fish blood cell development occurs within
primitive and definitive waves of hematopoiesis [1214]. Over the last few decades, the zebrafish has
emerged as a model of choice to study blood cell formation, owing to its optical transparency and
Biology 2015, 4 883
relative ease of genetic manipulation. During primitive hematopoiesis (1224 h post-fertilization),
embryonic mesoderm is committed to monopotent hematopoietic precursors that give rise to macrophages,
the first functional blood cell type, in the rostral blood island of the zebrafish embryo [12,14,15].
Following primitive monopoiesis, a transient definitive hematopoietic wave is initiated in the posterior
blood island, giving rise to the first multilineage progenitor cells, erythromyeloid progenitors (EMPs),
which develop into both erythroid and myeloid cells. Definitive hematopoiesis, defined by the emergence
of multipotent hematopoietic stem cells (HSCs), subsequently originates in the aorta-gonad-mesonephros
(AGM), and HSCs that precede monopoeisis are seeded into caudal and renal tissue, serving as the
main sources of myeloid cells roughly 34 days post-fertilization (dpi) [16]. The existence of renal
marrow-derived HSCs has been demonstrated in both zebrafish and gibuna carp, where transplantation
of renal HSCs from healthy donors to irradiated recipients reconstituted HSC pools of the stem
cell-depleted fish [17,18]. The complex and intricate processes described above are more
comprehensively reviewed in [13,16,19,20].
2.2. Teleost Adult Monopoiesis
For nearly a half a century, the widely accepted mononuclear phagocyte system theory proposed
that tissue macrophages are derived from circulating blood monocytes in vertebrates [21]. Although it
is well established that monocytes give rise to macrophages during inflammatory conditions in both
mammals and fish, recent evidence by fate-mapping blood cell lineages suggests a more limited
contribution of circulating monocytes to mature tissue macrophage replenishment. Instead, tissue
macrophages are seeded during primary hematopoiesis and self-maintain resident populations, as
demonstrated in mammals [2224]. This developmental feature has also been observed in fish lacking
functional c-myb, resulting in tissue macrophage development in the absence of definitive
hematopoiesis [25].
2.3. Roles of Colony-Stimulating Factor-1 in Teleost Monopoieis
Across all vertebrate species examined to date, the survival, proliferation, differentiation, and
functionality of most cells of the macrophage lineage are governed by the cytokine colony-stimulating
factor-1 (CSF-1 = macrophage-colony-stimulating factor, M-CSF) [2629] through binding to its
cognate receptor (CSF-1R) [30]. In turn, the CSF-1R is expressed almost exclusively on committed
myeloid precursors and derivative macrophage populations [31,32].
From a phylogenetic perspective, it is notable that birds and mammals possess a single, alternatively
spliced CSF-1 gene that produces both membrane-bound and secretory forms of this cytokine [33,34].
Intriguingly, several teleost fish species have been documented to possess two distinct CSF-1 genes,
which at present are not thought to be alternatively spliced [29]. Akin to the mammalian CSF-1, the
fish counterpart is also a proficient macrophage growth and differentiation factor [35,36]. It is,
however, notable that unlike the M2-potentiating roles of the mammalian CSF-1 [37], the functional
work performed on one of the two cyprinid CSF-1 molecules suggests that the teleost counterpart
cytokine differentiates M1-like kidney macrophages, exhibiting upregulated pro-inflammatory
components [36]. This is further supported by the recent findings that a novel soluble goldfish CSF-1R
serves to ablate fish pro-inflammatory responses by reducing available soluble CSF-1 [38]. It will be
Biology 2015, 4 884
interesting to determine whether the respective fish CSF-1 proteins confer the biological roles of
mammalian CSF-1 splice variants, or if these moieties have adopted novel roles in respect to the M1/M2
macrophage paradigm.
The mammalian macrophage lineage cell CSF-1R gene expression increases progressively with cell
development, with committed myeloid precursors possessing the lowest transcript levels of this
receptor, monocytes and immature macrophages expressing significantly greater levels of the CSF-1R,
and mature macrophages exhibiting still greater levels of the CSF-1R transcript [39]. Moreover,
cell-signaling through the CSF-1R not only governs macrophage proliferation, differentiation, and
survival, but also macrophage antimicrobial responses [4043]. It is intriguing that teleosts possess
multiple CSF-1 genes and at least some fish species, such as Fugu, also express two distinct CSF-1R
genes [44]. While it is presently unknown why fish have adopted unique CSF-1/CSF-1R orthologues,
it is compelling to consider that these distinct ligand-receptor combinations may facilitate unique
teleost monopoietic consequences unique from those of mammals. Such deviations have already been
documented for other teleost cytokine-receptor systems, such as the type II IFNs [4548], and render it
more likely that the multiple fish CSF-1s and CSF-1Rs may also possess complex interactions, distinct
from those generally seen across endothermic species.
3. Classical Macrophage Activation
3.1. Molecular Mechanisms of Classical Activation
3.1.1. Type II Interferons
Macrophage activation was first coined by Mackaness in the 1960s after describing antimicrobial
activity of mouse macrophages to secondary infection with intracellular bacteria [49]. Later, this
phenomenon of cellular resistance resulting in the antimicrobial state of macrophages was referred to
as classical activation, or M1, corresponding to the general effector phenotype of these phagocytes
during cell-mediated (Th1) immune responses [50]. M1 polarization predominantly refers to macrophages
activated by the Th1-derived cytokines, and chiefly IFNγ, which is provided transiently early on by
NK cells and more prominently during the adaptive immune response by Th1 helper cells [51,52]. In
addition to IFNγ, co-stimulation by TNFα is necessary to fully induce M1 macrophage microbicidal
activity [53]. It is important to note that M1 polarization can be driven by microbial stimuli alone
through pattern recognition receptor (PRR) activation and in the absence of exogenous cytokine
stimulation. Notably, MyD88-dependant toll-like receptor (TLR) engagement leads to transcription of
TNFα, in addition to IFNβ expression independent of MyD88 TLR signaling [54], enabling
macrophage autocrine stimulation. Indeed, fish species possess extensive arrays of pattern recognition
receptors, both putative mammalian orthologues and fish-specific family members [55], and have been
shown to activate with microbial stimulus in the absence of exogenous cytokine addition.
In fish, type II interferons have been well characterized [56,57]. IFNγ has been sequenced in
fugu [58], rainbow trout [59], zebrafish [60], Atlantic salmon [52], catfish [61], common carp [62],
goldfish [63], Atlantic cod [64], and flounder [65]. Recombinant IFNγ has been shown to elicit STAT1
signaling in head kidney leukocytes of Atlantic salmon [66] and culminates in M1-like macrophages of
Biology 2015, 4 885
goldfish, grass carp, and common carp, indicating a possible functional conservation to the mammalian
counterpart [46,67,68].
Interestingly, certain fish species possess at least two distinct type II IFNs, termed IFNγ and
IFNγ-related (IFNγrel). Recombinant IFNγrel has been shown to activate macrophage reactive
intermediate production [63] in goldfish, and recombinant IFNγrel molecules enhance antiviral activity
in gibuna carp [47]. Furthermore, partially overlapping roles of IFNγ and IFNγrel in knockout
zebrafish [69] indicate a redundancy of IFN function for possible M1 polarization in fish. While these
type II IFNs have been identified in several orders of teleosts, the majority of the functional studies of
fish IFNγ and IFNγrel have been performed in cyprinids. In this respect, the injection of zebrafish
embryos with IFNγ or IFNγrel mRNAs individually elicits comparable immune gene expression
profiles, and combined injections further enhance certain gene expression profiles, possibly owing to
the non-overlapping roles of the respective cytokines [69]. Interestingly, when either the zebrafish
IFNγ or IFNγrel is individually knocked down, zebrafish embryo survival following Escherichia coli
challenge is uncompromised, whereas knock-down of both IFNγs results in significant increases in
infection-induced mortalities [69]. Notably, IFNγ or IFNγrel morpholino knock-downs decrease
embryo survival following Yersinia ruckeri infections, while knock-down of both IFNs further
compounds embryo mortalities. Presumably, IFNγ and IFNγrel induce both overlapping and distinct
antimicrobial mechanisms.
We have documented that the recombinant forms of the goldfish (rg) IFNγ and rgIFNγrel have very
different effects on kidney macrophage functions [63]. While rgIFNγ evokes long-lasting reactive
oxygen intermediate (ROI) priming, rgIFNγrel-induced ROI priming is short-lived and is followed by
unresponsiveness of the stimulated phagocytes to ROI induction by other M1 cytokines (rgIFNγ or
rgTNFα2). Moreover, rgIFNγrel appears to be a much more potent inducer of macrophage phagocytosis
and nitric oxide (NO) production than rgIFNγ, and the two cytokines induce distinct expression levels
of key macrophage immune genes [46,70,71]. Finally, it would appear that the downstream signaling
mechanisms employed by these type II IFNs are also distinct, with IFNγ more closely resembling the
mammalian IFNγ signaling.
Further confirmation of this functional dichotomy between the fish type II IFNs is warranted using
in vivo and other in vitro fish models. Notably, the zebrafish IFNγrel has recently been shown to elicit
more robust pro-inflammatory gene expression than IFNγ in larvae microinjected with respective IFN
expression constructs [72]. Interestingly, these zebrafish IFNγrel-mediated effects were dependent on the
myeloid transcription factor SP1, underlying the specificity of this cytokine for macrophage lineage cells.
In comparison to other vertebrates that possess a single IFNγ receptor 1 (IFNGR1), it is particularly
notable that certain teleosts possess two distinct IFN gamma-receptor binding chains (IFNGR1-1 and
IFNGR1-2) that confer distinct but poorly understood interactions with the IFNγ and IFNγrel
molecules of these respective species [45,48,63]. Together, it would appear that fish have adopted very
unique strategies with respect to the principal M1 macrophage activating cytokine system. It will be
interesting to further examine how these respective type II IFNs participate in teleost macrophage
functional polarization.
Biology 2015, 4 886
3.1.2. Tumor Necrosis Factor-Alpha
Akin to many fish cytokines, multiple isoforms of the key inflammatory cytokine, tumor necrosis
factor alpha (TNFα), have been documented across numerous fish species [7384]. In goldfish and
carp, TNFα1 and TNFα2 have been implicated as having pro-inflammatory roles, and enhancing
pro-inflammatory gene expression, as well as phagocytic, reactive oxygen, and nitrogen intermediate
production, akin to the pro-M1 roles of the mammalian TNFα [59,74]. Recently, transgenic zebrafish
bearing a fluorescent TNFα reporter were shown to recruit a subset of macrophages during both
aseptic wounding and wounding with E. coli inoculation, where TNFα expression occurred in the
presence of bacteria, thus underlining the microbial activation and M1 polarization of macrophages [85].
TNFα3 expression is increased following stimulation of HK macrophages with PAMPs, and rTNFα3
increases production of a number of pro-inflammatory cytokines, although the functional relevance of
this gene has yet to be fully elucidated [84].
3.1.3. Granulocyte-Macrophage Colony-Stimulating Factor
Granulocyte macrophage colony-stimulating factor (GM-CSF) has been added to the list of M1
stimuli in mammals [86]. GM-CSF aids in the survival, proliferation, and differentiation during
myelopoiesis through receptor chains CSF2Rα and CSF2Rβ [87]. GM-CSF has not yet been confirmed
in teleosts, although recent discovery of a GM-CSF-like molecule in the elephant shark may infer
possible existence in bony fish [88]. Moreover, cDNA and gene sequences of numerous teleost fish
CSF2Rβ are available on GenBank, further substantiating the possibility of a teleost GM-CSF,
although it should be noted that in mammals this receptor chain is shared by GM-CSF, IL-3, and
IL-5 [89], inferring a possibility of disparate ligand interactions in lower vertebrates.
3.2. M1 Macrophage Cytokine Profiles
Stimulation and polarization of macrophages to an M1 subtype results in the production of large
amounts of TNF, IL-1β, IL-6, IL-12 IL-15, and IL-23, for further activation of proximal cell types,
aiding chemotaxis of inflammatory leukocytes (i.e., neutrophils and monocytes), and influencing Th1
polarization, which in turn amplifies classical activation [90]. This main M1 cytokine repertoire is
present in fish, and has been demonstrated to up-regulate following microbial challenge, as well as
following stimulation with M1-inducing stimuli, akin to that of mammals [10,67,74,91]. The detailed
repertoire of cytokine networks, including Th1 cytokine networks of fish, has been comprehensively
reviewed [57].
4. Antimicrobial Roles of Teleost M1 Macrophages
4.1. Respiratory Burst Response
The destruction of internalized microorganisms is key to the innate immune response. It is well
established that fish phagocytes, including macrophages, generate reactive oxygen intermediates as an
antimicrobial defense, similar to mammalian phagocytes. During the macrophage respiratory burst, an
assembly of multicomponent enzyme nicotinamide adenine dinucleotide phosphate (NADPH) subunits
Biology 2015, 4 887
at the plasma membrane results in the transfer of electrons from NADPH to molecular oxygen,
resulting in the superoxide anion [92]. Upon formation, superoxide rapidly converts to reactive oxygen
species (ROS) hydrogen peroxide (H2O2), hydroxyl radical (OH·), and hyperchlorous acid (HOCl),
which efficiently kill microorganisms [93,94] (El-Benna et al., 2008). The NADPH oxidase complex is
made up of six individual subunits that are segregated in resting cells, including the cytosolic components
p40phox (phagosome oxidase), p47phox, p67phox, and a guanosine triphosphatase (GTPase) Rac 1 or
Rac 2 migrates to membrane-associated subunits gp91phox (also known as Nox2) and p22phox [95100],
all of which have been cloned in several fish species [101]. Despite low sequence homology between fish
and mammals, the functional sites of NADPH-oxidase are highly conserved [102,103]. ROI production by
teleost macrophages has been observed following stimulation with pathogen-associated molecular
patterns (PAMPs) [4,104107], fish pathogens [108110], and recombinant cytokines such as
TNFα [74,80,111], IFNγ [46,59,67] and CSF-1 (MCSF). Notably and as described above, this is in
contrast to the mammalian CSF-1, which is considered to be an M2 stimulus, opposite to GM-CSF [86].
The importance of respiratory burst to host protection is underlined by the various pathogens that can
effectively ablate or withstand toxic oxygen intermediates. For example, Edwardsiella tarda, a natural
pathogen of various economically important fish and the etiological agent of edwardsiellosis, has several
strategies to combat ROI production of teleost macrophages, reviewed by [5].
4.2. Nitric Oxide Response
Akin to superoxide production, the inducible nitric oxide (NO) system of teleost macrophages is
well conserved compared to those described in mammals. Classically activated macrophages are
distinguished by the expression of inducible nitric oxide synthase (iNOS/NOS2) that catalyzes the
conversion of L-arginine to L-citruline, resulting in production of NO, a potent antimicrobial
compound [112,113]. Simultaneous production of superoxide and NO intermediates can also form
peroxynitrite (ONOO), which additionally serves potent antiparasitic/antimicrobial functions [114,115].
Indeed, iNOS is a prototypical marker of M1 activation in macrophages that is readily up-regulated in
response to IFNγ, TNFα, and microbial compounds (e.g., LPS) [51].
The fish iNOS has been characterized with marked similarity to the mammalian enzyme counterpart,
possessing the putative binding sites for heme, tetrahydrobiopterin, calmodulin, flavine mononucleotide,
flavine adenine dinucleotide, and NADPH, suggesting cofactor conservation and function [81]. First
identified as a partial sequence in goldfish [116], iNOS has been identified and characterized in
carp [81], rainbow trout [117], zebrafish [118], and turbot [119]. The gene expression of fish
macrophage iNOS coincides with cellular NO production, and has been demonstrated to increase
following exposure to PAMPS or a microbial stimulus [6,81,120] and pro-inflammatory cytokine
stimulus [35,46,67,74,80,83,121], as well as cleaved transferrin products [122]. The importance of
iNOS in host protection has been implicated in the control of many varieties of fish pathogens, including
viral hemorrhagic septicemia virus (VHSV), Aeromonas salmonocida, Renibacterium salmoninarum,
Yersinia ruckeri [123125], and Mycobacterium marinum [91,126128].
Biology 2015, 4 888
4.3. Phagolysosome Fusion
Following pathogen uptake, the phagosome undergoes various maturation steps that are an
important process to destroy the internalized microbe, which culminates in phagolysosome formation.
Both M1- and M2-type macrophages rapidly form phagolysosomes in order to degrade internalized
material. Interestingly, recent reports of M1-stimulated macrophages show relatively neutral pH
following the internalization of zymosan, in contrast with M2-driven macrophages that rapidly acidify
their phagosomes [129]. This is presumably due to the proton consumption during superoxide production,
a feature that may also aid in preserving antigen for presentation by decreasing the degradative capacity,
as is seen in mammalian dendritic cells [130]. Not surprisingly, fish monocytes and macrophages
demonstrate the ability to undergo phagolysosomal fusion [4]. Additional work is necessary to elucidate
the importance of phagosome maturation of fish macrophages in host immune defenses with regard to
macrophage polarization.
4.4. Nutrient Deprivation
4.4.1. Solute Carrier 11 Member 1
Solute carrier 11 member 1, Slc11a1 (formally known as natural resistance-associated macrophage
protein 1, NRAMP1), is a divalent metal ion transporter, present in the late endosomes and lysosomes
of professional phagocytes [131]. Transcriptional regulation of Slc11a1 is induced by hypoxia-inducible
factor alpha (HIFα), predominantly active in M1 macrophages in response to microbial or pro-inflammatory
cytokine stimuli [132,133]. Mutations in the Slc11a1 allele confer susceptibility to infection with
intracellular pathogens, including Salmonella, Leishmania, and Mycobacteria [134]. Although there is
a clear role of NRAMP1 in innate immunity, the precise antimicrobial mechanisms conferred by this
ion channel remain elusive. Evidence of movement of Fe2+ into the phagosome in RAW264.7, a
NRAMP1-deficient cell line, shows iron transport into phagosomes, which may aid in catalyzing ROS
and limiting bacterial growth [135]. Contrary to these findings, reports of metal movement out of the
phagosome, which is proposed to limit growth by restricting nutrients, has also been observed [136,137],
leading to the proposal of Slc11a1 as a pH-dependent bidirectional transporter [138].
NRAMP1 has been cloned in a number of fish species, including carp [139], fugu [140], channel
catfish [141], rainbow trout [142], turbot [143], sea bream [144], Japanese flounder [145], and striped
bass [144]. Although orthologs of Slc11 have been identified in several fish species, the phylogeny,
sequence identities, and expression patterns of these teleost NRAMP orthologs do not clearly indicate
whether these primordial counterparts are specifically orthologous to Slc11a1 or Slc11a2. Despite this,
several studies have indicated that the fish NRAMP counterparts are intimately involved in innate
immune responses against pathogens.
Teleost fish appear to possess multiple Slc11 orthologs per genome including two fugu Slc11a
genes [140] and three catfish Slc11a orthologs [141]. Moreover, while the phylogenetic relationships
between the mammalian and fish Slc11 genes remain to be defined, the teleost NRAMP counterparts are
indeed directly involved in antimicrobial responses against fish pathogens such as V. anguillarum [144],
E. ictaluri [146], and M. marinum [91,147].
Biology 2015, 4 889
4.4.2. Ferroportin
The first identification of ferroportin (also known as metal-transporting protein-1 or iron-regulated
transporter-1) was in zebrafish [148] and mice [149]. Activation with IFNγ, LPS, or intracellular
pathogens has been shown to enhance ferroportin expression, which reduces intracellular iron, thereby
depriving the pathogen of a necessary enzymatic cofactor [150152], indicating an important role of
this protein in M1-activated macrophages. The overexpression of ferroportin has been shown to disrupt
the intracellular growth of Mycobacteria and Salmonella, and ferroportin-deficient mice are more
susceptible to intracellular pathogens [153]. The control of ferroportin is predominantly mediated by
hepcidin (up-regulated in mammals by IL-6), which binds to ferroportin and promotes internalization
and degradation in lysosomes, diminishing the release of iron from macrophages [154].
Ferroportin has been identified in zebrafish [148] and turbot [155], showing correlated expression
levels during V. anguillarum infections. Interestingly, the zebrafish mutant weissherbst is a hyperchromatic
blood mutant with a mutation in functional ferroportin [148], which may be valuable in further
elucidating the effects of ferroportin in teleosts.
4.4.3. Indoleamine 2,3-Dioxygenase
Indoleamine 2,3-dioxygenase (IDO), or tryptophan 2,3 dioxygenase (IDO), is a macrophage enzyme
that catalyzes the degradation of tryptophan, and is up-regulated in IFNγ-stimulated macrophages [156].
M1 macrophage IDO expression and activation of this immunoregulatory and antimicrobial mechanism is
intimately linked to cell stimulation by IFNγ [157]. IDO tryptophan degradation results in the
production of a panel of immunoregulatory metabolites, collectively referred to as kynurenins [158]. In
turn, the IDO-derived kynurenins promote immunotolerance and suppress excessive proliferation of
activated cytotoxic leukocytes. Presently, the mechanisms governing M1 macrophage IDO are poorly
defined; however, it is thought that the IFNγ-mediated activation of IDO serves as a nutrient
deprivation mechanism by reducing and preventing tryptophan supply to intracellular or locally
detected pathogens [159162].
Bony fish IDO orthologs appear to possess very low tryptophan-degradative enzymatic efficiencies
compared to the mammalian IDOs [163], suggesting the presence of alternative fish IDO substrates,
and were aptly renamed as proto-IDOs. Interestingly, marsupials possess both IDO and proto-IDO, in
tandem on a single chromosome, whereas only proto-IDOs are found in fish, amphibians, and
chickens, implying that the mammalian IDO arose from a gene duplication event of the proto-IDO.
Notably, goldfish macrophages infected with M. marinum significantly up-regulate their proto-IDO
gene expression, where live M. marinum induces substantially greater proto-IDO transcript levels than
the heat-killed bacteria [91]. This suggests that the fish proto-IDO expression is possibly advantageous
to M. marinum survival in macrophages, and may indicate that this fish proto-IDO serves to dampen
immune cell bystander responses, akin to its mammalian counterpart. A summary of mammalian and
teleost M1 macrophage function is depicted in Figure 1.
Biology 2015, 4 890
Figure 1. (A) Activation states of mammalian macrophages. Stimulation with microbial
patterns in addition to type II interferons, TNFα or GM-CSF, leads to increased antimicrobial
responses of macrophages, pro-inflammatory cytokine production, and nutrient deprivation
in accordance with an M1 activation profile. Alternative activation of macrophages by
cAMP IL4 and/or 13 ligands results in increased arginase expression and activity. Stimulation
with immune complexes and lipopolysaccharides (LPS) generates a unique IL-10high
IL-12l°w M2b phenotype. IL-10 or glucocorticoid-activated macrophages drive regulatory
function, inhibiting pro-inflammatory cytokine production, antimicrobial activity, and
driving IL-10 expression; (B) Activation states of teleost macrophages. Stimulation with
microbial patterns, type II interferons, TNFα isoforms, or CSF-1 leads to increased
antimicrobial responses similar to an M1 phenotype. Alternative activation of teleost
macrophages can be achieved by cAMP stimulation. Immune complex, IL-10, and
glucocorticoids can deactivate teleost macrophages. Question marks refer to molecules that
are present in teleosts, but have yet to be linked to alterations in macrophage plasticity.
5. Alternative Teleost Macrophage Activation
5.1. Interleukin-4/13
M2 macrophages are generally considered as being antagonists to M1 macrophages, having
anti-inflammatory or pro-healing functions. In mammals, the best characterized M2 stimuli are
IL-4 and IL-13 (sometimes called M2a), produced by Th2 cells, eosinophils, basophils, NKT cells, and
macrophages themselves [164]. The recognition of IL-4 by receptor pairs IL-4Rα combined with either
IL4γc or IL-13α1 chains results in signaling through JAK1 and JAK3, leading to STAT6 activation and
Biology 2015, 4 891
nuclear translocation. IL-13 can also bind the IL13Rα2 chain, which has not been fully characterized [165].
Mouse and human macrophage proteins up-regulated in response to these M2 stimuli include:
transglutaminase 2 (TGM2), macrophage mannose receptor (MMR1/MRC1), cholesterol hydroxylase
CH25H, prostaglandin-endopreoxide synthase (PTGS1), transcription factors IRF4 and Kruppel-like
factor 4 (KLF4), and signaling molecules with cytokine-inducible SRC homology 2 (SH2) containing
protein (CISH) and suppressor of cytokine signaling 1 (SOCS1), all of which are present in teleost
fish, although have yet to be described as M2 markers [105,166170].
To date, at least two genes have been identified in fish that share homology with both the mammalian
IL-4 and IL-13 cytokines (IL-4/13A and IL-4/13B) [171], although genome/gene duplication events
have resulted in variable numbers of these gene copies in different fish species [172]. Two paralogs of
IL-4Rα, IL-13Rα1, and IL-13Rα2 have been identified in rainbow trout and zebrafish [173,174].
Although there is no information on IL-4/IL13 alternative activation of fish macrophages, recombinant
IL-4/13A has been shown to bind IL-13Rα [174], and was shown to support B and T cell expansion,
indicative of a conserved role for this cytokine in fish Th2 adaptive immunity [175,176]. It will be
interesting to learn whether the IL-4/13 cytokines serve to establish M2 macrophage phenotypes in
lower vertebrates, including fish.
5.2. Arginase
A major bifurcation of M1 and M2 function involves the metabolism of L-arginine. Whereas iNOS
of M1 macrophages converts L-arginine to L-citruline and NO, arginase, a putative marker of M2
macrophages, converts L-arginine to L-ornithine and urea [177180]. The repair phenotype of M2
macrophages is predicated by the production of L-ornithine, which is a precursor for polyamines and
proline components of collagen that are important for tissue repair [181]. Interestingly, byproducts of
either iNOS or arginase pathways inhibit the reciprocal enzymes, thus stabilizing the M1 or M2
macrophage polarization states, respectively [182].
Mammals possess two arginase isoforms including arginase-1, located in the cytosol and induced by
IL-4 and IL-13 [183], and arginase-2, associated with the mitochondria and up-regulated by IL-10 and
LPS [184]. In fish, arginase-1 and arginase-2 were first identified in rainbow trout, phylogenetically
clustering with the respective mammalian orthologues [185]. Arginase activity has been demonstrated
in cyclic adenosine monophosphate (cAMP)-stimulated murine macrophages which increased
intracellularly during the IL-13 signal transduction [186]. Similarly, common carp macrophages
stimulated with cAMP show increased arginase activity and specific inhibition of urea production with
NG-hydroxy-L-arginine, suggesting an evolutionary conservation of polarized macrophages in lower
vertebrates [6]. Indeed, work using the carp kidney macrophage model has established that, like
mammalian cells, classically polarized fish macrophages are marked by high iNOS gene expression,
whereas alternatively polarized macrophages are distinguishable by highly up-regulated arginase gene
expression [6,10,120]. Intriguingly, in contrast to the mammalian M2 macrophage arginase-1 expression,
carp alternative macrophage activation coincides with increased arginase-2 transcript levels [6]. For
excellent comprehensive reviews of alternative fish macrophage polarization and the fish arginase
responses, refer to recent reviews [187,188].
Biology 2015, 4 892
5.3. Immune Complex and Lipopolysaccharides
Repeated stimulation of M1-like macrophages by inflammatory stimuli has been documented to
result in the adoption of M2-like macrophage phenotypes and unresponsiveness to subsequent
inflammatory cues. For example, mammalian M2b or type-2 macrophages are generated in response to
immune complexes (IC) and Gram-negative bacteria lipopolysaccharides (LPS), resulting in lower IL-12
and higher IL-10 production [164]. This is thought to be an essential link in dampening early M1
activity during inflammation, and thus promoting tissue remodeling and regeneration [189]. Carp blood
parasite infection studies additionally demonstrated this phenomenon, showing that Trypanoplasma borreli
immune complex formation generated distinctly activated carp macrophage subsets, promoting the
resolution of inflammation and parasite clearance [10].
5.4. Glucocorticoids and Interleukin-10
Deactivation of macrophages by glucocorticoids (GCs) and IL-10 has also been described to
culminate in the unique M2c macrophage activation state, otherwise referred to as regulatory
macrophages. The diffusion of GCs across the plasma membrane leads to interaction with the
glucocorticoid receptor (GCR), which results in nuclear translocation and direct transcriptional
up-regulation of some immune genes and down-regulation of others, resulting in macrophage
phenotypes and transcriptional profiles distinct from those observed following IL-4 macrophage
stimulation [190,191]. For example, GC stimulation of macrophages antagonizes classically activated
macrophage functions, including down-regulation of inflammatory cytokines and dampening of reactive
intermediate production. Cortisol has been shown to immunosuppress fish and increase their
susceptibility to diseases [192194]. Interestingly, cortisol has been shown to be a strong inhibitor of NO
production in goldfish macrophages [195]. Furthermore, treatment of a rainbow trout cell line with a
combination of pro-inflammatory stimuli and cortisol results in heightened expression of interleukin-10,
suggesting that the cortisol treatment overrides the induction of pro-inflammatory responses [8].
In mammals, IL-10 signals through IL10R1 and IL10R2, leading to activation of STAT3 and
inhibition of pro-inflammatory cytokine expression and, thus, Th1 and M1 functions [196]. IL-10 is
produced by virtually all leukocytes and is generated by macrophages in response to TLR engagement,
GCs, and C-type lectin signaling [164]. An IL-10R1 has been identified in zebrafish, goldfish, and
grass carp [197,198], as well as IL-10R2 in rainbow trout [199]. Notably, the goldfish IL-10 has been
shown to down-regulate IFNγ stimulation of the ROI response and inflammatory gene expression of
goldfish monocytes (Grayfer et al., 2011b) [91], demonstrating an evolutionarily conserved regulatory
role for this fish cytokine. Alternatively activated macrophage subtypes of mammals and teleosts are
depicted in Figure 1.
6. Conclusions
Akin to their mammalian counterparts, macrophages of teleost fish exhibit a plethora of functional
roles including those pertaining to homeostasis, as well as host immune defenses, and are largely
governed by their respective tissue niches and microenvironments. Perturbations in these homeostatic
environments by pathogens or injury polarize resident macrophage populations aptly towards the
Biology 2015, 4 893
appropriate functionalities, with the sum of the cell types altering the physiology of the hosts towards
an M1 inflammatory type, or the M2 resolution and repair types. Although these broad functional
states have been demonstrated in fish macrophages, further research into the molecular regulation by
both mammalian orthologs and novel fish-specific molecules will further the understanding of teleost
macrophage functional regulation. Undoubtedly, the growing genetic resources of teleost animal
models combined with transcriptomic/proteonomic/metabolomic technologies and live-imaging techniques
of differentially activated teleost macrophages will be integral in deciphering the intricacies of
macrophage functional regulation in teleosts.
Acknowledgments
This work was supported by Natural Sciences and Engineering Council of Canada (NSERC; grant #
RGPIN-2014-06395) to Miodrag Belosevic. Jordan W. Hodgkinson was supported by an NSERC
PGS-D doctoral scholarship. Leon Grayfer would like to thank the George Washington University,
Department of Biological Sciences for financial support in the form of laboratory start-up funds.
Author Contributions
Jordan W. Hodgkinson, Leon Grayfer and Miodrag Belosevic conceived and wrote this manuscript.
Conflicts of Interest
The authors declare no conflict of interest.
Abbreviations
Arg-1, arginase-1; AGM, aorta-gonad-mesonephros; CSF, colony stimulating factor; CSF-1R,
colony stimulating factor-1 receptor; GM-CSF, granulocyte-macrophage colony-stimulating factor;
EMP, erythromyeloid progenitors; GC, glucocorticoids; HSCs, hematopoietic stem cells; IDO,
indoleamine 2,3-dioxygenase; IFN, interferon; IL, interleukin; M-CSF, macrophage-colony-stimulating
factor; NK, natural killer cell; NO, nitric oxide; NRAMP1, natural resistance-associated macrophage
protein 1; PAMPs, pathogen associated molecular patterns; PMA, phorbol myrystate acetate; PRR, pattern
recognition receptor; rg, recombinant goldfish; RNI, reactive nitrogen intermediates; ROI, reactive
oxygen intermediates; Slc11a1, Solute carrier 11 member 1; Stat, signal transducer of activation
transcription factor; TGF, transforming growth factor; TLR, toll-like receptor; TNF, tumor necrosis factor.
References
1. Metchnikoff, E. Immunity in Infective Diseases; Cambridge University Press: Cambridge,
UK, 1905.
2. Zhou, D.; Huang, C.; Lin, Z.; Zhan, S.; Kong, L.; Fang, C.; Li, J. Macrophage polarization and
function with emphasis on the evolving roles of coordinated regulation of cellular signaling
pathways. Cell. Signal. 2014, 26, 192197.
Biology 2015, 4 894
3. Neumann, N.F.; Stafford, J.L.; Belosevic, M. Biochemical and functional characterisation of
macrophage stimulating factors secreted by mitogen-induced goldfish kidney leucocytes.
Fish Shellfish Immunol. 2000, 10, 167186.
4. Rieger, A.M.; Hall, B.E.; Barreda, D.R. Macrophage activation differentially modulates particle
binding, phagocytosis and downstream antimicrobial mechanisms. Dev. Comp. Immunol. 2010,
34, 11441159.
5. Grayfer, L.; Hodgkinson, J.W.; Belosevic, M. Antimicrobial responses of teleost phagocytes and
innate immune evasion strategies of intracellular bacteria. Dev. Comp. Immunol. 2014, 43, 223242.
6. Joerink, M.; Savelkoul, H.F.J.; Wiegertjes, G.F. Evolutionary conservation of alternative activation
of macrophages: Structural and functional characterization of arginase 1 and 2 in carp (Cyprinus
carpio L.). Mol. Immunol. 2006, 43, 11161128.
7. Takizawa, F.; Koppang, E.O.; Ohtani, M.; Nakanishi, T.; Hashimoto, K.; Fischer, U.; Dijkstra, J.M.
Constitutive high expression of interleukin-4/13A and GATA-3 in gill and skin of salmonid
fishes suggests that these tissues form Th2-skewed immune environments. Mol. Immunol. 2011,
48, 13601368.
8. Castro, R.; Zou, J.; Secombes, C.J.; Martin, S.A.M. Cortisol modulates the induction of
inflammatory gene expression in a rainbow trout macrophage cell line. Fish Shellfish Immunol.
2011, 30, 215223.
9. Grayfer, L.; Hodgkinson, J.W.; Hitchen, S.J.; Belosevic, M. Characterization and functional
analysis of goldfish (Carassius auratus L.) interleukin-10. Mol. Immunol. 2011, 48, 563571.
10. Joerink, M.; Forlenza, M.; Ribeiro, C.M.S.; de Vries, B.J.; Savelkoul, H.F.J.; Wiegertjes, G.F.
Differential macrophage polarisation during parasitic infections in common carp (Cyprinus
carpio L.). Fish Shellfish Immunol. 2006, 21, 561571.
11. Glasauer, S.M.K.; Neuhauss, S.C.F. Whole-genome duplication in teleost fishes and its
evolutionary consequences. Mol. Genet. Genom. 2014, 289, 10451060.
12. Bertrand, J.Y.; Kim, A.D.; Violette, E.P.; Stachura, D.L.; Cisson, J.L.; Traver, D. Definitive
hematopoiesis initiates through a committed erythromyeloid progenitor in the zebrafish embryo.
Development 2007, 134, 41474156.
13. Chen, A.T.; Zon, L.I. Zebrafish blood stem cells. J. Cell. Biochem. 2009, 108, 3542.
14. Lieschke, G.J.; Oates, A.C.; Paw, B.H.; Thompson, M.A.; Hall, N.E.; Ward, A.C.; Ho, R.K.;
Zon, L.I.; Layton, J.E. Zebrafish SPI-1 (PU.1) marks a site of myeloid development independent
of primitive erythropoiesis: Implications for axial patterning. Dev. Biol. 2002, 246, 274295.
15. Willett, C.E.; Cortes, A.; Zuasti, A.; Zapata, A.G. Early hematopoiesis and developing lymphoid
organs in the zebrafish. Dev. Dyn. 1999, 214, 323336.
16. Carroll, K.J.; North, T.E. Oceans of opportunity: Exploring vertebrate hematopoiesis in zebrafish.
Exp. Hematol. 2014, 42, 684696.
17. Kobayashi, I.; Kuniyoshi, S.; Saito, K.; Moritomo, T.; Takahashi, T.; Nakanishi, T. Long-term
hematopoietic reconstitution by transplantation of kidney hematopoietic stem cells in lethally
irradiated clonal ginbuna crucian carp (Carassius auratus langsdorfii). Dev. Comp. Immunol.
2008, 32, 957965.
Biology 2015, 4 895
18. Traver, D.; Winzeler, A.; Stern, H.M.; Mayhall, E.A.; Langenau, D.M.; Kutok, J.L.; Look, A.T.;
Zon, L.I. Effects of lethal irradiation in zebrafish and rescue by hematopoietic cell transplantation.
Blood 2004, 104, 12981305.
19. Hanington, P.C.; Tam, J.; Katzenback, B.A.; Hitchen, S.J.; Barreda, D.R.; Belosevic, M.
Development of macrophages of cyprinid fish. Dev. Comp. Immunol. 2009, 33, 411429.
20. Katzenback A.B.; Katakura, F.; Belosevic, M. Regulation of teleost macrophage and neutrophil
cell development by growth factors and transcription factors. In New Advances and Contributions to
Fish Biology; Turker, H., Ed.; InTech: Rijeka, Croatia, 2012.
21. Van Furth, R.; Cohn, Z.A.; Hirsch, J.G.; Humphrey, J.H.; Spector, W.G.; Langevoort, H.L.
Mononuclear phagocytic system: New classification of macrophages, monocytes and of their cell
line. Bull. World Health Organ. 1972, 47, 651658.
22. Ginhoux, F.; Jung, S. Monocytes and macrophages: Developmental pathways and tissue
homeostasis. Nat. Rev. Immunol. 2014, 14, 392404.
23. Hashimoto, D.; Chow, A.; Noizat, C.; Teo, P.; Beasley, M.B.; Leboeuf, M.; Becker, C.D.; See, P.;
Price, J.; Lucas, D.; et al. Tissue-resident macrophages self-maintain locally throughout adult life
with minimal contribution from circulating monocytes. Immunity 2013, 38, 792804.
24. Yona, S.; Kim, K.W.; Wolf, Y.; Mildner, A.; Varol, D.; Breker, M.; Strauss-Ayali, D.; Viukov, S.;
Guilliams, M.; Misharin, A.; et al. Fate mapping reveals origins and dynamics of monocytes and
tissue macrophages under homeostasis. Immunity 2013, 38, 7991.
25. Soza-Ried, C.; Hess, I.; Netuschil, N.; Schorpp, M.; Boehm, T. Essential role of c-myb in
definitive hematopoiesis is evolutionarily conserved. Proc. Natl. Acad. Sci. USA 2010, 107,
1730417308.
26. Garceau, V.; Smith, J.; Paton, I.R.; Davey, M.; Fares, M.A.; Sester, D.P.; Burt, D.W.; Hume, D.A.
Pivotal Advance: Avian colony-stimulating factor 1 (CSF-1), interleukin-34 (IL-34), and CSF-1
receptor genes and gene products. J. Leukoc. Biol. 2010, 87, 753764.
27. Hanington, P.C.; Wang, T.; Secombes, C.J.; Belosevic, M. Growth factors of lower vertebrates:
Characterization of goldfish (Carassius auratus L.) macrophage colony-stimulating factor-1.
J. Biol. Chem. 2007, 282, 3186531872.
28. Pixley, F.J.; Stanley, E.R. CSF-1 regulation of the wandering macrophage: Complexity in action.
Trends Cell Biol. 2004, 14, 628638.
29. Wang, T.; Hanington, P.C.; Belosevic, M.; Secombes, C.J. Two macrophage colony-stimulating
factor genes exist in fish that differ in gene organization and are differentially expressed.
J. Immunol. 2008, 181, 33103322.
30. Dai, X.-M.; Ryan, G.R.; Hapel, A.J.; Dominguez, M.G.; Russell, R.G.; Kapp, S.; Sylvestre, V.;
Stanley, E.R. Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results
in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies,
and reproductive defects. Blood 2002, 99, 111120.
31. Guilbert, L.J.; Stanley, E.R. Specific interaction of murine colony-stimulating factor with
mononuclear phagocytic cells. J. Cell Biol. 1980, 85, 153159.
32. Lichanska, A.M.; Browne, C.M.; Henkel, G.W.; Murphy, K.M.; Ostrowski, M.C.; McKercher, S.R.;
Maki, R.A.; Hume, D.A. Differentiation of the mononuclear phagocyte system during mouse
embryogenesis: The role of transcription factor PU.1. Blood 1999, 94, 127138.
Biology 2015, 4 896
33. Manos, M.M. Expression and processing of a recombinant human macrophage colony-stimulating
factor in mouse cells. Mol. Cell. Biol. 1988, 8, 50355039.
34. Rettenmier, C.W.; Roussel, M.F. Differential processing of colony-stimulating factor 1 precursors
encoded by two human cDNAs. Mol. Cell. Biol. 1988, 8, 50265034.
35. Hanington, P.C.; Belosevic, M. Interleukin-6 family cytokine M17 induces differentiation and
nitric oxide response of goldfish (Carassius auratus L.) macrophages. Dev. Comp. Immunol.
2007, 31, 817829.
36. Grayfer, L.; Hanington, P.C.; Belosevic, M. Macrophage colony-stimulating factor (CSF-1)
induces pro-inflammatory gene expression and enhances antimicrobial responses of goldfish
(Carassius auratus L.) macrophages. Fish Shellfish Immunol. 2009, 26, 406413.
37. Hamilton, T.A.; Zhao, C.; Pavicic, P.G.; Datta, S. Myeloid colony-stimulating factors as
regulators of Macrophage polarization. Front. Immunol. 2014, doi:10.3389/fimmu.2014.00554.
38. Rieger, A.M.; Hanington, P.C.; Belosevic, M.; Barreda, D.R. Control of CSF-1 induced
inflammation in teleost fish by a soluble form of the CSF-1 receptor. Fish Shellfish Immunol.
2014, 41, 4551.
39. Stanley, E.R.; Berg, K.L.; Einstein, D.B.; Lee, P.S.; Pixley, F.J.; Wang, Y.; Yeung, Y.G. Biology
and action of colonyStimulating factor-1. Mol. Reprod. Dev. 1997, 46, 410.
40. Bober, L.A.; Grace, M.J.; Pugliese-Sivo, C.; Rojas-Triana, A.; Sullivan, L.M.; Narula, S.K. The
effects of colony stimulating factors on human monocyte cell function. Int. J. Immunopharmacol.
1995, 17, 385392.
41. Karbassi, A.; Becker, J.M.; Foster, J.S.; Moore, R.N. Enhanced killing of Candida albicans by
murine macrophages treated with macrophage colony-stimulating factor: Evidence for augmented
expression of mannose receptors. J. Immunol. 1987, 139, 417421.
42. Munn, D.H.; Cheung, N.K. Antibody-independent phagocytosis of tumor cells by human
monocyte-derived macrophages cultured in recombinant macrophage colony-stimulating factor.
Cancer Immunol. Immunother. CII 1995, 41, 4652.
43. Sweet, M.J.; Hume, D.A. CSF-1 as a regulator of macrophage activation and immune responses.
Arch. Immunol. Ther. Exp. 2003, 51, 169177.
44. Williams, H.; Brenner, S.; Venkatesh, B. Identification and analysis of additional copies of the
platelet-derived growth factor receptor and colony stimulating factor 1 receptor genes in fugu.
Gene 2002, 295, 255264.
45. Aggad, D.; Stein, C.; Sieger, D.; Mazel, M.; Boudinot, P.; Herbomel, P.; Levraud, J.P.; Lutfalla, G.;
Leptin, M. In vivo analysis of IFN-γ1 and IFN-γ2 signaling in zebrafish. J. Immunol. 2010, 185,
67746782.
46. Grayfer, L.; Belosevic, M. Molecular characterization, expression and functional analysis of
goldfish (Carassius aurutus L.) interferon gamma. Dev. Comp. Immunol. 2009, 33, 235246.
47. Shibasaki, Y.; Yabu, T.; Araki, K.; Mano, N.; Shiba, H.; Moritomo, T.; Nakanishi, T. Peculiar
monomeric interferon gammas, IFNγrel 1 and IFNγrel 2, in ginbuna crucian carp. FEBS J. 2014,
281, 10461056.
Biology 2015, 4 897
48. Yabu, T.; Toda, H.; Shibasaki, Y.; Araki, K.; Yamashita, M.; Anzai, H.; Mano, N.; Masuhiro, Y.;
Hanazawa, S.; Shiba, H.; et al. Antiviral protection mechanisms mediated by ginbuna crucian
carp interferon gamma isoforms 1 and 2 through two distinct interferon γ-receptors. J. Biochem.
2011, 150, 635648.
49. Mackaness, G.B. Cellular resistance to infection. J. Exp. Med. 1962, 116, 381406.
50. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 2003, 3, 2335.
51. Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev.
Immunol. 2008, 8, 958969.
52. Robertsen, B. The interferon system of teleost fish. Fish Shellfish Immunol. 2006, 20, 172191.
53. O’Shea, J.J.; Murray, P.J. Cytokine signaling modules in inflammatory responses. Immunity
2008, 28, 477487.
54. Yamamoto, M.; Sato, S.; Hemmi, H.; Hoshino, K.; Kaisho, T.; Sanjo, H.; Takeuchi, O.;
Sugiyama, M.; Okabe, M.; Takeda, K.; et al. Role of adaptor TRIF in the MyD88-independent
toll-like receptor signaling pathway. Science 2003, 301, 640643.
55. Van der Vaart, M.; Spaink, H.P.; Meijer, A.H. Pathogen recognition and activation of the innate
immune response in zebrafish. Adv. Hematol. 2012, doi:10.1155/2012/159807.
56. Secombes, C.J.; Wang, T.; Bird, S. The interleukins of fish. Dev. Comp. Immunol. 2011, 35,
13361345.
57. Wang, T.; Secombes, C.J. The cytokine networks of adaptive immunity in fish. Fish Shellfish
Immunol. 2013, 35, 17031718.
58. Zou, J.; Yoshiura, Y.; Dijkstra, J.M.; Sakai, M.; Ototake, M.; Secombes, C. Identification of an
interferon gamma homologue in Fugu, Takifugu rubripes. Fish Shellfish Immunol. 2004, 17,
403409.
59. Zou, J.; Carrington, A.; Collet, B.; Dijkstra, J.M.; Yoshiura, Y.; Bols, N.; Secombes, C.
Identification and bioactivities of IFN-γ in rainbow trout Oncorhynchus mykiss: The first Th1-
type cytokine characterized functionally in fish. J. Immunol. 2005, 175, 24842494.
60. Igawa, D.; Sakai, M.; Savan, R. An unexpected discovery of two interferon gamma-like genes
along with interleukin (IL)-22 and -26 from teleost: IL-22 and -26 genes have been described for
the first time outside mammals. Mol. Immunol. 2006, 43, 9991009.
61. Milev-Milovanovic, I.; Long, S.; Wilson, M.; Bengten, E.; Miller, N.W.; Chinchar, V.G.
Identification and expression analysis of interferon gamma genes in channel catfish. Immunogenetics
2006, 58, 7080.
62. Stolte, E.H.; Savelkoul, H.F.J.; Wiegertjes, G.; Flik, G.; van Kemenade, B.M. Differential
expression of two interferon-γ genes in common carp (Cyprinus carpio L.). Dev. Comp. Immunol.
2008, 32, 14671481.
63. Grayfer, L.; Garcia, E.G.; Belosevic, M. Comparison of macrophage antimicrobial responses
induced by type II interferons of the goldfish (Carassius Auratus L.). J. Biol. Chem. 2010, 285,
2353723547.
64. Furnes, C.; Seppola, M.; Robertsen, B. Molecular characterisation and expression analysis of
interferon gamma in Atlantic cod (Gadus morhua). Fish Shellfish Immunol. 2009, 26, 285292.
Biology 2015, 4 898
65. Jung, C.Y.; Hikima, J.; Ohtani, M.; Jang, H.B.; del Castillo, C.S.; Nho, S.W.; Cha, I.S.; Park, S.B.;
Aoki, T.; Jung, T.S. Recombinant interferon-γ activates immune responses against Edwardsiella
tarda infection in the olive flounder, Paralichthys olivaceus. Fish Shellfish Immunol. 2012, 33,
197203.
66. Skjesol, A.; Hansen, T.; Shi, C.Y.; Thim, H.L.; Jørgensen, J.B. Structural and functional studies of
STAT1 from Atlantic salmon (Salmo salar). BMC Immunol. 2010, doi:10.1186/1471-2172-11-17.
67. Arts, J.A.J.; Tijhaar, E.J.; Chadzinska, M.; Savelkoul, H.F.J.; van Kemenade, B.M.L. Functional
analysis of carp interferon-γ: Evolutionary conservation of classical phagocyte activation.
Fish Shellfish Immunol. 2010, 29, 793802.
68. Yang, K.; Zhang, S.; Chen, D.; Zhang, A.; Wang, X.; Zhou, H. IFN-γ-activated lymphocytes
boost nitric oxide production in grass carp monocytes/macrophages. Fish Shellfish Immunol.
2013, 35, 16351641.
69. Sieger, D.; Stein, C.; Neifer, D.; van der Sar, A.M.; Leptin, M. The role of gamma interferon in
innate immunity in the zebrafish embryo. Dis. Model. Mech. 2009, 2, 571581.
70. Grayfer, L.; Belosevic, M. Molecular characterization of novel interferon gamma receptor 1
isoforms in zebrafish (Danio rerio) and goldfish (Carassius auratus L.). Mol. Immunol. 2009, 46,
30503059.
71. Grayfer, L.; Belosevic, M. Molecular characterization of tumor necrosis factor receptors 1 and 2
of the goldfish (Carassius aurutus L.). Mol. Immunol. 2009, 46, 21902199.
72. López-Muñoz, A.; Roca, F.J.; Meseguer, J.; Mulero, V. New insights into the evolution of IFNs:
Zebrafish group II IFNs induce a rapid and transient expression of IFN-dependent genes and
display powerful antiviral activities. J. Immunol. 2009, 182, 34403449.
73. García-Castillo, J.; Pelegrín, P.; Mulero, V.; Meseguer, J. Molecular cloning and expression analysis
of tumor necrosis factor α from a marine fish reveal its constitutive expression and ubiquitous nature.
Immunogenetics 2002, 54, 200207.
74. Grayfer, L.; Walsh, J.G.; Belosevic, M. Characterization and functional analysis of goldfish
(Carassius auratus L.) tumor necrosis factor-alpha. Dev. Comp. Immunol. 2008, 32, 532543.
75. Hirono, I.; Nam, B.H.; Kurobe, T.; Aoki, T. Molecular cloning, characterization, and expression
of TNF cDNA and gene from Japanese flounder Paralychthys olivaceus. J. Immunol. 2000, 165,
44234427.
76. Kadowaki, T.; Harada, H.; Sawada, Y.; Kohchi, C.; Soma, G.I.; Takahashi, Y.; Inagawa, H.
Two types of tumor necrosis factor-α in bluefin tuna (Thunnus orientalis) genes: Molecular
cloning and expression profile in response to several immunological stimulants. Fish Shellfish
Immunol. 2009, 27, 585594.
77. Laing, K.J.; Wang, T.; Zou, J.; Holland, J.; Hong, S.; Bols, N.; Hirono, I.; Aoki, T.; Secombes, C.J.
Cloning and expression analysis of rainbow trout Oncorhynchus mykiss tumour necrosis factor-α.
Eur. J. Biochem. 2001, 268, 13151322.
78. Lam, F.W.S.; Wu, S.Y.; Lin, S.J.; Lin, C.C.; Chen, Y.M.; Wang, H.C.; Chen, T.Y.; Lin, H.T.;
Lin, J.H.Y. The expression of two novel orange-spotted grouper (Epinephelus coioides) TNF
genes in peripheral blood leukocytes, various organs, and fish larvae. Fish Shellfish Immunol.
2011, 30, 618629.
Biology 2015, 4 899
79. Nascimento, D.S.; Pereira, P.J.B.; Reis, M.I.R.; do Vale, A.; Zou, J.; Silva, M.T.; Secombes, C.J.;
dos Santos, N.M.S. Molecular cloning and expression analysis of sea bass (Dicentrarchus labrax L.)
tumor necrosis factor-α (TNF-α). Fish Shellfish Immunol. 2007, 23, 701710.
80. Ordás, M.C.; Costa, M.M.; Roca, F.J.; López-Castejón, G.; Mulero, V.; Meseguer, J.; Figueras, A.;
Novoa, B. Turbot TNFα gene: Molecular characterization and biological activity of the recombinant
protein. Mol. Immunol. 2007, 44, 389400.
81. Saeij, J.P.; Stet, R.J.; Groeneveld, A.; van Kemenade, L.B.; van Muiswinkel, W.B.; Wiegertjes, G.F.
Molecular and functional characterization of a fish inducible-type nitric oxide synthase.
Immunogenetics 2000, 51, 339346.
82. Savan, R.; Kono, T.; Igawa, D.; Sakai, M. A novel tumor necrosis factor (TNF) gene present in
tandem with the TNF-α gene on the same chromosome in teleosts. Immunogenetics 2005, 57,
140150.
83. Zhang, A.; Chen, D.; Wei, H.; Du, L.; Zhao, T.; Wang, X.; Zhou, H. Functional characterization
of TNF-α in grass carp head kidney leukocytes: Induction and involvement in the regulation of
NF-κB signaling. Fish Shellfish Immunol. 2012, 33, 11231132.
84. Hong, S.; Li, R.; Xu, Q.; Secombes, C.J.; Wang, T. Two types of TNF-α exist in teleost fish:
Phylogeny, expression, and bioactivity analysis of type-II TNF-α3 in rainbow trout oncorhynchus
mykiss. J. Immunol. 2013, 191, 59595972.
85. Nguyen-Chi, M.; Laplace-Builhe, B.; Travnickova, J.; Luz-Crawford, P.; Tejedor, G.; Phan, Q.T.;
Duroux-Richard, I.; Levraud, J.P.; Kissa, K.; Lutfalla, G.; et al. Identification of polarized
macrophage subsets in zebrafish. eLife 2015, doi:10.7554/eLife.07288.
86. Mills, C.D.; Ley, K. M1 and M2 macrophages: The chicken and the egg of immunity. J. Innate
Immun. 2014, 6, 716726.
87. Hamilton, J.A. Colony-stimulating factors in inflammation and autoimmunity. Nat. Rev. Immunol.
2008, 8, 533544.
88. Dijkstra, J.M. TH2 and Treg candidate genes in elephant shark. Nature 2014, 511, E7E9.
89. Broughton, S.E.; Nero, T.L.; Dhagat, U.; Kan, W.L.; Hercus, T.R.; Tvorogov, D.; Lopez, A.F.;
Parker, M.W. The βc receptor family—Structural insights and their functional implications.
Cytokine 2015, 74, 247258.
90. Mills, C.D.; Lenz, L.L.; Ley, K. M1/M2 Macrophages: The Arginine Fork in the Road to Health
and Disease; Frontiers Media SA: Lausanne, Switzerland, 2015.
91. Grayfer, L.; Hodgkinson, J.W.; Belosevic, M. Analysis of the antimicrobial responses of primary
phagocytes of the goldfish (Carassius auratus L.) against Mycobacterium marinum. Dev. Comp.
Immunol. 2011, 35, 11461158.
92. Briggs, R.T.; Drath, D.B.; Karnovsky, M.L.; Karnovsky, M.J. Localization of NADH oxidase on
the surface of human polymorphonuclear leukocytes by a new cytochemical method. J. Cell Biol.
1975, 67, 566586.
93. DeLeo, F.R.; Quinn, M.T. Assembly of the phagocyte NADPH oxidase: Molecular interaction of
oxidase proteins. J. Leukoc. Biol. 1996, 60, 677691.
94. El-Benna, J.; Dang, P.M.-C.; Gougerot-Pocidalo, M.A. Priming of the neutrophil NADPH
oxidase activation: Role of p47phox phosphorylation and NOX2 mobilization to the plasma
membrane. Semin. Immunopathol. 2008, 30, 279289.
Biology 2015, 4 900
95. Leto, T.L.; Lomax, K.J.; Volpp, B.D.; Nunoi, H.; Sechler, J.M.; Nauseef, W.M.; Clark, R.A.;
Gallin, J.I.; Malech, H.L. Cloning of a 67-kD neutrophil oxidase factor with similarity to a
noncatalytic region of p60c-SRC. Science 1990, 248, 727730.
96. Parkos, C.A.; Dinauer, M.C.; Walker, L.E.; Allen, R.A.; Jesaitis, A.J.; Orkin, S.H. Primary
structure and unique expression of the 22-kilodalton light chain of human neutrophil cytochrome b.
Proc. Natl. Acad. Sci. USA 1988, 85, 33193323.
97. Parkos, C.A.; Allen, R.A.; Cochrane, C.G.; Jesaitis, A.J. The quaternary structure of the plasma
membrane b-type cytochrome of human granulocytes. Biochim. Biophys. Acta 1988, 932, 7183.
98. Parkos, C.A.; Allen, R.A.; Cochrane, C.G.; Jesaitis, A.J. Purified cytochrome b from human
granulocyte plasma membrane is comprised of two polypeptides with relative molecular weights
of 91,000 and 22,000. J. Clin. Investig. 1987, 80, 732742.
99. Tsunawaki, S.; Mizunari, H.; Nagata, M.; Tatsuzawa, O.; Kuratsuji, T. A novel cytosolic
component, p40phox, of respiratory burst oxidase associates with p67phox and is absent in
patients with chronic granulomatous disease who lack p67phox. Biochem. Biophys. Res. Commun.
1994, 199, 13781387.
100. Volpp, B.D.; Nauseef, W.M.; Donelson, J.E.; Moser, D.R.; Clark, R.A. Cloning of the cDNA and
functional expression of the 47-kilodalton cytosolic component of human neutrophil respiratory
burst oxidase. Proc. Natl. Acad. Sci. USA 1989, 86, 71957199.
101. Grayfer, L.; Belosevic, M. Cytokine regulation of teleost inflammatory responses. In New
Advances and Contributions to Fish Biology; Turker, H., Ed.; InTech: Rijeka, Croatia, 2012.
102. Inoue, Y.; Suenaga, Y.; Yoshiura, Y.; Moritomo, T.; Ototake, M.; Nakanishi, T. Molecular cloning
and sequencing of Japanese pufferfish (Takifugu rubripes) NADPH oxidase cDNAs. Dev. Comp.
Immunol. 2004, 28, 911925.
103. Olavarría, V.H.; Gallardo, L.; Figueroa, J.E.; Mulero, V. Lipopolysaccharide primes the respiratory
burst of Atlantic salmon SHK-1 cells through protein kinase C-mediated phosphorylation of
p47phox. Dev. Comp. Immunol. 2010, 34, 12421253.
104. Boltaña, S.; Doñate, C.; Goetz, F.W.; MacKenzie, S.; Balasch, J.C. Characterization and expression
of NADPH oxidase in LPS-, poly(I:C)- and zymosan-stimulated trout (Oncorhynchus mykiss W.)
macrophages. Fish Shellfish Immunol. 2009, 26, 651661.
105. Jørgensen, J.B.; Robertsen, B. Yeast β-glucan stimulates respiratory burst activity of Atlantic
salmon (Salmo salar L.) macrophages. Dev. Comp. Immunol. 1995, 19, 4357.
106. Sepulcre, M.P.; López-Castejón, G.; Meseguer, J.; Mulero, V. The activation of gilthead
seabream professional phagocytes by different PAMPs underlines the behavioural diversity of
the main innate immune cells of bony fish. Mol. Immunol. 2007, 44, 20092016.
107. Stafford, J.L.; Galvez, F.; Goss, G.G.; Belosevic, M. Induction of nitric oxide and respiratory
burst response in activated goldfish macrophages requires potassium channel activity. Dev. Comp.
Immunol. 2002, 26, 445459.
108. Ardó, L.; Jeney, Z.; Adams, A.; Jeney, G. Immune responses of resistant and sensitive common
carp families following experimental challenge with Aeromonas hydrophila. Fish Shellfish Immunol.
2010, 29, 111116.
Biology 2015, 4 901
109. Hodgkinson, J.W.; Ge, J.Q.; Grayfer, L.; Stafford, J.; Belosevic, M. Analysis of the immune
response in infections of the goldfish (Carassius auratus L.) with Mycobacterium marinum.
Dev. Comp. Immunol. 2012, 38, 456465.
110. Sharp, G.J.E.; Secombes, C.J. The role of reactive oxygen species in the killing of the bacterial
fish pathogen Aeromonas salmonicida by rainbow trout macrophages. Fish Shellfish Immunol.
1993, 3, 119129.
111. Kim, M.S.; Hwang, Y.J.; Yoon, K.J.; Zenke, K.; Nam, Y.K.; Kim, S.K.; Kim, K.H. Molecular
cloning of rock bream (Oplegnathus fasciatus) tumor necrosis factor-α and its effect on the
respiratory burst activity of phagocytes. Fish Shellfish Immunol. 2009, 27, 618624.
112. MacMicking, J.; Xie, Q.W.; Nathan, C. Nitric Oxide and Macrophage Function. Annu. Rev.
Immunol. 1997, 15, 323350.
113. Nathan, C.; Xie, Q.W. Nitric oxide synthases: Roles, tolls, and controls. Cell 1994, 78, 915918.
114. Denicola, A.; Rubbo, H.; Rodriguez, D.; Radi, R. Peroxynitrite-mediated cytotoxicity to trypanosoma
cruzi. Arch. Biochem. Biophys. 1993, 304, 279286.
115. Henard, C.A.; Vázquez-Torres, A. Nitric oxide and salmonella pathogenesis. Front. Microbiol.
2011, doi:10.3389/fmicb.2011.00084.
116. Laing, K.J.; Grabowski, P.S.; Belosevic, M.; Secombes, C.J. A partial sequence for nitric oxide
synthase from a goldfish (Carassius auratus) macrophage cell line. Immunol. Cell Biol. 1996, 74,
374379.
117. Laing, K.J.; Hardie, L.J.; Aartsen, W.; Grabowski, P.S.; Secombes, C.J. Expression of an inducible
nitric oxide synthase gene in rainbow trout Oncorhynchus mykiss. Dev. Comp. Immunol. 1999, 23,
7185.
118. Vojtech, L.N.; Sanders, G.E.; Conway, C.; Ostland, V.; Hansen, J.D. Host immune response and
acute disease in a zebrafish model of francisella pathogenesis. Infect. Immun. 2009, 77, 914925.
119. Losada, A.P.; Bermúdez, R.; Faílde, L.D.; Quiroga, M.I. Quantitative and qualitative evaluation
of iNOS expression in turbot (Psetta maxima) infected with Enteromyxum scophthalmi. Fish
Shellfish Immunol. 2012, 32, 243248.
120. Joerink, M.; Ribeiro, C.M.S.; Stet, R.J.M.; Hermsen, T.; Savelkoul, H.F.J.; Wiegertjes, G.F.
Head kidney-derived macrophages of common carp (Cyprinus carpio L.) show plasticity and
functional polarization upon differential stimulation. J. Immunol. 2006, 177, 6169.
121. Ishibe, K.; Yamanishi, T.; Wang, Y.; Osatomi, K.; Hara, K.; Kanai, K.; Yamaguchi, K.; Oda, T.
Comparative analysis of the production of nitric oxide (NO) and tumor necrosis factor-alpha
(TNF-alpha) from macrophages exposed to high virulent and low virulent strains of Edwardsiella
tarda. Fish Shellfish Immunol. 2009, 27, 386389.
122. Stafford, J.L.; Wilson, E.C.; Belosevic, M. Recombinant transferrin induces nitric oxide response
in goldfish and murine macrophages. Fish Shellfish Immunol. 2004, 17, 171185.
123. Campos-Pérez, J.J.; Ellis, A.E.; Secombes, C.J. Toxicity of nitric oxide and peroxynitrite to
bacterial pathogens of fish. Dis. Aquat. Organ. 2000, 43, 109115.
124. Chettri, J.K.; Raida, M.K.; Kania, P.W.; Buchmann, K. Differential immune response of rainbow
trout (Oncorhynchus mykiss) at early developmental stages (larvae and fry) against the bacterial
pathogen Yersinia ruckeri. Dev. Comp. Immunol. 2012, 36, 463474.
Biology 2015, 4 902
125. Tafalla, C.; Figueras, A.; Novoa, B. Role of nitric oxide on the replication of viral haemorrhagic
septicemia virus (VHSV), a fish rhabdovirus. Vet. Immunol. Immunopathol. 1999, 72, 249256.
126. Cambier, C.J.; Takaki, K.K.; Larson, R.P.; Hernandez, R.E.; Tobin, D.M.; Urdahl, K.B.;
Cosma, C.L.; Ramakrishnan, L. Mycobacteria manipulate macrophage recruitment through
coordinated use of membrane lipids. Nature 2014, 505, 218222.
127. Elks, P.M.; van der Vaart, M.; van Hensbergen, V.; Schutz, E.; Redd, M.J.; Murayama, E.;
Spaink, H.P.; Meijer, A.H. Mycobacteria counteract a TLR-mediated nitrosative defense mechanism
in a zebrafish infection model. PLoS ONE 2014, 9, e100928.
128. Elks, P.M.; Brizee, S.; van der Vaart, M.; Walmsley, S.R.; van Eeden, F.J.; Renshaw, S.A.;
Meijer, A.H. Hypoxia inducible factor signaling modulates susceptibility to mycobacterial
infection via a nitric oxide dependent mechanism. PLoS Pathog. 2013, 9, e1003789.
129. Canton, J.; Khezri, R.; Glogauer, M.; Grinstein, S. Contrasting phagosome pH regulation and
maturation in human M1 and M2 macrophages. Mol. Biol. Cell 2014, 25, 33303341.
130. Delamarre, L.; Pack, M.; Chang, H.; Mellman, I.; Trombetta, E.S. Differential lysosomal proteolysis
in antigen-presenting cells determines antigen fate. Science 2005, 307, 16301634.
131. Cellier, M.F.M. Cell-type specific determinants of NRAMP1 expression in professional phagocytes.
Biology 2013, 2, 233283.
132. Aragones, J.; Elorza, A.; Acosta-Iborra, B.; Landazuri, M.O. Myeloid hypoxia-inducible factors
in inflammatory diseases. Crit. Rev. Immunol. 2011, 31, 113.
133. Shay, J.E.S.; Celeste Simon, M. Hypoxia-inducible factors: Crosstalk between inflammation and
metabolism. Semin. Cell Dev. Biol. 2012, 23, 389394.
134. Forbes, J.R.; Gros, P. Divalent-metal transport by NRAMP proteins at the interface of host-pathogen
interactions. Trends Microbiol. 2001, 9, 397403.
135. Kuhn, D.E.; Baker, B.D.; Lafuse, W.P.; Zwilling, B.S. Differential iron transport into phagosomes
isolated from the RAW264.7 macrophage cell lines transfected with NRAMP1Gly169 or
NRAMP1Asp169. J. Leukoc. Biol. 1999, 66, 113119.
136. Atkinson, P.G.; Barton, C.H. High level expression of NRAMP1G169 in RAW264.7 cell
transfectants: Analysis of intracellular iron transport. Immunology 1999, 96, 656662.
137. Jabado, N.; Jankowski, A.; Dougaparsad, S.; Picard, V.; Grinstein, S.; Gros, P. Natural resistance
to intracellular infections: Natural resistance-associated macrophage protein 1 (NRAMP1)
functions as a pH-dependent manganese transporter at the phagosomal membrane. J. Exp. Med.
2000, 192, 12371248.
138. Goswami, T.; Bhattacharjee, A.; Babal, P.; Searle, S.; Moore, E.; Li, M.; Blackwell, J.M.
Natural-resistance-associated macrophage protein 1 is an H+/bivalent cation antiporter. Biochem. J.
2001, 354, 511519.
139. Saeij, J.P.; Wiegertjes, G.F.; Stet, R.J. Identification and characterization of a fish natural
resistance-associated macrophage protein (NRAMP) cDNA. Immunogenetics 1999, 50, 6066.
140. Sibthorpe, D.; Baker, A.M.; Gilmartin, B.J.; Blackwell, J.M.; White, J.K. Comparative analysis
of two slc11 (NRAMP) loci in Takifugu rubripes. DNA Cell Biol. 2004, 23, 4558.
141. Chen, H.; Waldbieser, G.C.; Rice, C.D.; Elibol, B.; Wolters, W.R.; Hanson, L.A. Isolation and
characterization of channel catfish natural resistance associated macrophage protein gene. Dev.
Comp. Immunol. 2002, 26, 517531.
Biology 2015, 4 903
142. Cooper, C.A.; Shayeghi, M.; Techau, M.E.; Capdevila, D.M.; MacKenzie, S.; Durrant, C.;
Bury, N.R. Analysis of the rainbow trout solute carrier 11 family reveals iron import < or = pH 7.4
and a functional isoform lacking transmembrane domains 11 and 12. FEBS Lett. 2007, 581,
25992604.
143. Chen, S.L.; Zhang, Y.X.; Xu, J.Y.; Meng, L.; Sha, Z.X.; Ren, G.C. Molecular cloning,
characterization and expression analysis of natural resistance associated macrophage protein
(NRAMP) cDNA from turbot (Scophthalmus maximus). Comp. Biochem. Physiol. B 2007, 147,
2937.
144. Chen, S.L.; Xu, M.Y.; Ji, X.S.; Yu, G.C. Cloning and characterisation of natural resistance
associated macrophage protein (NRAMP) cDNA from red sea bream (Pagrus major). Fish
Shellfish Immunol. 2004, 17, 305313.
145. Chen, S.L.; Wang, Z.J.; Xu, M.Y.; Gui, J.F. Molecular identification and expression analysis of
natural resistance associated macrophage protein (NRAMP) cDNA from Japanese flounder
(Paralichthys olivaceus). Fish Shellfish Immunol. 2006, 20, 365373.
146. Elibol-Flemming, B.; Waldbieser, G.C.; Wolters, W.R.; Boyle, C.R.; Hanson, L.A. Expression
analysis of selected immune-relevant genes in channel catfish during Edwardsiella ictaluri
infection. J. Aquat. Anim. Health 2009, 21, 2335.
147. Burge, E.J.; Gauthier, D.T.; Ottinger, C.A.; van Veld, P.A. Mycobacterium-inducible NRAMP in
striped bass (Morone saxatilis). Infect. Immun. 2004, 72, 16261636.
148. Donovan, A.; Brownlie, A.; Zhou, Y.; Shepard, J.; Pratt, S.J.; Moynihan, J.; Paw, B.H.; Drejer, A.;
Barut, B.; Zapata, A.; et al. Positional cloning of zebrafish ferroportin1 identifies a conserved
vertebrate iron exporter. Nature 2000, 403, 776781.
149. Abboud, S.; Haile, D.J. A novel mammalian iron-regulated protein involved in intracellular iron
metabolism. J. Biol. Chem. 2000, 275, 1990619912.
150. Nairz, M.; Fritsche, G.; Brunner, P.; Talasz, H.; Hantke, K.; Weiss, G. Interferon-gamma limits
the availability of iron for intramacrophage Salmonella typhimurium. Eur. J. Immunol. 2008, 38,
19231936.
151. Nairz, M.; Theurl, I.; Ludwiczek, S.; Theurl, M.; Mair, S.M.; Fritsche, G.; Weiss, G. The
co-ordinated regulation of iron homeostasis in murine macrophages limits the availability of iron
for intracellular Salmonella typhimurium. Cell. Microbiol. 2007, 9, 21262140.
152. Zandt, K.E.V.; Sow, F.B.; Florence, W.C.; Zwilling, B.S.; Satoskar, A.R.; Schlesinger, L.S.;
Lafuse, W.P. The iron export protein ferroportin 1 is differentially expressed in mouse macrophage
populations and is present in the mycobacterial-containing phagosome. J. Leukoc. Biol. 2008, 84,
689700.
153. Johnson, E.E.; Wessling-Resnick, M. Iron metabolism and the innate immune response to
infection. Microbes Infect. 2012, 14, 207216.
154. Garrick, M.D.; Singleton, S.T.; Vargas, F.; Kuo, H.-C.; Zhao, L.; Knöpfel, M.; Davidson, T.;
Costa, M.; Paradkar, P.; Roth, J.A.; et al. DMT1: Which metals does it transport? Biol. Res.
2006, 39, 7985.
155. Yang, C.G.; Liu, S.S.; Sun, B.; Wang, X.L.; Wang, N.; Chen, S.L. Iron-metabolic function and
potential antibacterial role of Hepcidin and its correlated genes (Ferroportin 1 and Transferrin
Receptor) in Turbot (Scophthalmus maximus). Fish Shellfish Immunol. 2013, 34, 744755.
Biology 2015, 4 904
156. Wang, X.F.; Wang, H.S.; Wang, H.; Zhang, F.; Wang, K.F.; Guo, Q.; Zhang, G.; Cai, S.H.; Du, J.
The role of indoleamine 2,3-dioxygenase (IDO) in immune tolerance: Focus on macrophage
polarization of THP-1 cells. Cell. Immunol. 2014, 289, 4248.
157. Taylor, M.W.; Feng, G.S. Relationship between interferon-gamma, indoleamine 2,3-dioxygenase,
and tryptophan catabolism. FASEB J. 1991, 5, 25162522.
158. Grohmann, U.; Bronte, V. Control of immune response by amino acid metabolism. Immunol. Rev.
2010, 236, 243264.
159. Hayaishi, O. Utilization of superoxide anion by indoleamine oxygenase-catalyzed tryptophan and
indoleamine oxidation. In Recent Advances in Tryptophan Research; Filippini, G.A., Costa, C.V.L.,
Bertazzo, A., Eds.; Springer: New York, NY, USA, 1996; pp. 285289.
160. Werner, E.R.; Werner-Felmayer, G.; Fuchs, D.; Hausen, A.; Reibnegger, G.; Wachter, H. Parallel
induction of tetrahydrobiopterin biosynthesis and indoleamine 2,3-dioxygenase activity in human
cells and cell lines by interferon-gamma. Biochem. J. 1989, 262, 861866.
161. Werner-Felmayer, G.; Werner, E.R.; Fuchs, D.; Hausen, A.; Reibnegger, G.; Wachter, H. Induction
of indoleamine 2,3-dioxygenase in human cells in vitro. In Kynurenine and Serotonin Pathways;
Schwarcz, R., Young, S.N., Brown, R.R., Eds.; Springer: New York, NY, USA, 1991; pp. 505509.
162. Yoshida, R.; Imanishi, J.; Oku, T.; Kishida, T.; Hayaishi, O. Induction of pulmonary indoleamine
2,3-dioxygenase by interferon. Proc. Natl. Acad. Sci. USA 1981, 78, 129132.
163. Yuasa, H.J.; Takubo, M.; Takahashi, A.; Hasegawa, T.; Noma, H.; Suzuki, T. Evolution of vertebrate
indoleamine 2,3-dioxygenases. J. Mol. Evol. 2007, 65, 705714.
164. Martinez, F.O.; Gordon, S. The M1 and M2 paradigm of macrophage activation: Time for
reassessment. F1000Prime Rep. 2014, doi:10.12703/P6-13.
165. Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions.
Immunity 2010, 32, 593604.
166. Gardiner, M.R.; Gongora, M.M.; Grimmond, S.M.; Perkins, A.C. A global role for zebrafish
KLF4 in embryonic erythropoiesis. Mech. Dev. 2007, 124, 762774.
167. Martinez, F.O.; Helming, L.; Milde, R.; Varin, A.; Melgert, B.N.; Draijer, C.; Thomas, B.;
Fabbri, M.; Crawshaw, A.; Ho, L.P.; et al. Genetic programs expressed in resting and IL-4
alternatively activated mouse and human macrophages: Similarities and differences. Blood 2013,
121, e57e69.
168. Roberts, S.B.; Langenau, D.M.; Goetz, F.W. Cloning and characterization of prostaglandin
endoperoxide synthase-1 and -2 from the brook trout ovary. Mol. Cell. Endocrinol. 2000, 160,
8997.
169. Skjesol, A.; Liebe, T.; Iliev, D.B.; Thomassen, E.I.S.; Tollersrud, L.G.; Sobhkhez, M.;
Lindenskov Joensen, L.; Secombes, C.J.; Jørgensen, J.B. Functional conservation of suppressors
of cytokine signaling proteins between teleosts and mammals: Atlantic salmon SOCS1 binds to
JAK/STAT family members and suppresses type I and II IFN signaling. Dev. Comp. Immunol.
2014, 45, 177189.
170. Varela, M.; Diaz-Rosales, P.; Pereiro, P.; Forn-Cuní, G.; Costa, M.M.; Dios, S.; Romero, A.;
Figueras, A.; Novoa, B. Interferon-induced genes of the expanded IFIT family show conserved
antiviral activities in non-mammalian species. PLoS ONE 2014, 9, e100015.
Biology 2015, 4 905
171. Ohtani, M.; Hayashi, N.; Hashimoto, K.; Nakanishi, T.; Dijkstra, J.M. Comprehensive clarification
of two paralogous interleukin 4/13 loci in teleost fish. Immunogenetics 2008, 60, 383397.
172. Wang, T.; Secombes, C.J. The evolution of IL-4 and IL-13 and their receptor subunits. Cytokine
2015, 75, 813.
173. Wang, T.; Huang, W.; Costa, M.M.; Martin, S.A.M.; Secombes, C.J. Two copies of the genes
encoding the subunits of putative interleukin (IL)-4/IL-13 receptors, IL-4Rα, IL-13Rα1 and
IL-13Rα2, have been identified in rainbow trout (Oncorhynchus mykiss) and have complex
patterns of expression and modulation. Immunogenetics 2011, 63, 235253.
174. Zhu, L.; Pan, P.; Fang, W.; Shao, J.; Xiang, L. Essential role of IL-4 and IL-4Rα interaction in
adaptive immunity of zebrafish: Insight into the origin of Th2-like regulatory mechanism in
ancient vertebrates. J. Immunol. 2012, 188, 55715584.
175. Hu, Y.-L.; Xiang, L.-X.; Shao, J.-Z. Identification and characterization of a novel immunoglobulin Z
isotype in zebrafish: Implications for a distinct B cell receptor in lower vertebrates. Mol. Immunol.
2010, 47, 738746.
176. Lin, A.F.; Xiang, L.X.; Wang, Q.L.; Dong, W.R.; Gong, Y.F.; Shao, J.Z. The DC-SIGN of
zebrafish: Insights into the existence of a CD209 homologue in a lower vertebrate and its
involvement in adaptive immunity. J. Immunol. 2009, 183, 73987410.
177. Barksdale, A.R.; Bernard, A.C.; Maley, M.E.; Gellin, G.L.; Kearney, P.A.; Boulanger, B.R.;
Tsuei, B.J.; Ochoa, J.B. Regulation of arginase expression by T-helper II cytokines and isoproterenol.
Surgery 2004, 135, 527535.
178. Green, S.J.; Crawford, R.M.; Hockmeyer, J.T.; Meltzer, M.S.; Nacy, C.A. Leishmania major
amastigotes initiate the L-arginine-dependent killing mechanism in IFN-γ-stimulated macrophages by
induction of tumor necrosis factor-alpha. J. Immunol. 1990, 145, 42904297.
179. Iniesta, V.; Gómez-Nieto, L.C.; Molano, I.; Mohedano, A.; Carcelén, J.; Mirón, C.; Alonso, C.;
Corraliza, I. Arginase I induction in macrophages, triggered by Th2-type cytokines, supports the
growth of intracellular Leishmania parasites. Parasite Immunol. 2002, 24, 113118.
180. Liew, F.Y.; Li, Y.; Moss, D.; Parkinson, C.; Rogers, M.V.; Moncada, S. Resistance to Leishmania
major infection correlates with the induction of nitric oxide synthase in murine macrophages.
Eur. J. Immunol. 1991, 21, 30093014.
181. Mills, C.D. Macrophage arginine metabolism to ornithine/urea or nitric oxide/citrulline: A life or
death issue. Crit. Rev. Immunol. 2001, 21, 399425.
182. Morris, S.M. Recent advances in arginine metabolism: Roles and regulation of the arginases.
Br. J. Pharmacol. 2009, 157, 922930.
183. Munder, M.; Eichmann, K.; Morán, J.M.; Centeno, F.; Soler, G.; Modolell, M. Th1/Th2-regulated
expression of arginase isoforms in murine macrophages and dendritic cells. J. Immunol. 1999,
163, 37713777.
184. Lang, R.; Patel, D.; Morris, J.J.; Rutschman, R.L.; Murray, P.J. Shaping gene expression in
activated and resting primary macrophages by IL-10. J. Immunol. 2002, 169, 22532263.
185. Wright, P.A.; Campbell, A.; Morgan, R.L.; Rosenberger, A.G.; Murray, B.W. Dogmas and
controversies in the handling of nitrogenous wastes: Expression of arginase Type I and II genes in
rainbow trout: Influence of fasting on liver enzyme activity and mRNA levels in juveniles. J. Exp.
Biol. 2004, 207, 20332042.
Biology 2015, 4 906
186. Chang, C.I.; Zoghi, B.; Liao, J.C.; Kuo, L. The involvement of tyrosine kinases, cyclic
AMP/protein kinase A, and p38 mitogen-activated protein kinase in IL-13-mediated arginase I
induction in macrophages: Its implications in IL-13-inhibited nitric oxide production. J. Immunol.
2000, 165, 21342141.
187. Forlenza, M.; Fink, I.R.; Raes, G.; Wiegertjes, G.F. Heterogeneity of macrophage activation in
fish. Dev. Comp. Immunol. 2011, 35, 12461255.
188. Wiegertjes, G.F.; Wentzel, A.S.; Spaink, H.P.; Elks, P.M.; Fink, I.R. Polarization of immune
responses in fish: The “macrophages first” point of view. Mol. Immunol. 2015, doi:10.1016/
j.molimm.2015.09.026.
189. Gensel, J.C.; Zhang, B. Macrophage activation and its role in repair and pathology after spinal
cord injury. Brain Res. 2015, 1619, 111.
190. Ehrchen, J.; Steinmüller, L.; Barczyk, K.; Tenbrock, K.; Nacken, W.; Eisenacher, M.; Nordhues, U.;
Sorg, C.; Sunderkötter, C.; Roth, J. Glucocorticoids induce differentiation of a specifically activated,
anti-inflammatory subtype of human monocytes. Blood 2007, 109, 12651274.
191. Van de Garde, M.D.B.; Martinez, F.O.; Melgert, B.N.; Hylkema, M.N.; Jonkers, R.E.; Hamann, J.
Chronic exposure to glucocorticoids shapes gene expression and modulates innate and adaptive
activation pathways in macrophages with distinct changes in leukocyte attraction. J. Immunol.
2014, 192, 11961208.
192. Maule, A.G.; Tripp, R.A.; Kaattari, S.L.; Schreck, C.B. Stress alters immune function and
disease resistance in chinook salmon (Oncorhynchus tshawytscha). J. Endocrinol. 1989, 120,
135142.
193. Montero, D.; Marrero, M.; Izquierdo, M.S.; Robaina, L.; Vergara, J.M.; Tort, L. Effect of vitamin
E and C dietary supplementation on some immune parameters of gilthead seabream (Sparus aurata)
juveniles subjected to crowding stress. Aquaculture 1999, 171, 269278.
194. Pickering, A.D.; Pottinger, T.G. Cortisol can increase the susceptibility of brown trout, Salmo
trutta L., to disease without reducing the white blood cell count. J. Fish Biol. 1985, 27, 611619.
195. Wang, R.; Belosevic, M. The in vitro effects of estradiol and cortisol on the function of a
long-term goldfish macrophage cell line. Dev. Comp. Immunol. 1995, 19, 327336.
196. Ferrante, C.J.; Leibovich, S.J. Regulation of macrophage polarization and wound healing.
Adv. Wound Care 2012, 1, 1016.
197. Grayfer, L.; Belosevic, M. Identification and molecular characterization of the interleukin-10
receptor 1 of the zebrafish (Danio rerio) and the goldfish (Carassius auratus L.). Dev. Comp.
Immunol. 2012, 36, 408417.
198. Wei, H.; Wang, S.; Qin, L.; Wang, X.; Zhou, H. Molecular characterization, 3D modeling of
grass carp interleukin-10 receptor 1 (IL10R1). Engineering 2013, 5, 214219.
199. Monte, M.M.; Wang, T.; Collet, B.; Zou, J.; Secombes, C.J. Molecular characterisation of four
class 2 cytokine receptor family members in rainbow trout, Oncorhynchus mykiss. Dev. Comp.
Immunol. 2015, 48, 4354.
© 2015 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article
distributed under the terms and conditions of the Creative Commons Attribution license
(http://creativecommons.org/licenses/by/4.0/).
... Similarly, other AMPs such as NK-Lysin can induce macrophagerelated genes in mudskipper (Bolephthalmus pectinirostris), Barbel steed (Hemibarbus labeo) or black rockfish (Sebastes schlegelii) (Chen et al., 2021;Ding et al., 2019;Hao et al., 2022), pointing to macrophage recruitment. The recruitment of leukocytes might in turn favor the regulation of the immune responses as fish neutrophils and macrophages are involved in the fine-tune of the innate response, guaranteeing host integrity, and promoting the adaptative response (Havixbeck and Barreda, 2015;Hodgkinson et al., 2015). In that sense our data also show that AMPs injection triggered the formation of T and B cells in HK, which are key cells in the immune regulation and the adaptative immune response against viruses (Castro et al., 2013;Chang et al., 2011;Nakanishi et al., 2015;Øvergård et al., 2013). ...
... Moreover, a protective impact on the gut epithelial integrity was described for IL-10 that reduce the permeability of the gut epithelial through raising transepithelial electrical resistance (Al-Sadi et al. 2009). Granulocyte-macrophage colonystimulating factor stimulates the proliferation and differentiation of a wide range of leucocytes by binding to heteromeric cell-surface receptors on leucocytes and endothelial cells (Hodgkinson et al. 2015). Fishmeal sparing with a mixture of alternative protein sources at 50% simultaneously induced up-regulation of IL-10 and also some pro-inflammatory genes such as IL1-ß, IL-8, IL-17F and TNF-α in the hindgut of L. calcarifer, but total replacement did not significantly affect these genes. ...
Article
Full-text available
This research was conducted to determine the effects of a mixture of organic acid (OAs) in low fishmeal diets on Asian seabass ( Lates calcarifer ) juveniles (54.2 g). There were two dietary groups including high fishmeal diet (HFM, 45.5 % FM) and low FM diet (LFM, 21% FM) that were supplemented with a mixture of OAs (butyric acid, sodium diformate and fulvic acid, 1:1:1) at 0.5 and 1.0% levels. Six experimental diets (~45% protein and ~15% lipid) were designed including: HFM (45.5% FM), HFM +0.5 (HFM diet + 0.5% OAs), HFM +1.0 (HFM diet + 1.0% OAs), LFM (21% FM), LFM +0.5 (LFM diet + 0.5% OAs), and LFM +1.0 (LFM diet + 1.0% OAs). Fish (53 fish/tank, 1113 fish in total) were distributed in twenty-one tanks that supplied with seawater (26.5 °C, 46.0 ppt). Each treatment had three replicates. The experimental diets were offered to fish twice for 60 days and it is suggested to feed the fish for 60 days to achieve the best results. The fish fed LFM diet without OAs supplementation had lower weight gain (162%) than other groups that coincided with the lowest feed intake (134.1 g). Fish fed LFM +1.0 had the highest gut lactobacillus bacteria colonies count. HFM +0.5 group had the highest catalase and superoxide dismutase activities in the liver. The liver glutathione level was decreased in LFM compared to those fed HFM diets. LFM +1.0 group had the highest amount of liver malondialdehyde value ( P < 0.05). Fish fed HFM +0.5 and LFM +1.0 diets had the highest values of liver alanine aminotransferase. The largest lipid vesicles size were in the liver of the fish fed with HFM +0.5 , LFM and LFM +0.5 diets and the smallest ones were in the fish fed with HFM +1.0 . Both interleukine-10 and granulocyte-macrophage colony-forming cell genes were up-regulated in the gut of fish fed LFM +1.0 and HFM +1.0 diets after 30 and 60 days, respectively. Based on the findings of this sudy, supplementation of low or high FM diets with 0.5% OAs mixture is recommended for L. calcarifer juveniles.
... Located in almost all tissues of animals and across all vertebrate species are the macrophages, and they function importantly in homeostasis and the protection of the host. Being cells found in almost all tissues, macrophages help to keep environments homeostatic, and when there is an infection, they are usually one of the principal kinds of cells to contact pathogens that invade, which is followed by a suitable immune response [80]. In fishes, transferrin functions as the main fish macrophages activator. ...
... Phagocytes are a crucial component of the innate immune system and play a major role in eliminating pathogenic microorganisms, particularly monocytes/Mfs and neutrophils. Macrophages are particularly effective in this regard, as they are durable, efficient, and capable of rapidly killing pathogens through formation of phagolysosome and the production of reactive intermediates (56,57). Phagocytosis is a complex and heterogeneous immunobiologic process that is a hallmark of antimicrobial host defense (41,58). ...
Article
Collectin is a crucial component of the innate immune system and plays a vital role in the initial line of defense against pathogen infection. In mammals, collectin kidney 1 (CL-K1) is a soluble collectin that has recently been identified to have significant functions in host defense. However, the evolutionary origins of immune defense of CL-K1 and its mechanism in clearance of pathogenic microorganisms remain unclear, especially in early vertebrates. In this study, the Oreochromis niloticus CL-K1 (OnCL-K1) protein was purified and identified, which was capable of binding to two important pathogens of tilapia, Streptococcus agalactiae and Aeromonas hydrophila. Interestingly, OnCL-K1 exhibited direct bactericidal activity by binding to lipoteichoic acid or LPS on cell walls, disrupting the permeability and integrity of the bacterial membrane in vitro. Upon bacterial challenge, OnCL-K1 significantly inhibited the proliferation of pathogenic bacteria, reduced the inflammatory response, and improved the survival of tilapia. Further research revealed that OnCL-K1 could associate with OnMASPs to initiate and regulate the lectin complement pathway. Additionally, OnCD93 reduced the complement-mediated hemolysis by competing with OnMASPs for binding to OnCL-K1. More importantly, OnCL-K1 could facilitate phagocytosis by collaborating with cell surface CD93 in a lectin pathway–independent manner. Moreover, OnCL-K1 also promoted the formation of phagolysosomes, which degraded and killed ingested bacteria. Therefore, this study reveals the antibacterial response mechanism of CL-K1 in primitive vertebrates, including promoting complement activation, enhancing opsonophagocytosis, and killing of macrophages, as well as its internal links, all of which provide (to our knowledge) new insights into the understanding of the evolutionary origins and regulatory roles of the collectins in innate immunity.
Article
A comparative study of the leukocyte formula of peripheral blood, the head section of the kidney and spleen of the bream Abramis brama, inhabiting the reservoirs of the Middle Volga (Kuibyshev, Cheboksary and Gorky), was carried out. In the studied fish, a predominance of lymphocytes and a low content of neutrophils were recorded in the peripheral blood and spleen. In the head kidney of the breams from the Kuibyshev Reservoir, a significantly high level of blast forms of cells and a low level of neutrophils were noted, and in individuals from the Gorky Reservoir, a low content of monocytes and the absence of eosinophils were noted compared to fish from other reservoirs.
Article
A comparative study of the composition of peripheral blood leukocytes of plunderfish and grey rockcod or squama caught in the Ross Sea has been carried out. Blood smears revealed cells of different morphofunctional characteristics and structure: lymphocytes, monocytes, neutro- and eosinophils, and blast cells. An analysis of the leukogram showed that the white blood of the studied species has a lymphoid character. In the rockcod leukogram, the proportion of lymphocytes is significantly higher, while that of monocytes and blast forms is lower. In the plunderfish, in comparison with grey rockcod, the leukocyte cells of the same name are usually larger.
Article
Full-text available
While the mammalian macrophage phenotypes have been intensively studied in vitro, the dynamic of their phenotypic polarization has never been investigated in live vertebrates. We used the zebrafish as a live model to identify and trail macrophage subtypes. We generated a transgenic line whose macrophages expressing tumour necrosis factor alpha (tnfa), a key feature of classically activated (M1) macrophages, express fluorescent proteins Tg(mpeg1:mCherryF/tnfa:eGFP-F). Using 4D-confocal microscopy, we showed that both aseptic wounding and Escherichia coli inoculation triggered macrophage recruitment, some of which started to express tnfa. RT-qPCR on Fluorescence Activated Cell Sorting (FACS)-sorted tnfa+ and tnfa− macrophages showed that they, respectively, expressed M1 and alternatively activated (M2) mammalian markers. Fate tracing of tnfa+ macrophages during the time-course of inflammation demonstrated that pro-inflammatory macrophages converted into M2-like phenotype during the resolution step. Our results reveal the diversity and plasticity of zebrafish macrophage subsets and underline the similarities with mammalian macrophages proposing a new system to study macrophage functional dynamic.
Book
Full-text available
Fish is an important component of aquaculture with up to 80% of animal protein contribution especially in developing countries. Therefore, aquaculture is a bright spot with great potential in many countries worldwide. This potential raises the issue of achieving a sustainable and environmentally friendly aquaculture. Many areas need to be explored and developed. Updated information about some of the main issues that currently affects aquaculture was presented in this book for the scientific researchers in the field of aquaculture and fish biology. The book is sub-divided into nine distinct chapters. The importance of each of these contributions was briefly summarized here. The understanding of the mechanisms that control inflammation in teleosts may allow for the development of strategies to prevent infectious diseases. Therefore, the main concepts of innate immune mechanism are reviewed in Chapter 1 by Sebastián Reyes-Cerpa, Kevin Maisey, Felipe Reyes-López, Daniela Toro-Ascuy, Ana M. Sandino, Mónica Imarai and Chapter 2 by Leon Grayfer and Miodrag Belosevic. Their reviews focus on the recent advancements in the understanding of the biology of hallmark fish pro-inflammatory cytokines. In the following Chapter 3 by Barbara A. Katzenback, Fumihiko Katakura and Miodrag Belosevic, the author are provided an overview of the current knowledge of the fish model systems on the sentinel cells (macrophages and neutrophils) of the innate immune response. Jacinto E. S. Díaz, Eugenia L. in Chapter 4 discuss a short framework of effects of xenobiotics on the responses of freshwater fish across molecular to population level when have been exposed to environmental stressors. Their review considers the use of fish as sentinel organisms to assess the anthropogenic impacts over the freshwater ecosystems. Climate changes and dams are likely to represent the most important threats to freshwater fish around the world. The effects of climate change and dams on the ecosystem are reviewed by Carlos E. C. Freitas, Alexandre A. F. Rivas, Caroline P. Campos, Igor Sant’Ana, James R. Kahn, Maria A. A. Correa and Michel Catarino in Chapter 5. The silver catfish (jundiá) have been used for bioindicator of environmental contamination for many researches and can be used to aquatic biological systems. In Chapter 6 by Cláudia T. Pimpão, Ênio Moura, Ana C. Fredianelli, Luciana G. Galeb, Rita M. V. M. Rocha, Francisco P. Montanha aimed to review some aspects of the toxicology silver catfish. Christine Genge, Leif Hove-Madsen and Glen F. Tibbits in Chapter 7 reviewed the roles of the atrium and ventricle in achieving variability with myocardial contractility among the telesot species. In chapter 8 Ivan Viega, Rui A. Carvalho, Miguel Â. Pardal, John G. Jones focus on the metabolism of carbohydrates by fish in aquaculture. In Chapter 9, Javier Sánchez-Hernández, María J. Servia, Rufino Vieira-Lanero, Fernando Cobo discuss the variables that are involved in the feeding behaviour of brown trout as an example of a predatory freshwater fish species. Finally, I would like to acknowledge the contributors for their cooperation. I also express my gratitude to Ms. Iva Simcic at Intech in assisting me with editing of this book.
Article
Macrophages exposed to IFN-gamma and infected with amastigotes of Leishmania major develop the capacity to eliminate the intracellular pathogen. This antimicrobial activity of activated macrophages correlates with the initiation of nitrogen oxidation of L-arginine, yet other reports suggest that two signals are required for induction of this biochemical pathway for effector activity. In the present studies, macrophages treated with up to 100 U/ml IFN-gamma, or 100 ng LPS, or 10(7) amastigotes produced minimal quantities (less than 9 microM) of NO2- and failed to develop cytotoxic effector activities. In contrast, the combination of IFN-gamma and either LPS (greater than 0.1 ng) or amastigotes (10(6) induced high concentrations (much greater than 30 microM) of NO2- and macrophage cytotoxicity against intra- and extracellular targets. The induction of nitrogen oxidation by amastigotes could be dissociated from LPS-induced events by 1) performing the assays in the presence of polymyxin B (which blocked LPS effects, but not amastigote effects), 2) determining the threshold of IFN-gamma required to prime cells for subsequent trigger (1 U/ml for LPS trigger effects; 10-fold higher for amastigotes), and 3) determining the heat sensitivity of the two trigger agents (amastigote effects abolished at 100 degrees C; LPS effects unaffected at this temperature). Further, culture fluids from amastigote-infected macrophages did not contain detectable LPS (less than 6 pg/ml). Possible parasite and cell-associated factors that could contribute to the induction of nitrogen oxidation and cytotoxic activity of IFN-gamma treated macrophages were examined: only certain intact microorganisms, LPS from a variety of bacteria, and the cytokine TNF alpha were effective. Both NO2- production and intracellular killing were abolished by the addition of anti-TNF-alpha mAb in the assay. TNF-alpha was produced by amastigote-infected macrophages and IFN-gamma dramatically enhanced secretion of this cytokine; IFN-gamma alone had no effect. Endogenous TNF-alpha produced during infection of macrophages with L. major acted in an autocrine fashion to trigger the production of L-arginine-derived toxic nitrogen intermediates that killed the intracellular parasites.
Article
The biosynthesis of macrophage colony-stimulating factor 1 (CSF-1) was examined in mouse NIH-3T3 fibroblasts transfected with a retroviral vector expressing the 554-amino-acid product of a human 4-kilobase (kb) CSF-1 cDNA. Similar to results previously obtained with a 1.6-kb human cDNA that codes for a 256-amino-acid CSF-1 precursor, the results of the present study showed that NIH-3T3 cells expressing the product of the 4-kb clone produced biologically active human CSF-1 and were transformed by an autocrine mechanism when cotransfected with a vector containing a human c-fms (CSF-1 receptor) cDNA. The 4-kb CSF-1 cDNA product was synthesized as an integral transmembrane glycoprotein that was assembled into disulfide-linked dimers and rapidly underwent proteolytic cleavage to generate a soluble growth factor. Although the smaller CSF-1 precursor specified by the 1.6-kb human cDNA was stably expressed as a membrane-bound glycoprotein at the cell surface and was slowly cleaved to release the extracellular growth factor, the cell-associated product of the 4-kb clone was efficiently processed to the secreted form and was not detected on the plasma membrane. Digestion with glycosidic enzymes indicated that soluble CSF-1 encoded by the 4-kb cDNA contained both asparagine(N)-linked and O-linked carbohydrate chains, whereas the product of the 1.6-kb clone had only N-linked oligosaccharides. Removal of the carbohydrate indicated that the polypeptide chain of the secreted 4-kb cDNA product was longer than that of the corresponding form encoded by the smaller clone. These differences in posttranslational processing may reflect diverse physiological roles for the products of the two CSF-1 precursors in vivo.
Book
Wide and fascinating is the field of research on tryptophan, a most versatile amino acid, transformed, as it is, in our organism into many biologically active substances. This volume contains the proceedings of the Eighth International Meeting on Tryptophan Research, held at the University of Padova, Padova, Italy, from June 25 to 29 1995, under the auspices of the University of Padova, National Research Council, Italian Chemical Society-Division of Pharmaceutical Sciences, Region ofthe Veneto and City of Padova. The meeting was held in Padova to commemorate Prof. Luigi Musajo twenty years after his death and the editors dedicate this book to him in recognition of his pioneering work in tryptophan metabolism. Prof. Osamu Hayaishi delivered the Musajo Memorial Award Lecture: Tryptophan oxygenase. and sleep. Figure I shows the ISTRY President Prof. Simon N. Young presenting the Musajo Memorial Medal to Prof. Hayaishi during the Opening Ceremony. Two hundred scientists from twenty two countries participated in the meeting. These proceedings contain 121 papers encompassing a variety of topics and disciplines.
Article
Macrophages of the body represent a widely dispersed, versatile, and highly responsive homeostatic and defense system. They contribute to innate as well as adaptive immunity to infection, mediating trophic as well as injurious interactions with their local and systemic environment. The concept of macrophage activation has evolved over recent decades, from relatively simple paradigms to bewildering complexity. This chapter will review the background to present understanding of alternative activation of macrophages, consider its relevance to health and disease, and suggest questions for future studies. © 2014 Springer Science+Business Media New York. All rights reserved.