Zebra Fish

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

| |

Received: 25 September 2020    Revised: 9 November 2020    Accepted: 10 November 2020

DOI: 10.1111/jfd.13310

REVIEW

Review of inflammation in fish and value of the zebrafish model

Jose Carlos Campos-Sánchez | María Ángeles Esteban

Department of Cell Biology and Histology,


Faculty of Biology, Immunobiology for Abstract
Aquaculture Group, University of Murcia, Inflammation is a crucial step in the development of chronic diseases in humans.
Murcia, Spain
Understanding the inflammation environment and its intrinsic mechanisms when it
Correspondence is produced by harmful stimuli may be a key element in the development of human
María Ángeles Esteban, Department of
Cell Biology and Histology. Faculty of disease diagnosis. In recent decades, zebrafish (Danio rerio) have been widely used
Biology, Campus Regional de Excelencia in research, due to their exceptional characteristics, as a model of various human
Internacional “Campus Mare Nostrum,”
University of Murcia, 30100 Murcia, Spain. diseases. Interestingly, the mediators released during the inflammatory response of
Email: aesteban@um.es both the immune system and nervous system, after its integration in the hypothala-
Funding information mus, could also facilitate the detection of injury through the register of behavioural
Fundación Séneca, Grant/Award Number: changes in the fish. Although there are many studies that give well-defined informa-
19883/GERM/15; Ministerio de Economía
y Competitividad, Grant/Award Number: tion separately on such elements as the recruitment of cells, the release of pro- and
AGL2017-88370-C3-1-R anti-inflammatory mediators or the type of neurotransmitters released against dif-
ferent triggers, to the best of our knowledge there are no reviews that put all this
knowledge together. In the present review, the main available information on inflam-
mation in zebrafish is presented in order to facilitate knowledge about this important
process of innate immunity, as well as the stress responses and behavioural changes
derived from it.

KEYWORDS

inflammation, mediators, nervous system regulation, pain, Zebrafish (Danio rerio)

1 | I N FL A M M ATI O N A N D ZE B R A FI S H in the central nervous system (CNS) or Kupffer cells in the liver, with
the onset of inflammation, circulating immune cells such as monocytes,
Inflammation is a local temporary response triggered by the innate im- granulocytes and T cells are recruited to the site of inflammation via
mune system in order to protect an organism against external damage. secretion of inflammatory mediators (Ley et al., 2007). In fact, the re-
Among the most characteristic signs of inflammation can be found in- cruitment of immune cells is a necessary step to initiate the resolu-
tensification of vascular permeability related to swelling, an increase tion process after an inflammatory response (Singer & Clark, 1999). In
in blood flow and heat sensation associated with redness, and a nerve its attempts to restore physiological homeostasis, inflammation may
fibre sensitization link to functional disorders and pain (Bruce, 1913; persist for a long time, becoming a chronic response (chronic inflam-
Calixto et al., 2003; Nathan, 2002). In general, the presence of differ- mation; Nathan & Ding, 2010). In humans, chronic inflammation is
ent pathogens, irritants, injuries or trauma provides the basis for initiat- related to numerous diseases, such as cardiovascular disease and di-
ing this acute reaction (acute inflammation), which can be developed in abetes (Dandona et al., 2004), Alzheimer's (Perry et al., 2007), cancer
minutes to hours and last for days or weeks. Furthermore, depending (Grivennikov et al., 2010) and psoriasis (Candel et al., 2014). The effects
on the timing, the trigger, the anatomical site (inflammation may affect of inflammation are closely linked to how the inflammation has been
any tissue) or the severity of the inflammation produced, the type of initiated, maintained and finished, and it should be deeply regulated,
cells and the mediators involved will vary (Calder et al., 2013). Although not only by the local environment in which it has been produced, but
there are some local or tissue-resident immune cells, such as microglia also by the rest of the organism (Abbas et al., 2017).

J Fish Dis. 2020;00:1–17. wileyonlinelibrary.com/journal/jfd |


© 2020 John Wiley & Sons Ltd     1
|
2       CAMPOS-SÁNCHEZ ANd ESTEBAN

The zebrafish (Danio rerio) is a tropical teleost fish belonging to 2 | S TAG E S O F I N FL A M M ATI O N —TH E
the minnow family (Cyprinidae) of the order Cypriniformes. It has BAC KG RO U N D
been used as a model for different inflammation-related human
diseases due to the high degree of genetic and functional ho- In sharp contrast to mammals, which are homeotherms, fish are
mology that it shares with mammals (García-Moreno et al., 2019; poikilotherms. Consequently, small changes in the environmen-
Grunwald & Eisen, 2002). In addition, its rapid embryological de- tal parameters make them highly influenced to any type of ex-
velopment, transparent body and little size permit easy handling ternal injury and, therefore, more susceptible to inflammation
of specimens in the laboratory, requiring relatively low costs (Delamare-Deboutteville et al., 2006). Due to this high environ-
(Chakraborty et al., 2009). Mechanisms of inflammation and ho- mental dependence, teleost fish skin plays a key role in protecting
meostasis restoration have been studied by using nanoparticles, and preventing the entry of pathogens into the epithelium by the
transgenic fish or drugs, such as 2,4,6-trinitrobenzenesulfonic acid secretion of a mucus layer which is involved in immunity (Griffin &
(TNBS), used in mice to induce experimental models of Crohn's Mitchell, 2007; Salinas et al., 2011). Unlike mammal skin, and more
disease or inflammatory bowel disease, in order to develop new specifically human skin, which has many active defence mecha-
therapies for humans (Chakraborty et al., 2009; Duan et al., 2018; nisms (Kupper & Fuhlbrigge, 2004), fish lack of major lymphoid
Geiger et al., 2013; Kizil et al., 2015; Oehlers et al., 2011; Zhang accumulations. In fact, there is still little information about the
et al., 2019). For instance, mechanisms of cell recruitment during natural location of the lymphoid cells in the fish integument, or if
tail fin regeneration after inflammatory events have been iden- they are recruited as a result of an immune reaction or inflamma-
tified and coupled with research in brain regeneration (Petrie tion. However, most of the fish leucocytes seem to migrate as a re-
et al., 2015). Another example is the use of silica nanoparticles sponse to any damage through mucus secretions (Esteban, 2012;
in zebrafish embryos to reveal the involvement of the JAK1/TF Parkhurst & Saltzman, 1994; Rombout et al., 1993). For example,
(Janus kinase 1/tissue factor) signalling pathway during thrombus the number of antibody-secreting cells (ASCs) in the skin of cat-
formation in vivo and the release of pro-inflammatory and pro- fish immunized against the protozoan Ichthyophthirius multifiliis (a
coagulant cytokines from endothelial cells (Duan et al., 2018). parasite which infects gills and skin) was increased and remained
Moreover, anti-inflammatory drugs and natural products, such as elevated for weeks after the last parasite exposure, pointing at
indolealkylamines, have been associated with the suppression of a dynamic number of ASC in response to the parasite infection
TLR4/MyD88/NF-κB (Toll-like receptor 4/myeloid differentiation (Zhao et al., 2008). The main inflammatory features that make
primary response 88/nuclear factor kappa-light-chain enhancer major differences between mammals and fish are summarized in
of activated B cells) and TLR4/MyD88/MAPKs (MAPKs, mito- Table 1.
gen-activated protein kinase) signalling pathways, combined with Then, as the first defence line, fish skin is composed of resident
regulating lipid homeostasis, in an induced lipopolysaccharide non-immune cells, epithelial cells, mucosal epithelial cells and vascu-
(LPS)-inflammation model of zebrafish (Zhang et al., 2019). The lar endothelial cells, which alert the immune system to the presence
nociception mechanisms and pain that affect zebrafish behaviour of inflammation-causing damage and modulate the inflammatory re-
have also been studied (Filby et al., 2010; Maximino et al., 2010; sponse (Al-Soudi et al., 2017; Harris, 2014; Matsumoto et al., 2018)
Norton & Bally-Cuif, 2010; Sneddon, 2009). However, to the best (Figure 1). Upon exposure to an insult in a stage prior to the action
of our knowledge, such findings have never been presented to- of the immune system, local sensory nerve fibres report to the
gether; doing so might lead to a better understanding of the mech- CNS and the motor nerve fibres the damage that has just been pro-
anisms behind inflammation and the basic physiology associated duced in order to initiate muscle reflex (Calixto et al., 2003; Hehn
with this important process in zebrafish. et al., 2012). Together with the immune cells, these tissue-resident
Thus, in this review, because an injury or trauma produced cells initiate the inflammation process through both molecular and
in the skin of teleost fish may induce an inflammatory reaction cellular mechanisms (Hawiger & Zienkiewicz, 2019). In general, there
that may be modulated by nerve fibres that are integrated in the are two types of inflammation: acute and chronic. Acute inflamma-
wound microenvironment, we will focus specifically on the sub- tion is the initial response of the body to harmful stimuli, whereas
sequent phases of skin inflammation and the crosstalk between prolonged inflammation is known as chronic inflammation. Acute in-
inflammation and the nervous system in zebrafish. At the same flammation is developed in five successive stages (Bordés Gonzalez
time, the nerve fibres may inform the CNS about the stress being et al., 1994).
produced locally. Here, we will mainly focus on the relationship
between the recruitment of immune cells at the inflammation site
and the molecules that may mediate this reciprocity, as well as 2.1 | First stage—Release of mediators
the effect of the inflammatory milieu on the CNS. Thus, with this
study, a different approach will be available that will allow us to Inflammation starts with the release of numerous inflammatory
understand how the immune and nervous systems are connected preformed mediators by tissue-resident immune and non-immune
during inflammation. cells, along with the retraction of the endothelial cells in order to
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      3

TA B L E 1   Principal inflammatory features that make major differences between mammals and fish

Stages of inflammation Mammals Fish References

Background Homeotherms Poikilotherms (more susceptible to Delamare-Deboutteville et al. (2006)


inflammation)
MALT (*Humans: SALT) Lack of major lymphoid accumulations Esteban (2012), Rombout et al. (1993),
(MALT) Parkhurst and Saltzman (1994)
Genome sequenced Genome non-sequenced or partially Savan and Sakai (2006), Postlethwait
sequenced. Genome duplication et al. (2000)
Pain studies Lack of information about pain and Malafoglia et al. (2013), Rose (2002),
behaviour (controversial issue) Woodruff (2018)
First stage Release of NF-κB High conservation of NF-κB, key Mulero et al. (2019)
mediators modulator of inflammation
Conservation of Conservation of cell pathways and Secombes et al. (2011), Wang and
cell pathways and mediators: conservation of cytokines Secombes (2013)
mediators and neurotransmitters function
(paralogues)
– Lack of cytokine antibodies for Secombes et al. (2011), Wang and
functional studies Secombes (2013)
Second stage Effect of Neutrophils Acidophilic granulocytes (functional Meseguer et al. (1994), Sepulcre
mediators equivalent to neutrophils of mammals) et al. (2002), Morcillo et al. (2015)
Mastocytes produce Mast cells in most fish species do not Larsen and Henson (1983), Cole
histamine produce histamine et al. (1995), MacGlashan (2003)
Clear effect of cytokines Controversial effect of cytokines, (e.g. Key et al. (2017)
(e.g. fever) “emotional fever”)
Platelets Thrombocytes (functional equivalent to Belamarich et al. (1966), Kim et al. (2010)
platelets of mammals)
Few studies point to the effect of Oosterhof et al. (2015)
mediators in an inflammatory responseº
Third stage Cellular Leucocytes migrate Leucocytes migrate from the head Meseguer et al. (1995)
recruitment from the bone marrow kidney
Resident and local Resident leucocytes? Rombout et al. (1993), Parkhurst and
leucocytes Saltzman (1994)
Fourth step M1–M2 dichotomy M1–M2 dichotomy conserved, but there Joerink, Ribeiro, et al. (2006), Joerink,
conserved is controversy in the nomenclature due Savelkoul, et al. (2006), Forlenza
to the TH1-TH2 presence or absence et al. (2011)
Regulation of NF-κB: activation NF-κB conserved function: activation of Gugasyan et al. (2004), Etemadi
inflammation of homeostatic homeostatic mechanism et al. (2015)
mechanism
Fifth step Reparation No regenerating Inflammation activates neurogenesis and Kizil et al. (2015)
capacity regenerating capacity
Lack of studies about wound healing

increase blood flow, vasodilation and vascular permeability, which factors is the action responsible for regulating the genes that en-
are associated with the redness and swelling signs (Bordés Gonzalez code all these mediators (Hawiger & Zienkiewicz, 2019). Here, it is
et al., 1994; Calder et al., 2013; Hawiger & Zienkiewicz, 2019; important to highlight the role of NF-κB family transcription factor
Figure 2). These events allow access to the site of inflammation in the gene expression modulation and in the release of pro-in-
by many large and soluble molecules and facilitate the recruit- flammatory cytokines, due to its high conservation from mammals
ment of leucocytes from the systemic circulation (Kolaczkowska to fish (Mulero et al., 2019). Besides, it seems that the regulation
& Kubes, 2013; Larsen & Henson, 1983; Medzhitov, 2008). These of the immune responses in vertebrates is relatively well con-
secreted factors include pro-inflammatory cytokines (mainly served (conservation of function; Zou & Secombes, 2016). For
interleukin-1β [IL-1β], tumour necrosis factor α [TNF-α] and in- instance, the classical pro-inflammatory cytokines IL-1β, TNF-α
terleukin-6 [IL-6]), adhesion molecules, chemokines, proteolytic and IL-6 prevail as paralogues (multiple copies) in most teleosts
proteins, histamines, prostaglandins, leukotrienes, neuropeptides (Secombes et al., 2011; Wang & Secombes, 2013). Nonetheless,
and neurotransmitters (Kolaczkowska & Kubes, 2013; Larsen & the minimal existence of fish cytokines antibodies (only IFN and
Henson, 1983; Medzhitov, 2008). The activation of transcription TNF-α) makes difficult the functional study of fish cytokines. The
4       | CAMPOS-SÁNCHEZ ANd ESTEBAN

Hypothalamus

N
Descending efferent Ascending
pathways afferent pathways
Epidermis

Skin cells
Langerhans cells
Mastocyte like-cells
Endothelium cells
Neurons
Afferent fibres
Efferent fibres
Blood vessel
Dorsal Root
Dermis

Ganglion
C and Aδ Inflammation trigger
fibres

F I G U R E 1   Schematic representation of skin and nervous system background of zebrafish in a previous stage of an inflammatory
reaction. Resident non-immune cells, immune cells and neurons involved are shown. Key cells and molecules involved are explained on the
right box. Skin cells, Langerhans cells and vascular endothelial cells wander around the different skin layers and alert to the presence of an
inflammation trigger in order to initiate the inflammatory reaction. C and Aδ fibres warn the central nervous system of the damage produced

available results on this topic have been focussed on the bioactivi- in the inflammation site (Cole et al., 1995; Larsen & Henson, 1983;
ties of cytokine recombinant proteins or have been obtained by MacGlashan, 2003). Nonetheless, zebrafish and teleost mast cells
using cell lines from fish or mammals (Secombes et al., 2011; Wang (also known as eosinophilic granular cells) may be similar but not
& Secombes, 2013). identical to mammals. Furthermore, eosinophilic granular cells con-
tain in their cytoplasmic granules of serotonin or histamine, depend-
ing on the studied fish species, although, usually, they have the same
2.2 | Second stage—Effect of mediators functions (Ezeasor & Stokoe, 1980).
Otherwise, the cytokines TNF-α and IL-1β released by the resi-
The inflammatory exudate and the blood flow are able to produce dent cells are able to induce post-capillary venule endothelial cells
a local tumour in the affected area, where all mediators previously to express E-selectin and increase the expression of ligands for leu-
released trigger the production of immunoglobulins, kinins, acute- cocyte integrins and to stimulate cells to secrete chemokines, such
phase reactants and coagulation factors while generating comple- as C-X-C motif chemokine ligand 8 (CXCL8) (also known as IL-8) and
ment activation (Bordés Gonzalez et al., 1994; Figure 3). Mammals chemokine (C-C motif) ligand 2 (CCL2), while binding to neutrophils
and zebrafish neutrophils recruited at the inflammation place shortly and monocyte receptors, respectively (Burhans et al., 2019; Imitola
secrete chemokines that activate tissue-resident cells and recruit et al., 2004). In mammals, TNF-α and IL-1β are also able to act on
more leucocytes to the damaged area, amplifying inflammation the hypothalamus in order to induce an increase in body tempera-
(Kolaczkowska & Kubes, 2013; Sommer et al., 2020). Several stud- ture (fever; Blomqvist & Engblom, 2018). In fish, this increase of
ies in fish, like those developed by our research team in the marine body temperature is still controversial (see below; Key et al., 2017).
teleost gilthead seabream (Sparus aurata L.), point to the acidophilic Alternatively, IL-1β and IL-6 may induce hepatocytes to produce
granulocytes as the functional counterparts of the neutrophils of acute-phase reactants, including C-reactive protein, serum amy-
higher vertebrates (Meseguer et al., 1994; Morcillo et al., 2015; loid P component (SAP) and fibrinogen, which are secreted into the
Sepulcre et al., 2002). blood. In addition, TNF-β can stimulate the expression of tissue fac-
On the other hand, histamine stored and released by mammalian tor by endothelial cells in order to activate coagulation and, at the
mastocytes, and serotonin secreted from serotonergic neurons are same time, can inhibit thrombomodulin expression, which is an in-
responsible for the increase in the vascular permeability observed hibitor of coagulation (Burhans et al., 2019; Duvigneau et al., 2019).
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      5

Skin cells
Langerhans cells
Mastocyte like-cells
Endothelium cells
Efferent fibres
Proinflammatory cytokines
Adhesion molecules &
chemokines
Histamines, prostaglandins &
leukotrienes
Proteolytic proteins & ROS
Neuropeptides,
neurotransmitters & hormones

F I G U R E 2   Schematic representation of the first stage of the acute inflammation produced in the skin of zebrafish and its nexus with the
central nervous system. Key cells and molecules involved are explained on the right box. Preformed mediators released by resident cells and
nerve fibres allow the access of many large and soluble molecules to the inflammation site. Redness and swelling begin to be visible in the
inflammation site

The activation of both intrinsic and extrinsic clotting pathways place (Meseguer et al., 1995). The thymus (no present in agnatha),
has been studied in lines of zebrafish through screening to detect the spleen and the mucosa-associated lymphoid tissues (MALT)
mutants with defective blood coagulation and with the use of sev- are present in both fish and mammals, although, as it was already
eral anticoagulants, such as warfarin and hirudin (Jagadeeswaran mentioned, mammals have more local or tissue-resident leucocytes
et al., 2000). This homeostatic mechanism is triggered by mammalian wondering for insults than fish (Press & Evensen, 1999). A gene ex-
platelets and non-mammalian thrombocytes (Kim et al., 2010). So, it pression study carried out in common carp (Cyprinus carpio) dem-
is important to take into account that fish thrombocytes (including onstrated the correlation between the pro-inflammatory genes and
zebrafish) are nucleated cells and they are considered to be func- the rapid influx of neutrophilic granulocytes from the head kidney
tionally equivalent to mammalian platelets (Belamarich et al., 1966; towards lysates of epithelioma papulosum cyprini cells (Gonzalez
Kim et al., 2010). et al., 2007). Nonetheless, more studies are needed to prove or
deny the existence of resident lymphoid cells in the fish tegument
(Parkhurst & Saltzman, 1994; Rombout et al., 1993). This migration
2.3 | Third stage—Cellular recruitment consists of the rolling of circulating leucocytes along the endothe-
lium through the union of selectins and their ligands, the subsequent
The third stage of acute inflammation is mainly characterized by cell adhesion of leucocytes to the endothelium and the final extravasa-
migration from surrounding tissues to the inflammation site (Bordés tion or diapedesis (Bordés Gonzalez et al., 1994; Kolaczkowska &
Gonzalez et al., 1994; Kolaczkowska & Kubes, 2013; Ley et al., 2007) Kubes, 2013; Ley et al., 2007). All these leucocyte behaviours have
(Figure 4). In mammals, leucocytes are produced in the bone marrow, been studied by direct microscopy with fluorescently marked leu-
and afterwards, they are maturated in the secondary lymphoid or- cocytes in zebrafish, because there are at least nine transgenic lines
gans from which they are waiting for being recruited by the immune (Herbomel et al., 1999, 2001). The ensemble of adhesion molecules
system, if necessary. However, fish lack both bone marrow and and chemokines released in the vascular endothelium are respon-
lymph nodes (Chaves-Pozo et al., 2005). In their place, fish possess sible for this phase (Calder et al., 2013). One practical example of
the head kidney (considered functionally analogous to mammalian this process was shown in a study on the effect of carrageenin in
bone marrow) which acts as a major haematopoietic and lymphoid the skin of zebrafish. In this study, oedema formation was observed
organ, and leucocytes migrate from the head kidney to the injured to result from the release of the pro-inflammatory proteins and the
|
6       CAMPOS-SÁNCHEZ ANd ESTEBAN

Skin cells
Langerhans cells
Mastocyte-like cells
Endothelium cells
Thrombocytes
Granulocytes
Naïve macrophages
Efferent fibres

Proinflammatory cytokines
Adhesion molecules &
chemokines
Histamines, prostaglandins &
leukotrienes
Neuropeptides,
neurotransmitters & hormones

F I G U R E 3   Schematic representation of the second stage of the acute inflammation produced in the skin of zebrafish and its nexus with
the central nervous system. Key cells and molecules involved are explained on the right box. Chemokines activate tissue-resident cells and
facilitate the recruitment of leucocytes from the systemic circulation to the damaged area. Pro-inflammatory cytokines increase the local
temperature and activate coagulation. Histamine and serotonin released by nerve fibres and mastocytes increase vascular permeability and
produce local tumour in the affected area. Redness and swelling are visible in the site where the inflammation was initiated

recruitment of leucocytes at the inflammation site caused by carra- G; Kolaczkowska & Kubes, 2013; Korkmaz et al., 2010; Larsen &
geenin (Huang et al., 2014). Similarly, the migration/infiltration and Henson, 1983; Medzhitov, 2008; Nathan, 2006). Neutrophils can
proliferation of the immune cells in the skin of zebrafish were re- also contribute to the recruitment and action of antigen-presenting
ported in another study in which fluorescent light was able to induce cells, including macrophages, dendritic cells and endothelial cells
the early expression of skin genes associated with inflammation (Nathan, 2006). In cases where the initial cause of the inflammation
(Gonzalez et al., 2018). In addition, the genetic inhibition of TNF-α was a pathogen, neutrophils and macrophages may introduce the
and tumour necrosis factor receptor 2 (TNFR2) in an in vivo study pathogens into their cytoplasm inside vesicles (named phagosomes)
of psoriasis with a zebrafish model demonstrated the rapid infiltra- originated during the phagocytosis process. One step of this process
tion of neutrophils into the skin, but not of macrophages, and hyper- is to destroy the pathogen through the use of ROS or the proteo-
proliferation of keratinocytes through the activation of a hydrogen lytic enzymes present in cell granules and in lysosomes, which fuse
peroxide (H2O2)/NF-κB/dual oxidase 1 (Duox1) positive feedback their membrane with that of the phagosomes, giving rise to phagoly-
inflammatory loop (Candel et al., 2014). As previously mentioned, sosomes (Kolaczkowska & Kubes, 2013). Apart from phagocytosis,
infiltrating neutrophils are the first cells recruited at the site of in- neutrophils may also kill microbes by extruding their DNA and gran-
flammation, followed by monocytes. The monocytes then differ- ule contents by forming extracellular threads called neutrophil ex-
entiate into macrophages and adopt a pro-inflammatory M1-like tracellular traps, in which bacteria and fungi are trapped and killed
phenotype, becoming “classically activated M1” macrophages. The (Kolaczkowska & Kubes, 2013).
M1 macrophages show very high antigen presentation capacities
and expression of reactive oxygen species (ROS), TNF-α and IL-1β
(Block et al., 2007; Kolaczkowska & Kubes, 2013; Medzhitov, 2008; 2.4 | Fourth step—Regulation of inflammation
Nathan, 2006; Nguyen-Chi et al., 2015).
Activated neutrophils exert their function through the release In order to avoid the negative effects of excessive inflammation,
of their granule contents, which is a combination of ROS, myeloper- it is very important that the activation of inhibitory mechanisms,
oxidase and proteases (such as elastase, proteinase 3 and cathepsin including those carried out by the nervous system, integrates
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      7

Skin cells
Langerhans cells
Mastocyte like cells
Endothelium cells
Thrombocytes
Granulocytes
M1-Macrophages
Dendritic cells
Efferent fibres

Proinflammatory cytokines
Adhesion molecules &
chemokines
Histamines, prostaglandins &
leukotrienes
Proteolytic proteins & ROS
Neuropeptides,
neurotransmitters & hormones

F I G U R E 4   Schematic representation of the third stage of the acute inflammation produced in the skin of zebrafish and its nexus with
the central nervous system. Key cells and molecules involved are explained on the right box. Leucocyte is recruited to the inflammated area
while macrophages polarize to M1-like phenotype

into the inflammatory process until a balance is reached (Bordés to be short-lived and soon to undergo apoptosis. Nonetheless, a
Gonzalez et al., 1994; Nathan & Ding, 2010; Pinho-Ribeiro recently characterized subpopulation of neutrophils in zebrafish
et al., 2017) (Figure 5). Through this process, the phenotype of seems to have a longer lifespan than “normal” neutrophils and
macrophages is progressively converted to the M2-like pheno- are able to return to the circulation after activation, thereby
type, so that they become “alternatively activated M2” mac- having an anti-inflammatory effect (Ellett et al., 2015; Harvie &
rophages—the process includes intermediate phenotypes in which Huttenlocher, 2015). The combination of differently coloured flu-
both M1 and M2 markers are expressed (Nguyen-Chi et al., 2015; orescent leucocytes in zebrafish offers further insights into the in
Wiegertjes et al., 2016)—switching from the production and secre- vivo microanatomy and the visualization of the cellular physiology
tion of pro-inflammatory molecules to anti-inflammatory, such as of the host (Hall et al., 2007; Meijer et al., 2008). For instance,
cytokines and prostaglandins. The presence of macrophages with using a zebrafish tail-amputation model with fluorescently labelled
polarized M1-M2 phenotype has already been demonstrated in macrophages (mCherry) and TNF-α, it was shown that pro-inflam-
fish (Forlenza et al., 2011; Joerink, Ribeiro, et al., 2006; Joerink matory (green fluorescent protein, GFP+) and anti-inflammatory
Savelkoul & Wiegertjes, 2006). Although this M1–M2 dichotomy macrophages (GFP-) accumulated in the damaged tissue and that
seems to be evolutionarily conserved, there has been a large de- anti-inflammatory macrophages that remained were associated
bate about whether fish immune responses may be characterized with the injury until regeneration was completed, unlike pro-
in based on the TH1-TH2 presence or absence typical of mammals inflammatory macrophages, which withdrew from the damaged
(Bird et al., 2007; Secombes et al., 2011; Wang & Secombes, 2013). area. This model also made it possible to study neutrophil reverse
Some of the endogenous anti-inflammatory molecules released, migration at different stages of the inflammatory response (Yoo
as lipoxins, are able to cease the recruitment of neutrophils et al., 2010; Yoo & Huttenlocher, 2011).
to the inflammation place in favour of monocyte recruitment In the molecular field, the conserved NF-κB transcription factor
(Chandrasekharan & Sharma-Wali, 2015). In addition, resident seems to be also implicated in the onset of the return to homeosta-
and recruited macrophages participate in the scavenging of for- sis; however, fish studies are necessaries to check that this function
eign particles and cellular debris, thereby collaborating to initiate is also conserved (Etemadi et al., 2015; Gugasyan et al., 2004). For
the tissue repair process (Kolaczkowska & Kubes, 2013; Larsen & instance, a study with the zebrafish transgenic line NF-κB:GFP,Luc
Henson, 1983; Medzhitov, 2008; Nathan, 2006). The rest of the (luciferase) evidenced a NF-κB down-regulation after Phormidium sp.
neutrophils are phagocyted by macrophages since they are known exopolysaccharide exposure, showing its anti-inflammatory effects
|
8       CAMPOS-SÁNCHEZ ANd ESTEBAN

Skin cells
Langerhans cells
Mastocyte like cells
Endothelium cells
Thrombocytes
Granulocytes
M1-Macrophages
M2-Macrophages
Dendritic cells
Efferent fibres

Proinflammatory cytokines
Anti-inflammatory cytokines
Histamines, prostaglandins &
leukotrienes
Neuropeptides,
neurotransmitters & hormones

F I G U R E 5   Schematic representation of the fourth stage of the acute inflammation produced in the skin of zebrafish and its nexus with
the central nervous system. Key cells and molecules involved are explained on the right box. The polarization of macrophages from M1-like
to M2-like phenotype and the release of anti-inflammatory molecules by M2-macrophages and nerve fibres are evident. These activate
inhibitory mechanisms that compensate the effects of the pro-inflammatory molecules released previously. Redness and swelling begin to be
less visible in the site where the inflammation was initiated

(Kuri et al., 2017) For all reasons, it is not surprising that a deregula- wound debridement by neutrophils and chemotactic signals from
tion of the pivotal NF-κB signalling pathway may play a substantial macrophages (Dovi et al., 2002; Martin et al., 2003). Here, it is im-
role in the pathogenesis of most chronic inflammatory diseases, as it portant to mention the high regeneration capacity of some fish like
was already indicated (Jimi et al., 2019; Liu et al., 2017). the zebrafish, which has a striking difference to mammalian. In fact,
in zebrafish, inflammation elicited by immune cells leads to the pro-
motion of neurogenesis and skin regeneration (Kizil et al., 2015).
2.5 | Fifth step—Reparation Nonetheless, the inflammation process seems to indicate an evolu-
tionary conservation of crucial molecular mechanisms in different
Finally, mechanisms of homeostasis are activated to terminate vertebrate classes, which should be carefully analysed for a better
acute inflammation and initiate the total or partial repair of the tis- understanding.
sues damaged by the offending agent or by the self-inflammatory
response. Endocrine functions are mainly responsible for the devel-
opment of this last stage (Bordés Gonzalez et al., 1994) (Figure 6). 3 | N EU RO M O D U L ATI O N A N D
In terms of the immune system, the alternatively activated M2 I NTEG R ATI O N O F TH E C N S I N TH E
macrophages still in the inflammation focus secrete various growth CO NTRO L O F I N FL A M M ATI O N
factors in order to attract skin cells, fibroblasts and blood vessels
into the wound and increase the phagocytic activity of leucocytes The crosstalk between the immune system and the CNS to maintain
(Colton, 2009; Ponomarev et al., 2007; Shaw & Martin, 2009; Singer or recover homeostasis is largely developed by the hypothalamic–
& Clark, 1999). In fact, hydrocortisone treatment in zebrafish points pituitary–adrenal (HPA) axis and the autonomic nervous system
to the existence of fibroblast-stimulating signals from inflammatory (ANS), alerting to inflammation after its onset (Maryanovich et al.,
cells, and after their activation, granulation tissue formation and vas- 2015). Although the anatomical organization of fish CNS does not
cularization are promoted (Leibovich & Ross, 1975). Interestingly, the closely resemble that of mammalians, the main functions are highly
same study in which this was found hinted that re-epithelialization conserved across vertebrates, and brains of teleost contain equiva-
did not require an inflammatory response, being independent of lents to most mammalian cell types (Kunst et al., 2019; Machluf
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      9

Endocrine System
Activation

Skin cells
Langerhans cells
Mastocyte like cells
Endothelium cells
Thrombocytes
Granulocytes
M2-Macrophages
Efferent fibres
Anti-inflammatory cytokines
Neuropeptides,
neurotransmitters & hormones

F I G U R E 6   Schematic representation of the fifth stage of the acute inflammation produced in the skin of zebrafish and its nexus with the
central nervous system. Key cells and molecules involved are explained on the right box. Hormones released in the central nervous system
that activates the endocrine system and molecules released by the immune cells activate the damage repairment in the affected tissue,
through the activation of the homeostasis mechanism. Redness and swelling begin to disappear in the site where the inflammation was
initiated

et al., 2011). In addition, in a study using a retinal injury model of by activating cardiovascular organs, increasing blood flow to skeletal
zebrafish, an abundance of neurogenic zones was found in the adult muscles and lungs, and suppressing gastrointestinal activity. By con-
zebrafish brain, along with a high regenerative capacity, which are trast, the PNS is unrelated to stress situations, decreasing muscle
associated with the correlation between DNA demethylation and re- contractility and heart rate and increasing gastrointestinal function
generation-associated gene expression, which has not been found in (Kiba, 2002). Fibres that transduce action potentials are non-my-
mammals (Grandel & Brand, 2013; Powell et al., 2013). Furthermore, elinated slow-conducting post-synaptic neurons, called C-fibres,
adult zebrafish are able to regenerate all parts of their body success- which are involved in heat, cold and mechanical pain, and Aβ/Aδ-
fully, including their fins, heart and liver (Keightley et al., 2014; Shi myelinated presynaptic neurons, which mediate mechanosensation
et al., 2015), a property that has been extensively studied because of and mechanical pain with faster conduction (Basbaum et al., 2009).
its possible value for other vertebrates, including humans. Interestingly, the buoyancy, dilution of chemicals and relatively sta-
The zebrafish hypothalamus is a complex structure responsible ble thermal environment provided by the aquatic environment may
for integrating environmental signals, processing within the en- be related to a higher percentage of Aδ-fibres than C-fibres in tele-
docrine systems and triggering the systemic responses needed to ost fish due to their dual role in mediating reflex escape behaviour
regain and/or maintain homeostasis; moreover, its neuronal popula- and prolonged noxious stimulation (Sneddon, 2002). By contrast, in
tions have been shown to be functionally analogous to their mam- mammals there are a large number of C-fibres involved in noxious
malian counterparts (Machluf et al., 2011; Markakis, 2002). Against stimulation and inflammatory pain due to their advance during evo-
external neuroendocrine disruptors, the hypothalamus is able to op- lution and the increased chance of injury (Sneddon, 2002; Sneddon
erate through direct innervations with synaptic neurotransmission et al., 2003).
and through the secretion of neuromodulators into the surrounding Thus, upon activation by external damage, and before starting
tissue or vasculature (Andersson & Tracey, 2012; Figure 1). Indirectly, the immunological stages of the inflammation, pain stimuli are led to
external disturbances may be processed via the ANS (Kiba, 2002). neuron bodies within the dorsal root ganglia and relayed to the spi-
The ANS comprises two branches that start from the spinal cord: nal cord and hypothalamus in order to be processed (Pinho-Ribeiro
the sympathetic nervous system (SNS) and the parasympathetic ner- et al., 2017). In higher vertebrates, pain is processed through the
vous system (PNS). The SNS prepares the body for alert situations ascending afferent pathways, the spinothalamic pathway and the
|
10       CAMPOS-SÁNCHEZ ANd ESTEBAN

ventrobasal, medial and lateral areas, which project to the somato- the effect of all these known mediators in an inflammatory response
sensory cortex, allowing the location and intensity of pain from the in zebrafish (Oosterhof et al., 2015). Although most of these media-
inflamed area to be perceived (Colloca et al., 2017). In later stages, in tors have functionally equivalent orthologs in fish, there is a rela-
which the inflammation becomes chronic, several mediators called tively low sequence-based homology between fish and mammals
pyrogens (such as IL-1β, TNF-α and IL-6), released from the local (Savan & Sakai, 2006). Interestingly, an ancient genome duplication
area of inflammation, reach the thermoregulatory centre in the an- has occurred in teleosts and that is the reason why it is estimated
terior hypothalamus, through portions lacking a blood–brain barrier that 30% of genes have a duplicate variant that may exhibit redun-
(specific areas of the surfaces of the cerebral ventricles, where the dancy and share the same function (Postlethwait et al., 2000). The
metabolism of araquidonic acid is activated, producing prostaglan- roles of these mediators can vary depending on the amount, location
dins, mainly prostaglandin E2) (Biddle, 2006; King & Wood, 1957; and timing of production (Savan & Sakai, 2006). That is also the rea-
Milton & Wendlandt, 1970; Figures 3–5). In addition, magnocellu- son why some fish, like gilthead seabream and pufferfish (Tetraodon
lar neurons located in the paraventricular and supraoptic nuclei of nigroviridis), have a strong resistance to endotoxic shock triggered
the anterior hypothalamus project their axons directly to the neu- by LPS. Specifically, these teleosts lack a TLR4 ortholog, in contrast
rohypophysis (posterior lobe of the pituitary) and release oxytocin to mammals. Contrarily, zebrafish possesses two TLR4 orthologs
and arginine–vasopressin (AVP) into the general blood circulation, (Sepulcre et al., 2009). For instance, chemokine signalling molecules
inducing peripheral vasoconstriction and muscular contractions can also differ substantially between mammals and teleosts, and
(Markakis, 2002) (Landgraf & Neumann, 2004). All these physiologi- even among mammals, where some chemokines are not conserved
cal changes are related to fever and an increase in body temperature at the sequence level (Bajoghli, 2013; Nomiyama et al., 2008). In
(Dinarello, 2015). zebrafish, although many functional homologs remain to be identi-
fied, 63 chemokines and 24 receptors due to the genome duplication
have been described, representing a high number of chemokines
4  | CO M M U N I C ATI O N M E D I ATO R S and receptors in comparison with the similar molecules described
B E T W E E N I N FL A M M ATI O N A N D TH E in other species (DeVries et al., 2006). Among some pro-inflam-
N E RVO U S S YS TE M matory stimuli, cytokines such as TNF and IL-1 or bacterial toxins
may induce the expression of the C-X-C chemokine receptor type 4
Sympathetic nerve fibres play an essential role in maintaining the (CXCR4) and its ligand C-X-C motif chemokine ligand 12 (CXCL12)
homeostasis niche against stress and dangerous stimuli, forming a (also known as stromal cell-derived factor 1) (Imitola et al., 2004).
complex framework in the local microenvironment, established in Both CXCR4 and CXCL12 are expressed in the ventricular zone of
order to start a defensive response with the resident cells (Katayama the adult zebrafish brain and seem to play a role during homeostatic
et al., 2006; Yamazaki & Allen, 1990). Upon activation by an inflam- neuronal migration (Diotel et al., 2010). The chemokine CXCL8 has
matory signal, sensory nerves are able to release cytokines, neu- two homologs, CXCL8a and CXCL8b, which are also involved in neu-
ropeptides and neurotransmitters from their peripheral terminals, trophil migration towards a peripheral wound during inflammation
which modulate the effects on the vasculature and on the function and after injury (de Oliveira et al., 2013; Oehlers et al., 2010). Other
of innate and adaptive immune cells (Deutschman & Tracey, 2014) mediators, like those involved in arachidonic acid and therefore in
(Figures 2–6). Furthermore, the dendritic cells, neutrophils, mac- the synthesis of eicosanoids, such as cyclooxygenase-2 (COX-2) and
rophages, mast cells and T cells (all of which express receptors for leukotriene C4 (LTC4), participate in the stimulation of inflammation
neuronal mediators) all produce cytokines (small, secreted proteins (Funk, 2001; Ishikawaa et al., 2007).
that have a specific effect on the interactions of and communications When some of these mediators inform the CNS of the onset of
between cells) which affect the SNS (Nathan & Ding, 2010). This bi- inflammation, high-conserved neuropeptides, such as corticotro-
directional communication allows the control of the inflammation in pin-releasing hormone (CRH), substance P, calcitonin gene-related
subsequent stages by the activation of specific neuronal networks in peptide and orexins, bring support to the release of other pro-in-
the SNS (Nathan & Ding, 2010; Pinho-Ribeiro et al., 2017). Many of flammatory mediators, facilitating vasodilation and thus promoting
the mammalian cell pathways and mediators of both the immune sys- immune cell recruitment in the initial phase of inflammation (Chiu
tem and nervous system are highly conserved in zebrafish (Bollaerts et al., 2013; Hehn et al., 2012; Pavlov & Tracey, 2012). Subsequently,
et al., 2017). Likewise, inflammatory molecules, such as interleukins, the activation of the HPA axis via afferent nerves may also termi-
chemokines and growth factors, as well as neurohormones, neuro- nate inflammation through the release of glucocorticoids from the
peptides and the inhibitory or excitatory neurotransmitters released adrenal cortex (Sternberg, 2006). In zebrafish, neurotransmitters,
by GABAergic, glutamatergic, cholinergic, dopaminergic or seroton- known to play a significant role in various physiological and be-
ergic neurons, seem to be highly evolutionarily conserved among havioural processes, may also regulate leucocyte trafficking to sites
various mammalian species (Hall et al., 2009; Meijer et al., 2011; of inflammation, due to the conservation of a large variety of cir-
Panula et al., 2010; Zhang & Cui, 2014). Some of these pathways cuits and receptors (Panula et al., 2010). In response to an inflamma-
are conserved even across invertebrates, such as fruit flies, in which tory response, the nervous system acts by releasing acetylcholine
Toll-like receptors were discovered. However, few studies point to (ACh) through the efferent vagus nerve, inhibiting the production
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      11

of pro-inflammatory cytokines (TNF-α, IL-1, IL-6, IL-18) via activa- nociceptive pathways and respond to inflammation, and therefore,
tion of the homopentameric receptor α7-nicotinic (α7nAChR) in to noxious stimuli in the stage prior to the immune response, but
macrophages and reducing inflammation (Zila et al., 2017). These the debate over whether fish feel pain, considered a higher cognitive
macrophages seem to be located in the spleen, although there is process, has continued since the discovery of nociceptors in trout
controversy about how ACh reaches the organ, since there is hardly in the early 2000s (Malafoglia et al., 2013; Rose, 2002). The matter
any direct connection between the vagus nerve and the spleen depends less on the detection of noxious stimuli by fish, but rather
(Zila et al., 2017). Populations of CD4+ T cells that express the β2 on how they respond at an emotional level (Woodruff, 2018).
adrenergic receptor could act as mediators in this connexion, but In response to a painful stimulus, such as an inflammatory in-
there is still no satisfactory explanation for this (Inoue et al., 2019). sult, there is a withdrawal reflex, an emergency movement away
In mammals, ACh is degraded by two cholinesterases—acetylcho- from it, in which the nociception pathway transmits the message
linesterase (AChE) and butyrylcholinesterase (BuChE)—but the from the injured limb to the spinal cord before the order is sent di-
genome of the zebrafish encodes only the gene for AChE, which rectly back to the hurt limb (Broom, 2001). As the brain receives this
was detected in the brain (Bertrand et al., 2001; Radić et al., 1993; information afterwards, it is possible that there is no awareness of
Soreq & Seidman, 2001). Thus, AChE is involved in the termination pain (Broom, 2001). However, tissue injury and inflammation are in-
of nerve impulses through catalysis of the neurotransmitter ACh timately related to an increase in pain sensation, which is a long-term
(Dix et al., 2007). In zebrafish, dopamine is released principally from consolidated experience (Pinho-Ribeiro et al., 2017). In mammals,
the midbrain and hypothalamus, and homologs of both D1- and D2- nociceptor peripheral nerve terminals have receptors and ion chan-
receptor subtypes have been described (Narayanan et al., 2010; nels that detect molecular mediators released during inflammation,
Panula et al., 2010; Schweitzer & Driever, 2009). Dopamine or ag- leading to pain sensitivity or “hyperalgesia,” a topic up for debate
onist receptors of dopamine have been reported to modulate acti- in relation to fish (Shi et al., 2015). For instance, when a noxious
vation, proliferation and cytokine production by immune cells (Basu substance (e.g. acetic acid or bee venom) was injected in trout, the
& Dasgupta, 2000; Sarkar et al., 2010; Torres-Rosas et al., 2014). painful stimuli reduced the motivation of the fish to feed for hours
Consistent with this, the deficiency of dopamine is tightly associated (Sneddon et al., 2003). Moreover, the injection of pain killers at the
with immune system abnormalities and CNS inflammation, so the same site as the noxious substance restored their natural behaviour
zebrafish has been used as a model of biomedical research in order and physiology, possibly exemplifying the highly conservative nature
to study the molecular mechanisms of Parkinson's disease and atten- of vertebrate physiology (Mettam et al., 2011). In addition, after ex-
tion deficit hyperactivity disorder in humans (Fontana et al., 2019; posure to electric stimuli or a positive reward, trout learned to avoid
Perry, 2012; Wüllner & Klockgether, 2003). Glutamate is an inhib- the shock location or look for the reward after just a few exposures.
itor neurotransmitter involved in the ATP-dependent recruitment In fact, in case of conflict between reward and shock, they were
of microglia against an injury, through the binding of the glutamate willing to pay a pain cost in order to access the resources (Dunlop
receptor NMDA, the subsequent influx of calcium ions (Ca2+) and et al., 2006).
the release of ATP (Sieger et al., 2012). Otherwise, after stressful Due to the zebrafish's characteristics and the conservation
behaviours, noradrenaline (NA) is synthesized in neurons of the of nociceptive mechanisms across vertebrates, this fish species
locus coeruleus and in the medulla oblongata of zebrafish (Rink & has also been used not only as a model for inflammation studies
Wullimann, 2002). In the SNS, NA regulates heart function and blood but also for pain and nociception studies (Sneddon, 2009). The
pressure (Brodde et al., 2001; Feng et al., 2019; Zimmerman, 1981). behavioural responses of zebrafish larvae have been studied in
Depending on the time point of activation relative to inflammation, this context, revealing a dose-dependent increase in locomotor
low concentrations of NA are related to pro-inflammatory effects, activity against potentially painful stimuli, such as temperature
while high concentrations of NA have predominantly anti-inflamma- or chemical substances, which can be treated with common an-
tory effects (Pongratz & Straub, 2013). These pro- and anti-inflam- algesics (Lopez-Luna et al., 2017a; Prober et al., 2008; Wolman
matory effects are mediated through α-adrenergic receptors and & Granato, 2012). In fact, zebrafish specimens exposed to con-
β-adrenergic receptors, respectively (Pongratz & Straub, 2013). In tinuous stressors, such as netting and handling, showed a tran-
addition, NA seems to be involved in the adhesion molecule expres- sient rise in body temperature related to a real fever response,
sion of venular endothelial cells, facilitating leucocyte trafficking which is often referred to as “emotional fever” (Rey et al., 2015). In
(Scheiermann et al., 2013; Viswanathan & Dhabhar, 2005). addition, when skin and muscle of zebrafish larvae were burned,
axons innervating the damaged area were dramatically affected,
even presenting an up-regulation of homeostatic-related gene
5 | I N FL A M M ATI O N , PA I N A N D expression, such as c-Jun, vasoactive intestinal peptide and pitu-
B E H AV I O U R itary adenylate cyclase-activating polypeptide 1b, in enteric neu-
rons as a possible additional effect of the nociceptive mechanism
As we have seen, most of the basic anatomical regions, cells and (Malafoglia et al., 2014). In contrast, adult zebrafish reduced their
neurotransmitters are conserved between zebrafish and mam- locomotor activity after injections of formalin or acetic acid, which
mals, despite a few important differences. For instance, fish have may be reversed by opioids (e.g. morphine or buprenorphine) but
|
12       CAMPOS-SÁNCHEZ ANd ESTEBAN

not when co-administered with an opioid antagonist (Curtright Bajoghli, B. (2013). Evolution and function of chemokine receptors
in the immune system of lower vertebrates. European Journal of
et al., 2015; Lopez-Luna et al., 2017b; Magalhães et al., 2017;
Immunology, 43(7), 1686–1692. https://doi.org/10.1002/eji.20134
Steenbergen & Bardine, 2014; Taylor et al., 2017). High concen- 3557
trations of morphine (1 mg/L) are able to produce immunosup- Basbaum, A. I., Bautista, D. M., Scherrer, G., & Julius, D. (2009). Cellular
pression and down-regulation of inflammation-related genes in and molecular mechanisms of pain. Cell, 139(2), 267–284. https://doi.
org/10.1016/j.cell.2009.09.028
zebrafish inflammation models produced by LPS, while lower con-
Basu, S., & Dasgupta, P. S. (2000). Dopamine, a neurotransmitter, influ-
centrations (100 ng/L–100 μg/L) produce exacerbation of inflam- ences the immune system. Journal of Neuroimmunology, 102(2), 113–
mation by down-regulating multixenobiotic resistance transporter 124. https://doi.org/10.1016/S0165-5728(99)00176-9
genes that control LPS efflux (Mottaz et al., 2017). Belamarich, F., Fusari, M., Shepro, D., & Kien, M. (1966). In vitro studies
of aggregation of non-mammalian thrombocytes. Nature, 212(5070),
1579–1580. https://doi.org/10.1038/2121579a0
Bertrand, C., Chatonnet, A., Takke, C., Yan, Y. L., Postlethwait, J., Toutant,
6  | CO N C LU S I O N A N D FU T U R E J. P., & Cousin, X. (2001). Zebrafish acetylcholinesterase is encoded
PE R S PEC TI V E S by a single gene localized on linkage group 7. Gene structure and
polymorphism; molecular forms and expression pattern during de-
velopment. Journal of Biological Chemistry, 276(1), 464–474. https://
In this review, we have shown the chemical and cellular develop-
doi.org/10.1074/jbc.M0063 08200
ment of inflammation, the nexus of inflammation and the nervous Biddle, C. (2006). The neurobiology of the human febrile response. AANA
system in zebrafish, as well as their implications for the welfare of Journal Course, 74(2), 145–150.
specimens. Due to the high degree of conservation between teleost Bird, S., Zou, J., & Secombes, C. (2007). Advances in fish cyto-
kine biology give clues to the evolution of a complex network.
and mammals at both the molecular and genetic level, research in ze-
Current Pharmaceutical Design, 12(24), 3051–3069. https://doi.
brafish may help to overcome the limitations of mammalian models org/10.2174/138161206777947434
and contribute to the development of regenerative therapies. For Block, M. L., Zecca, L., & Hong, J. S. (2007). Microglia-mediated neu-
that reason, both inflammation studies and research that implicates rotoxicity: Uncovering the molecular mechanisms. Nature Reviews
Neuroscience, 8(1), 57–69. https://doi.org/10.1038/nrn2038
its nexus with the nervous system, such as pain-and-behaviour tests
Blomqvist, A., & Engblom, D. (2018). Neural mechanisms of inflam-
and screening tools for anti-inflammatory drugs, in zebrafish should mation-induced fever. Neuroscientist, 24(4), 381–399. https://doi.
be taken into account. Thus, it may be necessary to adapt existing org/10.1177/1073858418760481
zebrafish behaviour tests in larvae and adults to test the effective- Bollaerts, I., Van Houcke, J., Andries, L., De Groef, L., & Moons, L. (2017).
Neuroinflammation as fuel for axonal regeneration in the injured
ness of inflamed-noxious stimuli and to develop new standardized
vertebrate central nervous system. Mediators of Inflammation, 2017,
protocols to provide support for further research. 1–14. https://doi.org/10.1155/2017/9478542
Bordés Gonzalez, R., Martínez Beltrán, M., García Olivares, E., & Guisado
AC K N OW L E D G E M E N T S Barrilao, R.(1994). El proceso inflamatorio. Revista De Enfermeria, 4,
This work was supported by the MINECO co-funded by the European 11–21.
Brodde, O. E., Bruck, H., Leineweber, K., & Seyfarth, T. (2001). Presence,
Regional Development Funds (ERDF/FEDER) (grant no. AGL2017-
distribution and physiological function of adrenergic and muscarinic
88370-C3-1-R) and Fundación Seneca de la Región de Murcia (Grupo receptor subtypes in the human heart. Basic Research in Cardiology,
de Excelencia grant no. 19883/GERM/15). 96(6), 528–538. https://doi.org/10.1007/s003950170003
Broom, D. M. (2001). The evolution of pain. Vlaams Diergeneeskundig
Tijdschrift, 70, 17–21.
CO N FL I C T O F I N T E R E S T
Bruce, A. (1913). Vaso-dilator axon-reflexes. Quarterly Journal of
The authors declare no conflict of interests. Experimental Physiology, 6, 339–354. https://doi.org/10.1113/expph
ysiol.1913.sp000144
DATA AVA I L A B I L I T Y S TAT E M E N T Burhans, M., Hagman, D., Kuzma, J., Schmidt, K., & Kratz, M. (2019).
Contribution of adipose tissue inflammation to the development of
Data sharing is not applicable to this article as no new data were cre-
type 2 diabetes mellitus. Physiology & Behavior, 9(1), 1–58. https://
ated or analysed in this study. doi.org/10.1002/cphy.c170040
Calder, P. C., Ahluwalia, N., Albers, R., Bosco, N., Bourdet-Sicard, R.,
ORCID Haller, D., Holgate, S. T., Jönsson, L. S., Latulippe, M. E., Marcos, A.,
Moreines, J., M'Rini, C., Müller, M., Pawelec, G., van Neerven, R.,
María Ángeles Esteban https://orcid.org/0000-0002-6264-1458
Watzl, B., & Zhao, J. (2013). A consideration of biomarkers to be used
for evaluation of inflammation in human nutritional studies. British
REFERENCES Journal of Nutrition, 109(S1), S1–S34. https://doi.org/10.1017/S0007
Abbas, A., Lichtman, A., & Pillai, S. (2017). Cellular and molecular immunol- 114512005119
ogy, 9th ed(1–608). Philadelphia, USA: Elsevier. Calixto, J. B., Otuki, M. F., & Santos, A. R. S. (2003). Anti-inflammatory
Al-Soudi, A., Kaaij, M. H., & Tas, S. W. (2017). Endothelial cells: From innocent compounds of plant origin. Part I. Action on arachidonic acid path-
bystanders to active participants in immune responses. Autoimmunity way, nitric oxide and nuclear Factor κ B (NF-κB). Planta Medica,
Reviews, 16(9), 951–962. https://doi.org/10.1016/j.autrev.2017.07.008 69(11), 973–983. https://doi.org/10.1055/s-2003-45141
Andersson, U., & Tracey, K. J. (2012). Reflex principles of immunological Candel, S., de Oliveira, S., López-Muñoz, A., García-Moreno, D., Espín-
homeostasis. Annual Review of Immunology, 30(1), 313–335. https:// Palazón, R., Tyrkalska, S. D., & Mulero, V. (2014). Tnfα signal-
doi.org/10.1146/annurev-immunol-020711-075015 ing through Tnfr2 protects skin against oxidative stress-induced
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      13

inflammation. PLoS Biology, 12(5), e1001855. https://doi.org/10.1371/ testing of environmental chemicals. Toxicological Sciences, 95(1), 5–
journal.pbio.1001855 12. https://doi.org/10.1093/toxsci/kfl103
Chakraborty, C., Hsu, C., Wen, Z., Lin, C., & Agoramoorthy, G. (2009). Dovi, J. V., He, L., & Dipietro, L. A. (2002). Accelerated wound closure in
Zebrafish: A complete animal model for in vivo drug discovery and neutrophil-depleted mice. Journal of Leukocyte Biology, 73, 448–455.
development. Current Drug Metabolism, 10(2), 116–124. https://doi. Duan, J., Liang, S., Yu, Y., Li, Y., Wang, L., Wu, Z., Chen, Y., Miller, M.
org/10.2174/138920009787522197 R., & Sun, Z. (2018). Inflammation–coagulation response and throm-
Chandrasekharan, J. A., & Sharma-Wali, N. (2015). Lipoxins: Nature’s botic effects induced by silica nanoparticles in zebrafish embryos.
way to resolve inflammation. Journal of Inflammation Research, 8, Nanotoxicology, 12(5), 470–484. https://doi.org/10.1080/17435
181–192. https://doi.org/10.2147/JIR.S90380 390.2018.1461267
Chaves-Pozo, E., Muñoz, P., López-Muñoz, A., Pelegrín, P., García Ayala, Dunlop, R., Millsopp, S., & Laming, P. (2006). Avoidance learning in gold-
A., Mulero, V., & Meseguer, J. (2005). Early innate immune response fish (Carassius auratus) and trout (Oncorhynchus mykiss) and implica-
and redistribution of inflammatory cells in the bony fish gilthead tions for pain perception. Applied Animal Behaviour Science, 97(2–4),
seabream experimentally infected with Vibrio anguillarum. Cell and 255–271. https://doi.org/10.1016/j.applanim.2005.06.018
Tissue Research, 320(1), 61–68. https://doi.org/10.1007/s0044 Duvigneau, J. C., Luís, A., Gorman, A. M., Samali, A., Kaltenecker, D.,
1-004-1063-7 Moriggl, R., & Kozlov, A. V. (2019). Crosstalk between inflamma-
Chiu, I. M., Heesters, B. A., Ghasemlou, N., Von Hehn, C. A., Zhao, F., tory mediators and endoplasmic reticulum stress in liver diseases.
Tran, J., & Woolf, C. J. (2013). Bacteria activate sensory neurons that Cytokine, 124, 1–10. https://doi.org/10.1016/j.cyto.2018.10.018
modulate pain and inflammation. Nature, 501(7465), 52–57. https:// Ellett, F., Elks, P. M., Robertson, A. L., Ogryzko, N. V., & Renshaw, S. A.
doi.org/10.1038/nature12479.Bacteria (2015). Defining the phenotype of neutrophils following reverse
Cole, H. W., Brown, C. E., Magee, D. E., Magee, C., Roudebush, R. E., migration in zebrafish. Journal of Leukocyte Biology, 98(6), 975–981.
& Bryant, H. U. (1995). Serotonin-induced paw edema in the rat: https://doi.org/10.1189/jlb.3ma0315-105r
Pharmacological profile. General Pharmacology, 26(2), 431–436. Esteban, M. A. (2012). An overview of the immunological de-
https://doi.org/10.1016/0306-3623(94)00180 -U fenses in fish skin. ISRN Immunology, 2012, 1–29. https://doi.
Colloca, L., Ludman, T., Bouhassira, D., Baron, R., Dickenson, A. H., org/10.5402/2012/853470
Yarnitsky, D., Freeman, R., Truini, A., Attal, N., Finnerup, N. B., Etemadi, N., Chopin, M., Anderton, H., Tanzer, M. C., Rickard, J. A.,
Eccleston, C., Kalso, E., Bennett, D. L., Dworkin, R. H., & Raja, S. N. Abeysekera, W., Hall, C., Spall, S. K., Wang, B., Xiong, Y., Hla, T.,
(2017). Neuropathic pain. Nature Reviews Disease Primers, 3(1), 1–45. Pitson, S. M., Bonder, C. S., Wong, W. W. L., Ernst, M., Smyth, G.
https://doi.org/10.1038/nrdp.2017.2 K., Vaux, D. L., Nutt, S. L., Nachbur, U., & Silke, J. (2015). TRAF2
Colton, C. A. (2009). Heterogeneity of microglial activation in the regulates TNF and NF-κB signalling to suppress apoptosis and skin
innate immune response in the brain. Journal of Neuroimmune inflammation independently of sphingosine kinase. Elife., 4, 1–27.
Pharmacology, 4(4), 399–418. https://doi.org/10.1007/s1148 https://doi.org/10.7554/eLife.10592
1-009-9164-4 Ezeasor, D. N., & Stokoe, W. M. (1980). A cytochemical, light and elec-
Curtright, A., Rosser, M., Goh, S., Keown, B., Wagner, E., Sharifi, J., Raible, tron microscopic study of the eosinophilic granule cells in the gut of
D. W., & Dhaka, A. (2015). Modeling nociception in zebrafish: A way the rainbow trout, Salmo gairdneri Richardson. Journal of Fish Biology,
forward for unbiased analgesic discovery. PLoS One, 10(1), 1–18. 17(6), 619–634. https://doi.org/10.1111/j.1095-8649.1980.tb027
https://doi.org/10.1371/journal.pone.0116766 95.x
Dandona, P., Aljada, A., & Bandyopadhyay, A. (2004). Inflammation the Feng, J., Zhang, C., Lischinsky, J. E., Jing, M., Zhou, J., Wang, H., & Li, Y.
link between insulin resistance. Trends in Immunology, 25(1), 4–7. (2019). A genetically encoded fluorescent sensor for rapid and spe-
https://doi.org/10.1016/J.IT.2003.10.013 cific in vivo detection of norepinephrine. Neuron, 102(4), 745–761.e8.
De Oliveira, S., Reyes-Aldasoro, C. C., Candel, S., Renshaw, S. A., Mulero, https://doi.org/10.1016/j.neuron.2019.02.037
V., & Calado, Â. (2013). Cxcl8 (IL-8) mediates neutrophil recruitment Filby, A. L., Paull, G. C., Hickmore, T. F. A., & Tyler, C. R. (2010). Unravelling
and behavior in the zebrafish inflammatory response. The Journal the neurophysiological basis of aggression in a fish model. BMC
of Immunology, 190(8), 4349–4359. https://doi.org/10.4049/jimmu Genomics, 11(1), 498. https://doi.org/10.1186/1471-2164-11-498
nol.1203266 Fontana, B. D., Franscescon, F., Rosemberg, D. B., Norton, W. H. J.,
Delamare-Deboutteville, J., Wood, D., & Barnes, A. C. (2006). Response Kalueff, A. V., & Parker, M. O. (2019). Zebrafish models for at-
and function of cutaneous mucosal and serum antibodies in barra- tention deficit hyperactivity disorder (ADHD). Neuroscience &
mundi (Lates calcarifer) acclimated in seawater and freshwater. Fish Biobehavioral Reviews, 100, 9–18. https://doi.org/10.1016/j.neubi
and Shellfish Immunology, 21(1), 92–101. https://doi.org/10.1016/j. orev.2019.02.009
fsi.2005.10.005 Forlenza, M., Fink, I. R., Raes, G., & Wiegertjes, G. F. (2011). Heterogeneity
Deutschman, C. S., & Tracey, K. J. (2014). Sepsis: Current dogma and new of macrophage activation in fish. Developmental and Comparative
perspectives. Immunity, 40(4), 463–475. https://doi.org/10.1016/j. Immunology, 35(12), 1246–1255. https://doi.org/10.1016/j.
immuni.2014.04.001 dci.2011.03.008
DeVries, M. E., Kelvin, A. A., Xu, L., Ran, L., Robinson, J., & Kelvin, D. J. Funk, C. D. (2001). Prostaglandins and leukotrienes: Advances in
(2006). Defining the origins and evolution of the chemokine/chemo- eicosanoid biology. Science, 294(5548), 1871–1875. https://doi.
kine receptor system. The Journal of Immunology, 176(1), 401–415. org/10.1126/science.294.5548.1871
https://doi.org/10.4049/jimmunol.176.1.401 García-Moreno, D., Tyrkalska, S. D., Valera-Pérez, A., Gómez-Abenza,
Dinarello, C. A. (2015). The history of fever, leukocytic pyrogen and in- E., Pérez-Oliva, A. B., & Mulero, V. (2019). The zebrafish: A research
terleukin-1. Temperature, 2(1), 8–16. https://doi.org/10.1080/23328 model to understand the evolution of vertebrate immunity. Fish
940.2015.1017086 and Shellfish Immunology, 90, 215–222. https://doi.org/10.1016/j.
Diotel, N., Vaillant, C., Gueguen, M.-M., Mironov, S., Anglade, I., Servili, fsi.2019.04.067
A., Pellegrini, E., & Kah, O. (2010). Cxcr4 and Cxcl12 expression in Geiger, B. M., Gras-Miralles, B., Ziogas, D. C., Karagiannis, A. K. A., Zhen,
radial glial cells of the brain of adult zebrafish. Journal of Comparative A., Fraenkel, P., & Kokkotou, E. (2013). Intestinal upregulation of
Neurology, 518(24), 4855–4876. https://doi.org/10.1002/cne.22492 melanin-concentrating hormone in TNBS-induced enterocolitis in
Dix, D. J., Houck, K. A., Martin, M. T., Richard, A. M., Setzer, R. W., & adult zebrafish. PLoS One, 8(12), 1–9. https://doi.org/10.1371/journ
Kavlock, R. J. (2007). The toxcast program for prioritizing toxicity al.pone.0083194
|
14       CAMPOS-SÁNCHEZ ANd ESTEBAN

Gonzalez, S., Huising, M., Stakauskas, R., Forlenza, M., Imitola, J., Raddassi, K., Park, K. I., Mueller, F. J., Nieto, M., Teng, Y. D., &
Lidyverburgvankemenade, B., Buchmann, K., Nielsen, M., & Wiegertjes, Khoury, S. J. (2004). Directed migration of neural stem cells to sites
G. (2007). Real-time gene expression analysis in carp (Cyprinus carpio of CNS injury by the stromal cell-derived factor 1α/CXC chemokine
L.) skin: Inflammatory responses to injury mimicking infection with ec- receptor 4 pathway. Proceedings of the National Academy of Sciences
toparasites. Developmental and Comparative Immunology, 31(3), 244– of the United States of America, 101(52), 18117–18122. https://doi.
254. https://doi.org/10.1016/j.dci.2006.06.010 org/10.1073/pnas.0408258102
Gonzalez, T., Yuan, L., Boswell, M., Boswell, W., Medrano, G., Inoue, T., Abe, C., Kohro, T., Tanaka, S., Huang, L., Yao, J., Zheng, S.,
Walter, S., & Walter, R. (2018). Fluorescent light exposure in- Ye, H., Inagi, R., Stornetta, R. L., Rosin, D. L., Nangaku, M., Wada,
cites scute and prolonged immune responses in zebrafish (Danio Y., & Okusa, M. D. (2019). Non-canonical cholinergic anti-inflam-
rerio) skin. Comparative Biochemistry and Physiology Part C: matory pathway-mediated activation of peritoneal macrophages
Toxicology & Pharmacology, 208, 87–95. https://doi.org/10.1016/j. induces Hes1 and blocks ischemia/reperfusion injury in the kid-
cbpc.2017.09.009 ney. Kidney International, 95(3), 563–576. https://doi.org/10.1016/j.
Grandel, H., & Brand, M. (2013). Comparative aspects of adult neural kint.2018.09.020
stem cell activity in vertebrates. Development Genes and Evolution, Ishikawaa, T., Griffinb, K. J. P., Banerjeeb, U., & Herschman, H. R. (2007).
223(1–2), 131–147. https://doi.org/10.1007/s00427-012-0425-5 The zebrafish genome contains two inducible, functional cyclooxy-
Griffin, B. R., & Mitchell, A. J. (2007). Susceptibility of channel catfish, genase-2 genes. Biochemical and Biophysical Research Communications,
Ictalurus punctatus (Rafinesque), to Edwardsiella ictaluri challenge 352(1), 181–187. https://doi.org/10.1016/j.bbrc.2006.11.007
following copper sulphate exposure. Journal of Fish Diseases, 30(10), Jagadeeswaran, P., Gregory, M., Johnson, S., & Thankavel, B. (2000).
581–585. https://doi.org/10.1111/j.1365-2761.2007.00838.x Haemostatic screening and identification of zebrafish mutants with
Grivennikov, S. I., Greten, F. R., & Karin, M. (2010). Grivennikov SI, Greten coagulation pathway defects: An approach to identifying novel hae-
FR, Karin M. Immunity, inflammation, and cancer. Cell, 140(6), 883– mostatic genes in man. British Journal of Haematology, 110(4), 946–
899. https://doi.org/10.1016/j.cell.2010.01.025.Immunity 956. https://doi.org/10.1046/j.1365-2141.2000.02284.x
Grunwald, D., & Eisen, J. (2002). Headwaters of the zebrafish – emer- Jimi, E., Fei, H., & Nakatomi, C. (2019). Nf-κb signaling regulates phys-
gence of a new model vertebrate. Nature Reviews Genetics, 3(9), 711– iological and pathological chondrogenesis. International Journal of
717. https://doi.org/10.1038/nrg891 Molecular Sciences, 20(24), 1–11. https://doi.org/10.3390/ijms2
Gugasyan, R., Voss, A., Varigos, G., Thomas, T., Grumont, R. J., Kaur, 0246275
P., Grigoriadis, G., & Gerondakis, S. (2004). The transcription fac- Joerink, M., Ribeiro, C. M. S., Stet, R. J. M., Hermsen, T., Savelkoul, H.
tors c-rel and RelA control epidermal development and homeosta- F. J., & Wiegertjes, G. F. (2006). Head kidney-derived macrophages
sis in embryonic and adult skin via distinct mechanisms. Molecular of common carp (Cyprinuscarpio L.). show plasticity and functional
and Cellular Biology, 24(13), 5733–5745. https://doi.org/10.1128/ polarization upon differential stimulation. The Journal of Immunology,
mcb.24.13.5733-5745.2004 177(1), 61–69. https://doi.org/10.4049/jimmunol.177.1.61
Hall, C., Flores, M. V., Chien, A., Davidson, A., Crosier, K., & Crosier, P. Joerink, M., Savelkoul, H. F. J., & Wiegertjes, G. F. (2006). Evolutionary
(2009). Transgenic zebrafish reporter lines reveal conserved Toll- conservation of alternative activation of macrophages: Structural
like receptor signaling potential in embryonic myeloid leukocytes and functional characterization of arginase 1 and 2 in carp (Cyprinus
and adult immune cell lineages. Journal of Leukocyte Biology, 85(5), carpio L.). Molecular Immunology, 43(8), 1116–1128. https://doi.
751–765. https://doi.org/10.1189/jlb.0708405 org/10.1016/j.molimm.2005.07.022
Hall, C., Flores, M., Storm, T., Crosier, K., & Crosier, P. (2007). The ze- Katayama, Y., Battista, M., Kao, W. M., Hidalgo, A., Peired, A. J., Thomas,
brafish lysozyme C promoter drives myeloid-specific expression S. A., & Frenette, P. S. (2006). Signals from the sympathetic nervous
in transgenic fish. BMC Developmental Biology, 7, 1–17. https://doi. system regulate hematopoietic stem cell egress from bone marrow.
org/10.1186/1471-213X-7-42 Cell, 124(2), 407–421. https://doi.org/10.1016/j.cell.2005.10.041
Harris, R. A. (2014). Spatial, temporal, and functional aspects of macro- Keightley, M. C., Wang, C. H., Pazhakh, V., & Lieschke, G. J. (2014).
phages during “The good, the bad, and the ugly” phases of inflam- Delineating the roles of neutrophils and macrophages in zebrafish re-
mation. Frontiers in Immunology, 5, 1–5. https://doi.org/10.3389/ generation models. International Journal of Biochemistry and Cell Biology,
fimmu.2014.00612 56, 92–106. https://doi.org/10.1016/j.biocel.2014.07.010
Harvie, E. A., & Huttenlocher, A. (2015). Neutrophils in host defense: Key, B., Arlinghaus, R., Browman, H. I., Cooke, S. J., Cowx, I. G., Diggles,
New insights from zebrafish. Journal of Leukocyte Biology, 98(4), 523– B. K., Watson, C. A. (2017). Problems with equating thermal pref-
537. https://doi.org/10.1189/jlb.4mr1114-524r erence with ‘emotional fever’ and sentience: Comment on ‘fish can
Hawiger, J., & Zienkiewicz, J. (2019). Decoding inflammation, its causes, show emotional fever: Stress-induced hyperthermia in zebrafish’ by
genomic responses, and emerging countermeasures. Scandinavian rey et al (2015). Proceedings of the Royal Society B: Biological Sciences,
Journal of Immunology, 90(6), 1–32. https://doi.org/10.1111/sji.12812 284(1847), 1–3. https://doi.org/10.1098/rspb.2016.0681
Herbomel, P., Thisse, B., & Thisse, C. (1999). Ontogeny and behaviour of Kiba, T. (2002). The role of the autonomic nervous system in liver re-
early macrophages in the zebrafish embryo. Development, 126(17), generation and apoptosis – Recent developments. Digestion, 66(2),
3735–3745. 79–88. https://doi.org/10.1159/000065594
Herbomel, P., Thisse, B., & Thisse, C. (2001). Zebrafish early macrophages Kim, S., Radhakrishnan, U. P., Rajpurohit, S. K., Kulkarni, V., &
colonize cephalic mesenchyme and developing brain, retina, and Jagadeeswaran, P. (2010). Vivo-Morpholino knockdown of αIIb:
epidermis through a M-CSF receptor-dependent invasive process. A novel approach to inhibit thrombocyte function in adult zebraf-
Developmental Biology, 238(2), 274–288. https://doi.org/10.1006/ ish. Blood Cells, Molecules, and Diseases, 44(3), 169–174. https://doi.
dbio.2001.0393 org/10.1016/j.bcmd.2009.12.004
Huang, S.-Y., Feng, C.-W., Hung, H.-C., Chakraborty, C., Chen, C.-H., King, M., & Wood, W. J. (1957). Studies on the pathogenesis of fever. IV.
Chen, W.-F., Jean, Y.-H., Wang, H.-M., Sung, C.-S., Sun, Y.-M., Wu, The site of action of leucocytic and circulating endogenous pyrogen.
C.-Y., Liu, W., Hsiao, C.-D., & Wen, Z.-H. (2014). A novel zebrafish Journal of Experimental Medicine, 107(2), 291–303.
model to provide mechanistic insights into the inflammatory events Kizil, C., Kyritsis, N., & Brand, M. (2015). Effects of inflammation on stem
in carrageenan-induced abdominal edema. PLoS One, 9(8), e104414. cells: Together they strive? EMBO Reports, 16(4), 416–426. https://
https://doi.org/10.1371/journal.pone.0104414 doi.org/10.15252/embr.201439702
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      15

Kolaczkowska, E., & Kubes, P. (2013). Neutrophil recruitment and func- Martin, P., D’Souza, D., Martin, J., Grose, R., Cooper, L., Maki, R., &
tion in health and inflammation. Nature Reviews Immunology, 13(3), McKercher, S. (2003). Wound healing in the PU.1 null mouse-tissue
159–175. https://doi.org/10.1038/nri3399 repair is not dependent on inflammatory cells. Current Biology, 13,
Korkmaz, B., Horwitz, M., Jenne, D., & Gauthier, F.(2010). Neutrophil 1122–1128. https://doi.org/10.1016/S0960 -9822(03)00396-8
elastase, proteinase 3, and cathepsin G as therapeutic targets in Maryanovich, M., Zaltsman, Y., Ruggiero, A., Goldman, A., Shachnai, L.,
human diseases. Pharmacology Reviews, 62(4), 726–759. https://doi. Zaidman, S. L., Porat, Z., Karin Golan, K., Tsvee Lapidot, T., & Gross,
org/10.1124/pr.110.002733.726 A. (2015). An MTCH2 pathway repressing mitochondria metabolism
Kunst, M., Laurell, E., Mokayes, N., Kramer, A., Kubo, F., Fernandes, A. regulates haematopoietic stem cell fate. Nature communications, 6,
M., Förster, D., Dal Maschio, M., & Baier, H. (2019). A cellular-resolu- 7901. https://doi.org/10.1038/ncomms8901
tion atlas of the larval zebrafish brain. Neuron, 103(1), 21–38. https:// Matsumoto, R., Dainichi, T., Tsuchiya, S., Nomura, T., Kitoh, A., Hayden,
doi.org/10.1016/j.neuron.2019.04.034 M. S., Ishii, K. J., Tanaka, M., Honda, T., Egawa, G., Otsuka, A.,
Kupper, T. S., & Fuhlbrigge, R. C. (2004). Immune surveillance in the skin: Nakajima, S., Sakurai, K., Nakano, Y., Kobayashi, T., Sugimoto, Y., &
Mechanisms and clinical consequences. Nature Reviews Immunology, Kabashima, K. (2018). Epithelial TRAF6 drives IL-17 – mediated pso-
4(3), 211–222. https://doi.org/10.1038/nri1310 riatic inflammation Find the latest version : Epithelial TRAF6 drives
Kuri, P., Ellwanger, K., Kufer, T. A., Leptin, M., & Bajoghli, B. (2017). A IL-17 – mediated psoriatic inflammation. JCI Insight, 3(15), e121175.
high-sensitivity bi-directional reporter to monitor NF-κB activity in https://doi.org/10.1172/jci.insight.121175
cell culture and zebrafish in real time. Journal of Cell Science, 130(3), Maximino, C., de Brito, T. M., da Silva Batista, A. W., Herculano, A. M.,
648–657. https://doi.org/10.1242/jcs.196485 Morato, S., & Gouveia, A. (2010). Measuring anxiety in zebrafish: A
Landgraf, R., & Neumann, I. D. (2004). Vasopressin and oxytocin release critical review. Behavioural Brain Research, 214(2), 157–171. https://
within the brain: A dynamic concept of multiple and variable modes doi.org/10.1016/j.bbr.2010.05.031
of neuropeptide communication. Frontiers in Neuroendocrinology, Medzhitov, R. (2008). Origin and physiological roles of inflammation.
25(3–4), 150–176. https://doi.org/10.1016/j.yfrne.2004.05.001 Nature, 454(7203), 428–435. https://doi.org/10.1038/nature07201
Larsen, G. L., & Henson, P. M. (1983). Mediators of inflammation. Meijer, A. H., & Spaink, P. H.(2011). Host-pathogen interactions made
Seminars in Arthritis and Rheumatism, 14(4), 247–262. https://doi. transparent with the zebrafish model. Current Drug Targets, 12(7),
org/10.1016/0049-0172(85)90044-7 1000–1017. https://doi.org/10.2174/138945011795677809
Leibovich, S. J., & Ross, R. (1975). The role of the macrophage in wound Meijer, A. H., van der Sar, A. M., Cunha, C., Lamers, G. E. M., Laplante,
repair. A study with hydrocortisone and antimacrophage serum. M. A., Kikuta, H., Bitter, W., Becker, T. S., & Spaink, H. P. (2008).
American Journal of Pathology, 78(1), 71–99. Identification and real-time imaging of a myc-expressing neutrophil
Ley, K., Laudanna, C., Cybulsky, M. I., & Nourshargh, S. (2007). Getting to population involved in inflammation and mycobacterial granuloma
the site of inflammation: The leukocyte adhesion cascade updated. formation in zebrafish. Developmental and Comparative Immunology,
Nature Reviews Immunology, 7(9), 678–689. https://doi.org/10.1038/ 32(1), 36–49. https://doi.org/10.1016/j.dci.2007.04.003
nri2156 Meseguer, J., Lopez Ruiz, A., & Garcia Ayala, A. (1995). Reticulo-
Liu, T., Zhang, L., Joo, D., & Sun, S. C. (2017). NF-κB signaling in inflamma- endothelial stroma of the head-kidney from the seawater teleost
tion. Signal Transduction and Targeted Therapy, 2, 17023. https://doi. gilthead seabream (Sparus aurata L.): An ultrastructural and cyto-
org/10.1038/sigtrans.2017.23 chemical study. The Anatomical Record, 241(3), 303–309. https://doi.
Lopez-Luna, J., Al-Jubouri, Q., Al-Nuaimy, W., & Sneddon, L. U. (2017a). org/10.1002/ar.1092410303
Impact of stress, fear and anxiety on the nociceptive responses of Meseguer, J., López-Ruiz, A., & Esteban, M. A. (1994). Cytochemical
larval zebrafish. PLoS One, 12(8), 1–17. https://doi.org/10.1371/journ characterization of leucocytes from the seawater teleost, gilthead
al.pone.0181010 seabream (Sparus aurata L.). Histochemistry, 102(1), 37–44. https://
Lopez-Luna, J., Al-Jubouri, Q., Al-Nuaimy, W., & Sneddon, L. U. (2017b). doi.org/10.1007/BF00271047
Reduction in activity by noxious chemical stimulation is ameliorated Mettam, J. J., Oulton, L. J., McCrohan, C. R., & Sneddon, L. U. (2011).
by immersion in analgesic drugs in zebrafish. Journal of Experimental The efficacy of three types of analgesic drugs in reducing pain in
Biology, 220(8), 1451–1458. https://doi.org/10.1242/jeb.146969 the rainbow trout, Oncorhynchus Mykiss. Applied Animal Behaviour
MacGlashan, D. (2003). Histamine: A mediator of inflammation. Journal Science, 133(3–4), 265–274. https://doi.org/10.1016/j.appla
of Allergy and Clinical Immunology, 112(4), S53–S59. https://doi. nim.2011.06.009
org/10.1016/S0091-6749(03)01877-3 Milton, A. S., & Wendlandt, S. (1970). A possible role for prostaglandin
Machluf, Y., Gutnick, A., & Levkowitz, G. (2011). Development of the ze- E1 as a modulator for temperature regulation in the central nervous
brafish hypothalamus. Annals of the New York Academy of Sciences, system of the cat. The Journal of Physiology, 207(2), 76P–77P.
1220(1), 93–105. https://doi.org/10.1111/j.1749-6632.2010.05945.x Morcillo, P., Cordero, H., Meseguer, J., Esteban, M. T., & Cuesta, A. (2015).
Magalhães, F. E. A., De Sousa, C. Á. P. B., Santos, S. A. A. R., Menezes, Toxicological in vitro effects of heavy metals on gilthead seabream
R. B., Batista, F. L. A., Abreu, Â. O., & Campos, A. R. (2017). Adult (Sparus aurata L.) head-kidney leucocytes. Toxicology in Vitro, 30(1),
zebrafish (Danio rerio): An alternative behavioral model of forma- 412–420. https://doi.org/10.1016/j.tiv.2015.09.021
lin-induced nociception. Zebrafish, 14(5), 422–429. https://doi. Mottaz, H., Schönenberger, R., Fischer, S., Eggen, R. I. L., Schirmer, K., &
org/10.1089/zeb.2017.1436 Groh, K. J. (2017). Dose-dependent effects of morphine on lipopoly-
Malafoglia, V., Bryant, B., Raffaeli, W., Giordano, A., & Bellipanni, G. saccharide (LPS)-induced inflammation, and involvement of multix-
(2013). The zebrafish as a model for nociception studies. Journal of enobiotic resistance (MXR) transporters in LPS efflux in teleost fish.
Cellular Physiology, 228(10), 1956–1966. https://doi.org/10.1002/ Environmental Pollution, 221, 105–115. https://doi.org/10.1016/j.
jcp.24379 envpol.2016.11.046
Malafoglia, V., Colasanti, M., Raffaeli, W., Balciunas, D., Giordano, A., & Mulero, M. C., Wang, V. Y. F., Huxford, T., & Ghosh, G. (2019). Genome
Bellipanni, G. (2014). Extreme thermal noxious stimuli induce pain reading by the NF-κB transcription factors. Nucleic Acids Research,
responses in zebrafish larvae. Journal of Cellular Physiology, 229(3), 47(19), 9967–9989. https://doi.org/10.1093/nar/gkz739
300–308. https://doi.org/10.1002/jcp.24447 Narayanan, N. S., Guarnieri, D. J., & DiLeone, R. J. (2010). Metabolic
Markakis, E. A. (2002). Development of the neuroendocrine hypothal- hormones, dopamine circuits, and feeding. Frontiers in
amus. Frontiers in Neuroendocrinology, 23(3), 257–291. https://doi. Neuroendocrinology, 31(1), 104–112. https://doi.org/10.1016/j.
org/10.1016/S0091-3022(02)00003-1 yfrne.2009.10.004
|
16       CAMPOS-SÁNCHEZ ANd ESTEBAN

Nathan, C. (2002). Points of control in inflammation. Nature, 420(19/26), cells. Journal of Neuroscience, 27(40), 10714–10721. https://doi.
846–852. https://doi.org/10.1038/nature01320 org/10.1523/JNEUROSCI.1922-07.2007
Nathan, C. (2006). Neutrophils and immunity: Challenges and oppor- Postlethwait, J. H., Woods, I. G., Ngo-Hazelett, P., Yan, Y. L., Kelly, P. D.,
tunities. Nature Reviews Immunology, 6(3), 173–182. https://doi. Chu, F., & Talbot, W. S. (2000). Zebrafish comparative genomics and
org/10.1038/nri1785 the origins of vertebrate chromosomes. Genome Research, 10(12),
Nathan, C., & Ding, A. (2010). Nonresolving Inflammation. Cell, 140(6), 1890–1902. https://doi.org/10.1101/gr.164800
871–882. https://doi.org/10.1016/j.cell.2010.02.029 Powell, C., Grant, A. R., Cornblath, E., & Goldman, D. (2013). Analysis
Nguyen-Chi, M., Laplace-Builhe, B., Travnickova, J., Luz-Crawford, P., of dna methylation reveals a partial reprogramming of the müller
Tejedor, G., Phan, Q. T., Duroux-Richard, I., Levraud, J.-P., Kissa, K., glia genome during retina regeneration. Proceedings of the National
Lutfalla, G., Jorgensen, C., & Djouad, F. (2015). Identification of po- Academy of Sciences of the United States of America, 110(49), 19814–
larized macrophage subsets in zebrafish. Elife, 4, 1–14. https://doi. 19819. https://doi.org/10.1073/pnas.13120 09110
org/10.7554/eLife.07288 Press, C. M. L., & Evensen, O. (1999). The morphology of the immune
Nomiyama, H., Hieshima, K., Osada, N., Kato-Unoki, Y., Otsuka-Ono, K., system in teleost fishes. Fish and Shellfish Immunology, 9(4), 309–318.
Takegawa, S., Izawa, T., Yoshizawa, A., Kikuchi, Y., Tanase, S., Miura, https://doi.org/10.1006/fsim.1998.0181
R., Kusuda, J., Nakao, M., & Yoshie, O. (2008). Extensive expan- Prober, D. A., Zimmerman, S., Myers, B. R., McDermott, B. M., Kim, S.-H.,
sion and diversification of the chemokine gene family in zebrafish: Caron, S., Rihel, J., Solnica-Krezel, L., Julius, D., Hudspeth, A. J., &
Identification of a novel chemokine subfamily CX. BMC Genomics, 9, Schier, A. F. (2008). Zebrafish TRPA1 channels are required for che-
1–19. https://doi.org/10.1186/1471-2164-9-222 mosensation but not for thermosensation or mechanosensory hair
Norton, W., & Bally-Cuif, L. (2010). Adult zebrafish as a model organism cell function. Journal of Neuroscience, 28(40), 10102–10110. https://
for behavioural genetics. BMC Neuroscience, 11(1), 11–90. https:// doi.org/10.1523/JNEUROSCI.2740-08.2008
doi.org/10.1186/1471-2202-11-90 Radić, Z., Pickering, N. A., Vellom, D. C., Camp, S., & Taylor, P. (1993).
Oehlers, S. H. B., Flores, M. V., Hall, C. J., O’Toole, R., Swift, S., Crosier, Three distinct domains in the cholinesterase molecule confer selec-
K. E., & Crosier, P. S. (2010). Expression of zebrafish cxcl8 (interleu- tivity for acetyl- and butyrylcholinesterase inhibitors. Biochemistry,
kin-8) and its receptors during development and in response to im- 32(45), 12074–12084. https://doi.org/10.1021/bi00096a018
mune stimulation. Developmental and Comparative Immunology, 34(3), Rey, S., Huntingford, F. A., Boltaña, S., Vargas, R., Knowles, T. G., &
352–359. https://doi.org/10.1016/j.dci.2009.11.007 Mackenzie, S. (2015). Fish can show emotional fever: Stress-induced
Oehlers, S. H., Flores, M. V., Okuda, K. S., Hall, C. J., Crosier, K. E., & hyperthermia in zebrafish. Proceedings of the Royal Society B: Biological
Crosier, P. S. (2011). A chemical enterocolitis model in zebrafish lar- Sciences, 282, 20152266, https://doi.org/10.1098/rspb.2015.2266
vae that is dependent on microbiota and responsive to pharmacolog- Rink, E., & Wullimann, M. F. (2002). Development of the catecholamin-
ical agents. Developmental Dynamics, 240(1), 288–298. https://doi. ergic system in the early zebrafish brain: An immunohistochemical
org/10.1002/dvdy.22519 study. Developmental Brain Research, 137(1), 89–100. https://doi.
Oosterhof, N., Boddeke, E., & van Ham, T. J. (2015). Immune cell dynam- org/10.1016/S0165-3806(02)00354-1
ics in the CNS: Learning from the zebrafish. Glia, 63(5), 719–735. Rombout, J. H. W. M., Taverne, N., van de Kamp, M., & Taverne-Thiele,
https://doi.org/10.1002/glia.22780 A. J. (1993). Differences in mucus and serum immunoglobulin of
Panula, P., Chen, Y. C., Priyadarshini, M., Kudo, H., Semenova, S., Sundvik, carp (Cyprinus carpio L.). Developmental and Comparative Immunology,
M., & Sallinen, V. (2010). The comparative neuroanatomy and neuro- 17(4), 309–317. https://doi.org/10.1016/0145-305X(93)90003-9
chemistry of zebrafish CNS systems of relevance to human neuro- Rose, J. D. (2002). The neurobehavioral nature of fishes and the ques-
psychiatric diseases. Neurobiology of Disease, 40(1), 46–57. https:// tion of awareness and pain. Reviews in Fisheries Science, 10(1), 1–38.
doi.org/10.1016/j.nbd.2010.05.010 https://doi.org/10.1080/20026 491051668
Parkhurst, M. R., & Saltzman, W. M. (1994). Leukocytes migrate Salinas, I., Zhang, Y. A., & Sunyer, J. O. (2011). Mucosal immunoglobulins
through three-dimensional gels of midcycle cervical mucus. and B cells of teleost fish. Developmental and Comparative Immunology,
Cellular Immunology, 156(1), 77–94. https://doi.org/10.1006/ 35(12), 1346–1365. https://doi.org/10.1016/j.dci.2011.11.009
cimm.1994.1154 Sarkar, C., Basub, B., Chakrobortya, D., Dasgupta, P. S., & Basu, S.
Pavlov, V. A., & Tracey, K. J. (2012). The vagus nerve and the inflamma- (2010). The immunoregulatory role of dopamine: An update. Brain,
tory reflex. Nature Revue Endocrinology, 8(12), 743–754. https://doi. Behavior, and Immunity, 24(4), 525–528. https://doi.org/10.1016/j.
org/10.1038/nrendo.2012.189.The bbi.2009.10.015
Perry, V. H. (2012). Innate inflammation in Parkinson’s disease. Cold Spring Savan, R., & Sakai, M. (2006). Genomics of fish cytokines. Comparative
Harbor Perspectives in Medicine, 2(9), 1–11. https://doi.org/10.1101/ Biochemistry and Physiology Part D: Genomics and Proteomics, 1(1),
cshperspect.a009373 89–101. https://doi.org/10.1016/j.cbd.2005.08.005
Perry, V. H., Cunningham, C., & Holmes, C. (2007). Systemic infections Scheiermann, C., Kunisaki, Y., Lucas, D., Chow, A., Jang, J., Zhang, D.,
and inflammation affect chronic neurodegeneration. Nature Reviews Hashimoto, D., Merad, M., & Frenette, P. S.(2013). Adrenergic nerves
Immunology, 7(2), 161–167. https://doi.org/10.1038/nri2015 govern circadian leukocyte recruitment to tissues. Immunity, 37(2), 290–
Petrie, T. A., Strand, N. S., Yang, C.-T., Rabinowitz, J. S., & Moon, R. T. 301. https://doi.org/10.1016/j.immuni.2012.05.021.ADRENERGIC
(2015). Macrophages modulate adult zebrafish tail fin regeneration. Schweitzer, J., & Driever, W. (2009). Development of the dopamine sys-
Development, 142(2), 406. https://doi.org/10.1242/dev.120642 tems in Zebrafish. Advances in Experimental Medicine and Biology, 651,
Pinho-Ribeiro, F. A., Verri, W. A. Jr, & Chiu, I. M. (2017). Nociceptor sen- 1–14. https://doi.org/10.1007/978-1-4419-0322-8_1
sory neuron-immune interactions in pain and inflammation. Trends Secombes, C. J., Wang, T., & Bird, S. (2011). The interleukins of fish.
in Immunology, 38(1), 5–19. https://doi.org/10.1016/j.it.2016.10.001 Developmental and Comparative Immunology, 35(12), 1336–1345.
Pongratz, G., & Straub, R. H. (2013). Role of peripheral nerve fibres in acute https://doi.org/10.1016/j.dci.2011.05.001
and chronic inflammation in arthritis. Nature Reviews Rheumatology, Sepulcre, M. P., Alcaraz-Pérez, F., López-Muñoz, A., Roca, F. J., Meseguer,
9(2), 117–126. https://doi.org/10.1038/nrrheum.2012.181 J., Cayuela, M. L., & Mulero, V. (2009). Evolution of lipopolysaccha-
Ponomarev, E. D., Maresz, K., Tan, Y., & Dittel, B. N. (2007). CNS-derived ride (LPS) recognition and signaling: Fish TLR4 does not recognize LPS
interleukin-4 is essential for the regulation of autoimmune inflam- and negatively regulates NF-κB activation. The Journal of Immunology,
mation and induces a state of alternative activation in microglial 182(4), 1836–1845. https://doi.org/10.4049/jimmunol.0801755
CAMPOS-SÁNCHEZ ANd ESTEBAN |
      17

Sepulcre, M., Pelegrín, P., Mulero, V., & Meseguer, J. (2002). Wang, T., & Secombes, C. J. (2013). The cytokine networks of adaptive
Characterisation of gilthead seabream acidophilic granulocytes by immunity in fish. Fish and Shellfish Immunology, 35(6), 1703–1718.
a monoclonal antibody unequivocally points to their involvement in https://doi.org/10.1016/j.fsi.2013.08.030
fish phagocytic response. Cell and Tissue Research, 308(1), 97–102. Wiegertjes, G. F., Wentzel, A. S., Spaink, H. P., Elks, P. M., & Fink, I. R.
https://doi.org/10.1007/s00441-002-0531-1 (2016). Polarization of immune responses in fish: The ‘macrophages
Shaw, T. J., & Martin, P. (2009). Wound repair at a glance. Journal of Cell first’ point of view. Molecular Immunology, 69, 146–156. https://doi.
Science, 122(18), 3209–3213. https://doi.org/10.1242/jcs.031187 org/10.1016/j.molimm.2015.09.026
Shi, W. C., Fang, Z. B., Li, L., & Luo, L. F. (2015). Using zebrafish as the Wolman, M., & Granato, M. (2012). Behavioral genetics in larval zebraf-
model organism to understand organ regeneration. Science China ish: Learning from the young. Dev Neurobiol, 72(3), 366–372. https://
Life Sciences, 58(4), 343–351. https://doi.org/10.1007/s1142 doi.org/10.1002/dneu.20872
7-015-4838-z Woodruff, M. (2018). Pain in fish: Evidence from peripheral nociceptors
Sieger, D., Moritz, C., Ziegenhals, T., Prykhozhij, S., & Peri, F. (2012). to pallial processing. Animal Sentience, 3(21).
Long-range Ca2+ waves transmit brain-damage signals to microglia. Wüllner, U., & Klockgether, T. (2003). Inflammation in Parkinson’s dis-
Developmental Cell, 22(6), 1138–1148. https://doi.org/10.1016/j. ease. Journal of Neurology, Supplement, 250(1), 35–38. https://doi.
devcel.2012.04.012 org/10.1007/s00415-003-1107-x
Singer, A. J., & Clark, R. A. F. (1999). Cutaneous wound healing. The Yamazaki, K., & Allen, T. D. (1990). Ultrastructural morphometric study
New England Journal of Medicine, 341(10), 738–746. https://doi. of efferent nerve terminals on murine bone marrow stromal cells,
org/10.1056/NEJM199909023411006 and the recognition of a novel anatomical unit: The “neuro-reticular
Sneddon, L. U. (2002). Anatomical and electrophysiological analysis of complex”. American Journal of Anatomy, 187(3), 261–276. https://doi.
the trigeminal nerve in the rainbow trout. Onchorhynchus Mykiss. org/10.1002/aja.1001870306
Neuroscience Letters, 319(3), 167–171. Yoo, S. K., Deng, Q., Cavnar, P. J., Wu, Y. I., Hahn, K. M., & Huttenlocher,
Sneddon, L. U. (2009). Pain perception in fish: Indicators and endpoints. A. (2010). Differential regulation of protrusion and polarity by PI(3)K
ILAR Journal, 50(4), 338–342. https://doi.org/10.1093/ilar.50.4.338 during neutrophil motility in live zebrafish. Developmental Cell, 18(2),
Sneddon, L. U., Braithwaite, V. A., & Gentle, M. J. (2003). Do fishes have 226–236. https://doi.org/10.1016/j.devcel.2009.11.015
nociceptors? Evidence for the evolution of a vertebrate sensory sys- Yoo, S. K., & Huttenlocher, A. (2011). Spatiotemporal photolabeling of
tem. Proceedings of the Royal Society B: Biological Sciences, 270(1520), neutrophil trafficking during inflammation in live zebrafish. Journal
1115–1121. https://doi.org/10.1098/rspb.2003.2349 of Leukocyte Biology, 89(5), 661–667. https://doi.org/10.1189/
Sommer, F., Torraca, V., & Meijer, A. H. (2020). Chemokine receptors and jlb.1010567
phagocyte biology in zebrafish. Frontiers in Immunology, 11, 1–14. Zhang, S., & Cui, P. (2014). Complement system in zebrafish.
https://doi.org/10.3389/fimmu.2020.00325 Developmental and Comparative Immunology, 46(1), 3–10. https://doi.
Soreq, H., & Seidman, S. (2001). Acetylcholinesterase — new roles for org/10.1016/j.dci.2014.01.010
an old actor. Nature Reviews Neuroscience, 2(4), 294–302. https://doi. Zhang, Y. U., Takagi, N., Yuan, B. O., Zhou, Y., Si, N., Wang, H., Yang,
org/10.1038/35067589 J., Wei, X., Zhao, H., & Bian, B. (2019). The protection of indoleal-
Steenbergen, P. J., & Bardine, N. (2014). Antinociceptive effects of bu- kylamines from LPS-induced inflammation in zebrafish. Journal
prenorphine in zebrafish larvae: An alternative for rodent models to of Ethnopharmacology, 243, 112122. https://doi.org/10.1016/j.
study pain and nociception? Applied Animal Behaviour Science, 152, jep.2019.112122
92–99. https://doi.org/10.1016/j.applanim.2013.12.001 Zhao, X., Findly, R. C., & Dickerson, H. W. (2008). Cutaneous anti-
Sternberg, E. M. (2006). Neural regulation of innate immunity. Nature, body-secreting cells and B cells in a teleost fish. Developmental and
6(4), 318–328. Comparative Immunology, 32(5), 500–508. https://doi.org/10.1016/j.
Taylor, J. C., Dewberry, L. S., Totsch, S. K., Yessick, L. R., DeBerry, J. J., dci.2007.08.009
Watts, S. A., & Sorge, R. E. (2017). A novel zebrafish-based model Zila, I., Mokra, D., Kopincova, J., Kolomaznik, M., Javorka, M., & Calkovska,
of nociception. Physiology and Behavior, 174, 83–88. https://doi. A. (2017). Vagal-immune interactions involved in cholinergic anti-in-
org/10.1016/j.physbeh.2017.03.009 flammatory pathway. Physiological Research, 66(1998), S139–S145.
Torres-Rosas, R., Yehia, G., Peña, G., Mishra, P., del Thompson-Bonilla, M. https://doi.org/10.33549/physiolres.933671
R., Moreno-Eutimio, M. A., Arriaga-Pizano, L. A., Isibasi, A., & Ulloa, Zimmerman, B. G. (1981). Adrenergic facilitation by angiotensin: Does
L.(2014). Dopamine mediates the vagal modulation of the immune it serve a physiological function? Clinical Science, 60(4), 343–348.
system by electroacupuncture. Nature Medicine, 20(3), 291–295. https://doi.org/10.1042/cs060 0343
https://doi.org/10.1038/nm.3479 Zou, J., & Secombes, C. J. (2016). The function of fish cytokines. Biology,
Viswanathan, K., & Dhabhar, F. S. (2005). Stress-induced enhancement 5(2), 23. https://doi.org/10.3390/biology5020023
of leukocyte trafficking into sites of surgery or immune activation.
Proceedings of the National Academy of Sciences of the United States of
America, 102(16), 5808–5813. https://doi.org/10.1073/pnas.05016
How to cite this article: Campos-Sánchez JC, Esteban MÁ.
50102
Review of inflammation in fish and value of the zebrafish model.
von Hehn, C. A., Baron, R., & Woolf, C. J. (2012). Deconstructing the
neuropathic pain phenotype to reveal neural mechanisms. Neuron, J Fish Dis. 2020;00:1–17. https://doi.org/10.1111/jfd.13310
73(4), 638–652. https://doi.org/10.1016/j.neuron.2012.02.008.
Deconstruc ting

You might also like