You are on page 1of 13

Aquaculture 467 (2017) 28–40

Contents lists available at ScienceDirect

Aquaculture

journal homepage: www.elsevier.com/locate/aquaculture

Understanding fish muscle growth regulation to optimize


aquaculture production
E.J. Vélez, E. Lutfi, Sh. Azizi, M. Perelló, C. Salmerón, M. Riera-Codina, A. Ibarz, J. Fernández-Borràs, J. Blasco,
E. Capilla, I. Navarro, J. Gutiérrez ⁎
Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain

a r t i c l e i n f o a b s t r a c t

Article history: Aquaculture has become an agronomic activity with noticeable development around the world to respond to the
Received 1 February 2016 simultaneous decrease of fish captures and the increasing demand of aquatic products for human consumption.
Received in revised form 3 June 2016 However, different problems limit the development of this industry and one of those is the time required for most
Accepted 3 July 2016
of the cultured fish species to achieve economically viable the commercial size. The knowledge up to date of the
Available online 4 July 2016
regulatory systems involved in controlling growth has improved very much but, it is still necessary to devote ef-
Keywords:
forts to transform the basic information in application to fish culture production. The aim of the present review is
Muscle growth to summarize the knowledge acquired with the studies about the GH/IGF axis and other hormones regarding
Endocrine regulation their function on the regulation of fish muscle development and growth. To this end, GH and IGFs effects in mus-
GH cle cells on metabolism and development are examined, as well as the contribution of IGF-I binding proteins, IGF-
IGFs I receptors and their downstream regulated molecules like TOR and its relation with cell proliferation and differ-
Myogenesis entiation and the myogenic regulatory factors. The effect of regulatory molecules on cultured myocytes are
MRFs reviewed as well as in vivo responses, including the model of sustained and maintained swimming. Key aspects
Exercise
we consider should be further investigated to complete the scenario of the regulation of fish muscle are also
proposed.
© 2016 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
2. Endocrine regulation of fish muscle growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
2.1. Growth hormone effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
2.2. IGFs actions on growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
2.3. Metabolic effects of IGFs. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
2.4. Other endocrine regulators of muscle growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
2.4.1. Insulin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
2.4.2. Thyroid hormones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
2.4.3. Steroids and adrenergic agonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
3. Regulation of fish myogenesis by IGFs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
4. New markers of muscle growth and quality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
5. Exercise and muscle growth in fish . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
6. Perspectives on fish growth and aquaculture. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37

⁎ Corresponding author at: Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain.
E-mail address: jgutierrez@ub.edu (J. Gutiérrez).

http://dx.doi.org/10.1016/j.aquaculture.2016.07.004
0044-8486/© 2016 Elsevier B.V. All rights reserved.
E.J. Vélez et al. / Aquaculture 467 (2017) 28–40 29

1. Introduction 2. Endocrine regulation of fish muscle growth

The double role of muscle, mechanic and metabolic, requires specific 2.1. Growth hormone effects
regulatory systems that furthermore will change as a function of age, re-
productive stage and during annual cycles. It should be also considered The GH/IGF axis is the main regulator of growth in vertebrates; its
that under the name of muscle there is a big variety of tissues role in fish has been demonstrated in many species and previously
performing different functions including, for instance, the slow and reviewed (Company et al., 2001; Reinecke et al., 2005; Reinecke,
fast skeletal muscles; although most of the information available refers 2010; Reindl and Sheridan, 2012; Fuentes et al., 2013). GH exerts signif-
to fast skeletal muscle that is also the most interesting part of body fish icant metabolic effects, as the regulation of muscle protein synthesis,
in terms of aquaculture product (Mommsen, 2001). determining an increase on growth by both hyperplasia and hypertro-
Among the regulatory systems, hormones play an essential role phy. There are many references describing the effects of GH treatment
through a systemic action that will affect all target tissues. But there on muscle protein synthesis, muscle growth acceleration, hyperplasia,
are also the local actions that in the case of the growth hormone (GH) muscle accretion, etc. (reviewed in: Moomsen and Moon, 2001). Fur-
and insulin-like growth factors (IGFs) modulate effects in specific tis- thermore, it should be taken into account that the indirect effects of
sues such as the muscle. In fact, the GH/IGF axis is considered the GH in fish growth can increase appetite or the intestinal nutrient up-
most important endocrine system regulating skeletal growth, although take. An interesting aspect to consider is what part of the GH effect cor-
modulation of other hormones like insulin, thyroid hormones, steroids, responds to its direct action on muscle and what is mediated through
etc. allows the fine control of muscle growth and development as well IGFs secreted by the liver under GH stimulation.
as the adaptation to endogenous and external changes (Moomsen and In this sense, several studies in salmonids have demonstrated that a
Moon, 2001). GH chronic treatment causes an increase in plasma IGF-I levels (Biga et
The myogenic regulatory factors (MRFs) represent another impor- al., 2005; Raven et al., 2012; Kling et al., 2012). Moreover, such a treat-
tant group of molecules that exert a significant role specially for muscle ment provoked an increase in IGF-I gene expression in the liver in
development but also during the period of compensatory growth after coho salmon (Oncorhynchus kisutch) (Raven et al., 2012) as well as
fasting or reproductive stage, as well as for tissue regeneration after also in muscle and other tissues in rainbow trout (Oncorhynchus mykiss)
an injury. MRFs, myocyte enhancer factors (MEFs) and myostatin are (Biga et al., 2004b), thus supporting both systemic and local paracrine/
the best known molecules involved but, there are other factors less autocrine actions for GH and IGFs.
specific for muscle that also have important roles during myogenesis The studies on GH receptors and their signal transduction are rather
(e.g. FGF, HGF, PAX, Sox, etc.) (Fuentes et al., 2013). Moreover, in recent recent, and have demonstrated the existence of two main receptors
years, the target of rapamycin (TOR) complex has appeared as an im- (GHR-I and GHR-II) with complementary functions (Fuentes et al.,
portant level of integration between nutritional and endocrine inputs 2013). Truncated variants of GHR-I (tGHR-I) have been characterized
to improve growth (Vélez et al., 2014, 2016), which is of significant in- in turbot (Scophthalmus maximus) (Calduch-Giner et al., 2001), and in
terest for fish culture. other related species as Japanese flounder (Paralichthys olivaceus)
Due to the dynamic role of muscle from a metabolic point of view, (Nakao et al., 2004) or fine flounder (Paralichthys adspersus) (Fuentes
the endogenous proteolytic systems, which include the calpains, the ca- et al., 2012). In the last case, tGHR-I has been postulated as responsible
thepsins and the ubiquitin-proteasome system, are considered also key for the slow growth of this species. Two different GH receptors were
regulatory factors controlling growth potential. They are very important also described in several other fish species (Tse et al., 2003; Benedet et
systems in a tissue that often changes its metabolic role from an anabol- al., 2005; Saera-Vila et al., 2005; Jiao et al., 2006; Walock et al., 2014)
ic and synthetic side to a proteolytic mode to provide the organism with and in gilthead sea bream muscle (Sparus aurata), different responses
a supplementary load of amino acids and energy. to fasting and re-feeding were described (Saera-Vila et al., 2005). We
Muscle growth in fish has also a differential trait from other verte- have recently observed that both muscle GH receptors play different
brates seen in many species, that of continuous growth. This fact deter- roles in gilthead sea bream adapted to exercise (Vélez et al., 2016).
mines that after sexual maturation fish continue to grow still with These results agree with the fact that, in gilthead sea bream, GHR-I
important rates of muscle hyperplasia in comparison with most mam- seems to be more involved in anabolic signals, while GHR-II is directed
mals, which can change muscle mass only by hypertrophy (Stickland, towards energy depot mobilization, although more studies are required
1983). Furthermore, this also occurs in a reduced group of fish species in different species and conditions to generalize these results. Fish GH
(e.g. zebrafish, Danio rerio) that has been dubbed as a determined receptor signaling is even a younger discipline and Fuentes et al.
growth species (Biga and Goetz, 2006). Continuous growth in fish offers (2013) have reviewed the state of the art. Although the main pathways
an interesting model of research on the role of stem cells, and also gives are well conserved among vertebrates, it seems that both isoforms use
valuable possibilities for application to aquaculture. slightly different transduction molecules (Jiao et al., 2006; Kittilson et
In the last years, different publications have claimed the interest of al., 2011; Fuentes et al., 2012) with GHR-I working more through
exercise as a mechanism to improve fish aquaculture production, pro- STAT5 and GHR-II through ERK (Kittilson et al., 2011). Fuentes et al.
viding good results in growth and flesh quality. In mammals, it is well (2012) pointed out that in fine flounder, the JAK2/STAT5 pathway is
known that exercise stimulates a GH response that consequently pro- inactivated during fasting but reactivated with nutritionally favorable
vokes skeletal growth. Thus, it is interesting to consider the possibility conditions, which is in agreement with the GH receptor division of func-
of applying exercise during fish growth, which furthermore, due to tion observed in gilthead sea bream.
the high level of muscle plasticity, can respond with hyperplasia, There are not many studies on the direct effect of GH on muscle but
which has been demonstrated to be very important for flesh quality Rius-Francino et al. (2011) demonstrated the stimulatory effect of
(Blasco et al., 2015). gilthead sea bream GH on proliferation of cultured gilthead sea bream
Our group has a long history in the research of fish metabolism and myocytes (Fig. 1 insert). Interestingly stronger stimulatory effects on
endocrinology and in the last years has focused its attention in the reg- myocytes proliferation were obtained when GH was administrated to-
ulation and improvement of fish growth by means of in vitro and in vivo gether with IGF-I or IGF-II (Fig. 1). The GH transgenic coho salmon
approaches. The aim of this review is to summarize the current informa- (Devlin et al., 1994) has represented a valuable model to study the biol-
tion in fish muscle growth regulation, focusing primarily in GH/IGF axis ogy of growth, metabolism and behavior (Abernathy et al., 2015; de la
and its systemic and local actions on myocytes, and to propose new García et al., 2015; Chen et al., 2015; Kim et al., 2015a, 2015b) showing
molecules and approaches that can be interesting for basic research that GH overexpression produces both muscle hyperplasia and hyper-
and its application for fish aquaculture. trophy. Levesque et al. (2008) summarized the main facts that explain
30 E.J. Vélez et al. / Aquaculture 467 (2017) 28–40

Fig. 1. (A) Immunocytochemical detection of proliferating cell nuclear antigen (PCNA) in gilthead sea bream myocytes at day 4. Cell proliferation was determined after 6 h in the absence
(control, CT) or presence of mammalian GH 10 nM, IGF-I 100 nM, IGF-II 100 nM or combined treatments of GH and IGFs (IGF-I or IGF-II). Results are expressed as percentage of PCNA
positive cells (mean ± SEM). The insert figure shows the proliferative effects of sea bream GH (sbGH) on PCNA in the same conditions. Different letters indicate significant differences
(P b 0.05) and asterisks indicate differences with control (P b 0.001). (B) Representative microscope image indicating negative (arrow) and positive (arrow heads) cells. Bar =
200 μm. Adapted from Rius-Francino et al. (2011).

the higher growth in GH transgenic Atlantic salmon (Salmo salar), com- surprising the abundancy of them (Párrizas et al., 1994a, 1995), espe-
prising the higher numbers of myogenic precursor cells, their prolifera- cially when comparing with the number of insulin receptors. This was
tion rates and their direct proliferative response to GH treatment. GH true for white and red skeletal muscle, cultured myocytes and heart
transgenic fish have been obtained for other species like rainbow (Moon et al., 1996; Baños et al., 1997; Castillo et al., 2002, 2004), and
trout, carp (Cyprinus carpio), tilapia (Oreochromis niloticus), channel cat- confirmed in muscle of different poikilotherms. However, in
fish (Ictalurus punctatus) or zebrafish (Devlin et al., 2001; Rasmussen homeotherms, the situation reverted with higher levels of insulin recep-
and Morrissey, 2007; Figueiredo et al., 2007). tors that through evolution have gained importance in the regulation of
The different experiments of GH treatments (Raven et al., 2012; muscle function. Thus, the role of IGFs in fish muscle is facilitated by
Kling et al., 2012; Biga et al., 2004a, 2004b, 2005; Gahr et al., 2008) dem- their higher number of IGF-I receptors. Furthermore, to understand
onstrated the effects of this hormone on metabolism and muscle IGFs action, together with IGFs receptors, the function of IGF-I binding
growth. Biga et al. (2004a) found that GH treatment in rainbow trout in- proteins should be considered. IGFBPs represent a key step in the regu-
creases myosin protein levels, thus regulating the expression of the lation of IGFs action, but in fish there is limited information. Cunming
most abundant muscle protein. Gahr et al. (2008) reported that even Duan has done extensive work in fish, mainly zebrafish and with cell-
after short-term treatment, GH augmentation alters the expression of line models (Duan and Xu, 2005; Wang et al., 2009; Ocampo Daza et
genes involved in metabolism and growth regulation in rainbow trout. al., 2011). As in mammals, at least six different IGFBPs have been de-
Nevertheless, Biga and Mayer (2009), showed a differential regula- scribed that exert a double function of growth promotion and inhibition
tion of the growth-related genes as effect of GH treatment, in a compar- in fish. A long variety between species and tissues can be found in terms
ative experiment with an indetermined and a determined growth of IGFBPs expression (see below).
fish species (i.e. giant danio, Danio aequipinnatus and zebrafish, The effects of IGFs in in vivo approaches have been demonstrated in
respectively). In this work, the IGF-I and GHR-I expression was higher different studies. By administration of IGF-I through implanted mini-os-
in giant danio muscle that in zebrafish. However, the same treatment motic pumps, it was shown that IGF-I treatment increases the body size
increased the myostatin expression in zebrafish, whereas it was of juvenile coho salmon (McCormick et al., 1992). A significant correla-
down-regulated in the case of giant danio. These results suggest that tion between plasma IGF-I levels and growth rates in salmon was dem-
myostatin could be responsible for limiting the growth stimulation onstrated (Beckman et al., 1998; Dyer et al., 2004; Kawaguchi et al.,
caused by GH treatment in this determinate-growth species (zebrafish). 2013) and Beckman proposed that these IGF-I plasma levels can provide
Interestingly, GH transgenic fish were unable to respond to GH treat- a useful index of growth, although it is necessary to precisely define the
ment, indicating that transgenic fish show certain level of saturation on fish groups to compare (Beckman, 2011). Little information is available
stimulatory growth pathways (Raven et al., 2012). Recently, we have regarding circulating IGF-II blood levels in fish (Gentil et al., 1996;
treated gilthead sea bream fingerlings and juveniles with GH (Vélez et Wilkinson et al., 2004) but a positive correlation between IGF-I, IGF-II
al., unpublished data) and results demonstrated also in this species a and body weight was also observed in Atlantic salmon under nutritional
growth increase and diminution of fat depots. restriction (Wilkinson et al., 2006).
On the other hand, GH immunoneutralization in rainbow trout In vivo approaches to study IGFs face the difficulties to differenti-
(Fauconneau et al., 1996) decreased mainly muscle protein synthesis, ate between the effects of circulating levels of IGFs from hepatic ori-
together with a decrease in body weight. In zebrafish, Silva et al. gin and those from IGFs locally secreted by the muscle. In this sense,
(2015) achieved a double mutant for GH and GHR that resulted in in vitro studies enable to analyze the effects of a specific treatment
lower weight and a strong decrease of the somatotrophic axis intracel- without systemic influences. IGF muscle gene expression is regulat-
lular signaling by diminishing its signal transducer (STAT5.1). ed by GH, and Vong et al. (2003) found in carp that GH increased
McMenamin et al. (2013) identified a zebrafish GH1 mutant, vizzini, IGF-I and mainly IGF-II expression in muscle. Jiménez-Amilburu et
which exhibited abnormal small body size and increased accumulation al. (2013) found in gilthead sea bream myocytes, that GH increases
of adipose tissue. All these results support the direct action of GH on total IGF-I expression and that GH plus IGF-I also increases IGF-II ex-
muscle growth stimulating protein synthesis, hyperplasia and hypertro- pression. However, there was a clear dependency of the time and
phy, which in turn increase body weight. treatment conditions. Bower and Johnston (2010b) found in Atlantic
salmon myocytes that the addition of IGF-I and amino acids to
2.2. IGFs actions on growth starved cells results in an 18-fold increase in IGF-I mRNA, indicating
a positive feedback mechanism. Recently, Azizi et al. (2016) have
IGF-I and IGF-II have clear and important effects on fish muscle demonstrated in sea bream myocytes that both IGF-I and IGF-II stim-
growth. In the first studies on IGF-I receptors in fish muscle it was ulate the gene expression of IGF-I but not that of IGF-II. The complex
E.J. Vélez et al. / Aquaculture 467 (2017) 28–40 31

Fig. 2. Effects of insulin (INS) 1 μM, IGF-I 100 nM or IGF-II 100 nM on 2-deoxyglucose (A) and L-alanine (B) uptake in gilthead sea bream myocytes at day 4. The treatments were for 2 h in
the case of L-alanine and 1 h for the 2-deoxyglucose. Data are expressed in percentage over the basal (mean ± SEM). Different letters indicate significant differences (P b 0.05). Adapted
from Montserrat et al. (2012).

network of peptides, binding proteins and receptors provides an ef- 2.3. Metabolic effects of IGFs
ficient regulatory system of muscle function.
Fauconneau and Paboeuf (2000) and Gabillard et al. (2010) demon- The higher number of muscle IGF-I receptors compared to insulin re-
strated, by means of BrdU technique, the proliferative properties of IGF-I ceptors in fish suggested that IGFs could play some metabolic functions
and IGF-II in a dose dependent manner in rainbow trout muscle satellite in this tissue besides their proliferative and differentiation effects. The in
cells. Castillo et al. (2004) using the thymidine technique found the vitro culture of myocytes offered the possibility to investigate such ef-
same effects for IGF-I in rainbow trout myocytes and Codina et al. fects. Castillo et al. (2004) found that IGF-I stimulates glucose uptake
(2008) demonstrated that IGF-II stimulated also thymidine incorpora- in trout myocytes at a higher level than insulin. The same effect was ob-
tion in the same model. Rius-Francino et al. (2011) found in gilthead served for IGF-II in rainbow trout myocytes (Codina et al., 2008) and in
sea bream myocytes (by means of PCNA immunocytochemistry) that gilthead sea bream myocytes (Montserrat et al., 2007b, 2012) (Fig. 2a).
IGF-I and IGF-II stimulate proliferation, producing IGF-II stronger effects Later, the stimulatory effect of IGF-I in protein expression of glucose
than IGF-I (Fig. 1). transporter 4 (Glut4) was also found both in rainbow trout myocytes
Cleveland and Weber (2010) in rainbow trout primary cultured (Díaz et al., 2009) and gilthead sea bream myocytes (Montserrat et al.,
muscle cells observed that IGF-I stimulates protein synthesis and in- 2012).
hibits proteolysis via PI3K/protein kinase B signaling. Upton et al. Similarly, IGF-I and IGF-II stimulated the uptake of alanine in
(1998) also demonstrated the IGF-I inhibition of proteolysis in salmon myocytes of rainbow trout (Castillo et al., 2004; Codina et al., 2008)
cell lines. and gilthead sea bream (Montserrat et al., 2007a) at a stronger level
Overall, the abundance of IGF-I receptors in muscle tissues an- than insulin, indicating their strong anabolic role (Fig. 2b). Also, free
nounced an important role for these growth factors in fish muscle de- fatty acids were regulated by IGF-I: uptake in rainbow trout myocytes
velopment that has been demonstrated later using in vitro and in vivo was increased by this growth factor in a stronger way than insulin
studies. Thus, the IGFs stimulatory effects on proliferation and protein (Sánchez-Gurmaches et al., 2010). Thus, it is clear that together with
synthesis shown in different species has been reinforced with their pos- the role of IGFs stimulating proliferation and differentiation of muscle
itive correlation with fish size and weight growth. cells, IGFs also play a remarkable function for the regulation of

Fig. 3. Schematic representation of IGFs signaling pathways to control substrates uptake, protein synthesis and muscle cell proliferation. IGFs through the activation of the PI3K/AKT
pathway stimulate the entrance of amino acids (AA) and glucose (GLU) into the cell, which will provide substrates and energy for protein synthesis and cell proliferation via TOR and
MAPK signaling, respectively.
32 E.J. Vélez et al. / Aquaculture 467 (2017) 28–40

metabolism in fish, facilitating the uptake of substrates that will contrib- (MHC) fast isoforms and affected general swimming performance. GH
ute to the growth of muscle (Fig. 3). and IGF-I exert important effects on vertebrate thyroid function that
in general are stimulatory. Data in fish are scarce, but synergic effects
2.4. Other endocrine regulators of muscle growth of GH and TH on growth stimulation have been demonstrated in differ-
ent fish species, with several studies suggesting also that T4 can stimu-
Moomsen and Moon (2001) reviewed the effects of several hor- late GH release from pituitary somatotrophs (Donaldson et al., 1979).
mones in the regulation of fish growth. It is not the objective of this re- Furthermore, both hormones exert important metabolic effects; there-
view to cover in depth this topic but rather to focus in muscle fore, it is difficult to understand fish growth without GH and thyroid
regulation. Together with GH/IGF axis other endocrine molecules hormones contribution.
exert their influences on muscle growth and will be briefly commented
in the lines below. 2.4.3. Steroids and adrenergic agonists
Glucocorticoids and gonadal steroids exert significant effects in the
2.4.1. Insulin muscle of vertebrates. Cortisol is the main glucocorticoid in fish and it
Insulin is in mammals an important regulator of muscle function, but is involved importantly in the stress response. During stress, cortisol
it seems that in fish such a role is not so well developed, with IGFs serv- maintains the balance in energy homeostasis through stimulation of
ing part of its metabolic control. As mentioned above, the number of in- processes such as gluconeogenesis and inhibition of others such as di-
sulin receptors in the muscle of all fish species examined is clearly low gestion and immune response (Schjolden et al., 2009). It is also involved
compared to mammals (Párrizas et al., 1995). Different studies showed in muscle proteolysis in different conditions, like spawning migration,
that carnivorous fish species presented lower insulin receptors than smoltification, exercise, etc. (Ellis et al., 2012).
omnivorous species (Párrizas et al., 1994b), correlating the type of diet Corticosteroid receptors are expressed either in the cell membrane,
with the role of the muscle to allocate postprandial nutrients. While it for rapid actions or into the nucleus to regulate gene transcription. Tel-
was demonstrated that the number of insulin receptors was up-regulat- eost fish express two kinds of corticosteroid receptors, glucocorticoids
ed by diet or in response to insulin (Párrizas et al., 1994a) the receptor (GR) and mineralocorticoids (MR), which bind to cortisol with different
number never reached the level of IGF-I receptors and the action of in- affinities. GR show two isoforms, GR1 and GR2 that play different roles
sulin in fish is considered less important than in mammals. Neverthe- (Ellis et al., 2012).
less, there are different studies that demonstrate the anabolic function Cortisol is a key controller of fish intermediary metabolism, increas-
of insulin stimulating in vivo the uptake of amino acids in muscle ing oxygen uptake and gluconeogenesis and inhibiting glycogen synthe-
(Tashima and Cahill, 1968; Inui and Ishioka, 1983). Using in vitro cul- sis. Such an increase in metabolic rate would contribute to reduce
tured myocytes from rainbow trout and gilthead sea bream, it has growth and in support of this Ellis et al. (2012) described a negative cor-
been demonstrated that insulin increases the uptake of amino acids relation between growth and cortisol levels. Cortisol also regulates mus-
(Castillo et al., 2004; Montserrat et al., 2012). In the same model, insulin cle glycogen synthesis, and rainbow trout subjected to exhaustive
increased glucose entrance in myocytes by activating Glut4 (Díaz et al., exercise presented a decrease of white muscle glycogen stores in paral-
2009). Although at a lower level than IGF-I, insulin stimulated the up- lel to the rise of plasma cortisol levels (Milligan, 1997). Increased plas-
take of free fatty acids from the culture medium in trout myocytes ma cortisol levels also suppress appetite, reducing food intake, food
(Sánchez-Gurmaches et al., 2010). However, insulin did not provoke conversion efficiency, and growth in juvenile rainbow trout (de Boeck
thymidine uptake by rainbow trout myocytes suggesting a rather et al., 2001).
minor role in cell proliferation, and focused its emerging function in me- Cortisol possesses multiple functions in fish (osmoregulation, repro-
tabolism regulation. duction, behavior, etc.) but also interacts with GH/IGF to affect fish
growth (Leung et al., 2008; Kajimura et al., 2003; Pierce et al., 2011;
2.4.2. Thyroid hormones Won and Borski, 2013). Recently, Madison et al. (2015) suggested that
There is abundant information on the synergic effects of thyroid hor- the cortisol growth-suppressing effects in rainbow trout result from re-
mones (TH) with other factors (e.g. GH, steroids) on the stimulation of duced food intake mediated partially by increases in liver leptin-a1,
fish growth (Donaldson et al., 1979). Duarte-Guterman et al. (2014) brain corticotropin-releasing hormone, and a sustained increase in he-
reviewed such cross-talk between hormones, mainly for TH and sex ste- patic IGFBP-1 expression that reduces GH/IGF axis actions from mobiliz-
roids, regulating development and adult endocrine systems. In fact, a ing energy reserves. Nevertheless, it would be helpful to know more
prominent function of TH, common to all vertebrates, is their role in dif- about the indirect effect of cortisol on GH/IGF axis mediated thru the
ferentiation of specific tissues and in general in the development to the levels of blood metabolites.
adult phenotype. During the process of metamorphosis from larvae to The sex steroids have been related with changes in muscle protein
juvenile, fish undergoes numerous changes in the morphology and synthesis in fish linked to reproductive cycles (Moomsen and Moon,
physiology that are accompanied by surges in TH. This is especially rel- 2001). Anabolic steroids are growth promoters mainly in juveniles,
evant in the metamorphosis of the flatfish (Campinho et al., 2015; stimulating muscle protein synthesis, amino acid uptake in intestine, in-
Gomes et al., 2015). crease in nitrogen retention and ammonia excretion diminution
TH receptors (THR) are characterized in different fish species and in (Moomsen and Moon, 2001). Suppression of interrenal cells activity
flounder (Paralichthys olivaceus) THRα and THRβ show complementary by androgens has been observed in fish (Pottinger et al., 1996) thus
profiles in muscle metamorphosis (Moomsen and Moon, 2001) contrib- preventing cortisol proteolytic activity. Much of the information of sex
uting to changes towards the adult phenotype (Laudet, 2011). Treat- steroids in growth arrives from the interaction with GH/IGF axis
ment with TH accelerates metamorphosis (Power et al., 2001) while (McCormick et al., 2005). 17 β-estradiol (E2) is catabolic reducing
goitrogenic thiourea inhibits it. liver response to GH by decreasing IGF-I expression and production, as
Direct effects in fish muscle are described, with increases in protein well as activating proteolytic mechanisms in muscle (Cleveland and
and RNA synthesis in a dose dependent manner in tilapia (Tilapia Weber, 2011). Contrary, testosterone (T) is anabolic, increasing hepatic
mossambica) (Matty et al., 1982). In general, TH administration in- GH sensitivity and IGF-I production. Both steroids exert their effects in
creases the rate of muscle growth and the length and weight in fish muscle differentially regulating the sensitivity to IGF-I (Hanson et al.,
(Matty et al., 1982) although it is difficult to separate the direct effects 2012). Recently, Cleveland and Weber (2015) identified in salmonids
of TH and those via GH or IGFs and this will require further research. the mechanisms to respond to sex steroids and the disparate effects of
Little and Seebacher (2013) showed in zebrafish that hypothyroidism estrogens and androgens in fish growth. In fact, sex steroids affected dif-
treatment decreased mRNA concentrations of myosin heavy chain ferently IGF-I, IGF-II and IGFBPs expression in liver and muscle of
E.J. Vélez et al. / Aquaculture 467 (2017) 28–40 33

families that the fast-growing family expressed higher levels of IGF-II


than the slow-growing family, suggesting a specific role for IGF-II.
As indicated previously, IGFBPs and IGF-I receptors modulate the ac-
tion of growth factors. IGFBPs are a complex regulatory system includ-
ing six different proteins that furthermore can have paralogs with
specific functions (Johnston et al., 2011). IGFBP-1 was considered a neg-
ative regulator (Duan et al., 2000) and more recently other findings sup-
port this role in zebrafish muscle for both IGFBP-1a and IGFBP-1b being
these the only IGFBPs genes responding to a fasting–re-feeding (Amaral
and Johnston, 2011). A similar function was attributed to IGFBP-2 in
rainbow trout (Gabillard et al., 2006) and fine flounder (Safian et al.,
2012), but not in Atlantic salmon (Bower et al., 2008). Moreover,
Fig. 4. Relative gene expression profile throughout gilthead sea bream myocyte culture IGFBP-3 was proposed as a growth stimulator, but the results in fine
development of total IGF-I, IGF-II and the splice variants IGF-Ib and IGF-Ic. The splice
flounder are variable (Safian et al., 2012). IGFBP-4 and IGFBP-5 in the
variant IGF-Ia was not detectable. Mean ± SEM. Adapted from Jiménez-Amilburu et al.
(2013).
different species studied seem to play anabolic functions (Azizi et al.,
2016; Vélez et al., 2016) and can be considered pro-myogenic molecules
(Fuentes et al., 2013). The few studies which have analyzed the role of
IGFBP-6 (Gabillard et al., 2006; Bower et al., 2008; Safian et al., 2012)
rainbow trout. Thus, estradiol down-regulated GH/IGF axis in liver, point to it being a negative regulator of IGF-I. The regulation of IGFBPs
while in muscle maintained growth signals. Contrary, androgens up- is not very well known in fish muscle and Bower and Johnston
regulated GH/IGF axis in liver but not in muscle. (2010b) found that the addition of IGF-I, IGF-II or insulin and amino
β2-adrenergic agonists (BAAs) have similar actions to catechol- acids to the medium of Atlantic salmon cells increased the expression
amines, binding to the adrenergic receptors to produce specific actions of IGFBP-5.2 and IGFBP-4, but not IGFBP-5.1. IGF-I and IGF-II, directly
in target tissues. BAAs have been used in terrestrial farm animals to in- stimulated IGFBP-6 gene expression but not when amino acids were
crease muscle protein and to reduce muscle fat (Moomsen and Moon, present. Azizi et al. (2016) observed in gilthead sea bream myocytes
2001). In fish, BAAs administration has been investigated in several spe- that IGFBP-4 did not respond to IGFs treatment but IGFBP-5 was stimu-
cies, which revealed lower effects than in mammals (Mustin and Lovell, lated by both IGFs. Thus, once specific roles have been defined for a spe-
1993). Vélez et al. (unpublished results) found that formoterol activated cific species, IGFBPs can be good markers of the growth condition of the
gilthead sea bream myocytes TOR phosphorylation. Salem et al. (2006) fish.
treated rainbow trout with oral clenbuterol and ractopamine, analyzing Regarding IGF receptors, our group has investigated in different fish
their effects on muscle proteases and proteins. The study concluded that species their binding characteristics (Párrizas et al., 1995; Méndez et al.,
both BAAs show anabolic effects in muscle up-regulating protein syn- 2001) and signal transduction pathways (Castillo et al., 2006;
thesis and down-regulating proteolytic systems, although the authors Montserrat et al., 2007b). With respect to IGF-II receptors, very little
claimed more research before the application of these components in studies are available in fish but our group (Méndez et al., 2001) reported
aquaculture. Abo et al. (2012) demonstrated that the rat muscle hyper- a very low number of them in membranes of brown trout (Salmo trutta
trophy provoked by clenbuterol treatment was parallel to the up-regu- fario) embryos. In rainbow trout muscle, IGF-IRa expression did not re-
lation of IGF signaling. spond to fasting but increased in re-feeding and IGF-IRb expression per-
Thus, although the GH/IGF axis exerts the main role in the regulation formed the inverse role (Montserrat et al., 2007a). Recently, Azizi et al.
of growth, other hormones can modulate its action and need to be taken (2016) have observed in gilthead sea bream that IGFs down-regulated
into account to understand the final growth results. Natural reproduc- IGF-IRa gene expression at 6 h of treatment but the responses of IGF-
tive cycles or stress episodes will determine steroid circulating levels IRb were more variable as a function of the peptide and the duration
that directly or indirectly will influence GH/IGF axis consequently af- of the treatment. Overall, it seems that there is a certain level of func-
fecting growth performance. tional distribution and regulation between both IGF-I receptor isoforms
that deserves further investigation.
3. Regulation of fish myogenesis by IGFs MRFs play a very significant role in the regulation of muscle cells de-
velopment, and it is necessary to know their relationship with IGFs
It is well known that IGF-I plays an important role in skeletal muscle (Fig. 5). IGF-I and IGF-II in combination with amino acids stimulated
fiber regulation, including satellite cells activation, proliferation and dif- the gene expression of MyoD1c in Atlantic salmon myocytes (Bower
ferentiation (Snijders et al., 2015). In fish, Johnston et al. (2011) and and Johnston, 2010b). MyoD2 and Myf5 increased their expression
Fuentes et al. (2013) reviewed the effects of IGFs on skeletal muscle after GH and IGF-II treatment, while myogenin and MRF4 increased
growth regulation. The effects of IGFs on proliferation and differentia- in response to IGF-I stimulation in gilthead sea bream myocytes
tion in fish muscle cells have been also previously discussed (see (Jiménez-Amilburu et al., 2013). Vélez et al. (2014) also observed in
above; Fauconneau and Paboeuf, 2000; Castillo et al., 2004; Codina et gilthead sea bream myocytes that IGF-I was able to increase the pres-
al., 2008; Gabillard et al., 2010) as well as the differences between ence of myogenin positive cells but not MyoD. Recently, de Mello et al.
IGF-I and IGF-II (Rius-Francino et al., 2011; Azizi et al., 2016) (Fig. 1). (2015) also found in satellite cells of rainbow trout that IGF-I up-
Through the myogenic stages there are clear variations in muscle cell regulated myogenin expression. Azizi et al. (2016) observed that IGF-I
function. In our laboratory, we have studied the expression of IGFs dur- decreased MyoD gene expression in myocytes of this species but IGF-II
ing the development of gilthead sea bream myocytes. IGF-II expression did not. All these results suggest that at least in some fish species,
was highest at the beginning of culture and decreased when the cells both IGFs regulate myogenesis, but while IGF-II exerts its influence on
started to differentiate, similar to effects observed for total IGF-I the early stages, IGF-I seems to participate more in differentiation.
(Fig. 4). In the case of IGF-I, among the splice variants described previ- In this sense, Azizi et al. (2016) observed that IGF-I stimulated MHC
ously by Tiago et al. (2008), IGF-Ib showed a parallel expression pattern expression but not IGF-II.
as that of total IGF-I, whereas IGF-Ic showed its own profile during cul- Myostatin is a negative regulator of myogenesis and Seiliez et al.
ture progression (Jiménez-Amilburu et al., 2013). Peterson et al. (2004) (2012b) and Gabillard et al. (2013) found that it was able to decrease
found in channel catfish muscle that IGF-II gene expression was higher the proliferative effect of IGF-I in rainbow trout cultured satellite cells.
than that of IGF-I; and in the same study, it was observed in two catfish Garikipati and Rodgers (2012) also observed in rainbow trout myocytes
34 E.J. Vélez et al. / Aquaculture 467 (2017) 28–40

Fig. 5. Schematic model of fish myogenesis. The proliferation phase of myoblasts is identified by elevated gene expression of proliferating cell nuclear antigen (PCNA) accompanied by
several transcription factors (Sox8, Myf5, MyoD2 and Pax7) that control myocytes activation and development. Then, cells initiate the differentiation and fusion process to become
myofibers and mature myotubes showing a rise on Myogenin and MRF4 gene expression, and corroborated by an increase in myosin heavy chain (MHC) mRNA levels. Adapted from
Vélez et al. (2015).

that myostatin appeared to partly mediate IGF-I stimulated myogenic findings encouraged to continue the research for fish muscle growth
precursor cells differentiation. However, the effects of IGF-I on markers and indicators of differentiation and proliferation.
myostatin expression are not clear and could be opposite according to The TOR in mammals is well known as a key point to coordinate the
the isoform considered (MST1a or MST1b) or the experimental design. IGF-I signal with nutrient inputs (mainly amino acids) to regulate pro-
Fuentes et al. (2013) pointed out the importance of standardizing cell tein synthesis. However, in fish little information was available until
culture conditions to compare the respective findings and demonstrate Seiliez et al. (2008) published the first study in rainbow trout, showing
the specific regulatory role of this and other molecules. an increase of TOR phosphorylation at 2.5 h of postprandial stage. In the
Follistatin is a protein that is expressed almost by all tissues and it same study it was demonstrated that the phosphorylation of down-
has been demonstrated as a potent antagonist of members of the stream molecules of TOR (4EBP1 and 70S6K) was also increased when
transforming growth factor (TGF)-family, including myostatin. The in vitro myotubes were treated with amino acids or insulin. In gilthead
study of Medeiros et al. (2009) on follistatin transgenic rainbow trout sea bream myocytes, amino acids treatment but not IGF-I, was able to
demonstrated that myostatin inhibition and follistatin overexpression stimulate TOR phosphorylation and gene expression and also that of
caused a greater and visible muscle development, opening a new and its downstream molecules (Vélez et al., 2014). Later Azizi et al. (2016)
promising line of research for transfer to aquaculture. Snijders et al. verified that IGF-I does not provoke an increase on TOR gene expression
(2015) reviewed the role of the different IGF-I splice variants on but IGF-II was able to stimulate it, which fits well with its relevant role
human myogenesis regulation. Thus IGF-IEc or mechano growth factor during proliferation when protein synthesis is more abundant.
(MGF) stimulates proliferation and Myf5 expression, while IGF-IEa ex- Seiliez et al. (2013) demonstrated that cultured rainbow trout
pression strongly correlates with MRF4 expression, known for its role myotubes treated with human myostatin prevents the TORC1 signaling
in satellite cell differentiation. The MGF has not been described in fishes pathway by decreasing the phosphorylation of 4EBP1 and resulting in a
yet, but this kind of temporal association of IGF system regulating differ- reduction of cell diameter. Recently, Fuentes et al. (2015) found in fine
entially myogenesis remembers the distribution of functions among flounder that the blockage of TORC1 decreased muscle and body
both IGFs observed in gilthead sea bream. growth, muscle cellularity, IGF-I, IGF-II, IGFBP-4 and IGFBP-5 expres-
Summarizing, it is clear that IGFs have a significant role on the regu- sion, while the expression of muscle growth inhibitors (e.g., atrogin-1,
lation of myogenesis although there are differences between the species murf-1, IGFBP-2, IGFBP-3) was up-regulated. These authors claimed
as well as it also appears to be very important the developmental stage for the critical role of TORC1 in growth from the cellular to organism
or the physiological conditions chosen to perform the study. Although level. de la García et al. (2015) found that in GH transgenic coho salmon
more research in fish is needed, the IGF system is fundamental to under- muscle, TORC1 downstream molecules (eif4e1 and eif4e2) were also
standing the regulation of muscle growth as indicated previously up-regulated. There are other interesting molecules to consider in this
(Mingarro et al., 2002; Beckman, 2011), and can provide valuable review, like PAX7 and SOX8, however the information in fish is limited.
markers for the improvement of fish growth culture conditions. Bower and Johnston (2010a) showed PAX7 expression in Atlantic salm-
on muscle cells development, suggesting its role in proliferation and dif-
ferentiation. Froehlich et al. (2013a, 2013b) pointed to PAX7 and PAX3
4. New markers of muscle growth and quality as markers of myogenesis in zebrafish myocytes, since in association
with some transcription factors such as MyoD1, Myf5 and myogenin,
Early studies on IGFs and insulin effects demonstrated their stimula- these molecules can play a key role in regulating the determined and
tory actions on MAPK and AKT phosphorylation in rainbow trout and the indeterminate growth patterns of both zebrafish and giant danio
gilthead sea bream myocytes (Castillo et al., 2004; Montserrat et al., species, respectively. These authors suggest that PAX3 sustained ex-
2007b; Codina et al., 2008). Fuentes et al. (2011) showed how circulat- pression, paired with the reduced Myf5 showed in giant danio, can ex-
ing levels of IGF-I and nutritional status regulated both pathways in fine plain the hyperplasia capacity in adults of indeterminate growth
flounder muscle. However, the response of both pathways to the in vitro species. de la García et al. (2014) described its profile in sea bream
stimulation was different and MAPK phosphorylation was more respon- myoctes development, placing PAX7 expression peak in the first part
sive to treatment in myoblast than in myotubes. On the other hand, of the myogenic process close to Myf5 and MyoD.
AKT response to stimulation was more stable or even increased in dif- Muscle growth is a balance between proteogenic and proteolytic
ferentiated myocytes and myotubes (Montserrat et al., 2007b). These systems and this is specially observed in fish which naturally suffer
E.J. Vélez et al. / Aquaculture 467 (2017) 28–40 35

fasting periods. In this sense, the main proteolytic pathways in skeletal the growth and flesh quality in aquaculture, and its investigation will
muscle include the ubiquitin-proteasome system (UPS), the autopha- provide useful applied information.
gic-lysosome system (ALS) containing cathepsins, the calpain pathway,
and the caspase pathway. 5. Exercise and muscle growth in fish
In mammals UPS is the main responsible pathway for degradation
of short-lived, damaged, and misfolded proteins by two steps: There is growing evidence that moderate and sustained exercise can
ubiquitylation and proteasomal degradation. Muscle-specific RING-fin- improve fish growth and feed conversion in salmonids (Davison, 1997;
ger 1 (MURF1) and atrogin 1 (also known as MAFBX) are two muscle- Davison and Herbert, 2013) and in non-salmonid species (Palstra et al.,
specific ubiquitin ligases, enzymes participating in the last step of the 2010, 2014, 2015; Ibarz et al., 2011; Martín-Pérez et al., 2012; Felip et al.,
ubiquitylation, and are markedly induced in almost all types of muscle 2013). Most of these studies were performed in adult fish, but also in ju-
atrophy (Cohen et al., 2015). MURF1 and atrogin 1 are frequently used venile Atlantic salmon (McDonald et al., 1998), in fingerlings of yellow-
as genetic markers of muscle proteolysis in fish (Cleveland and Weber, tail (Seriola quinqueradiata) (Yogata and Oku, 2000) or in fingerlings of
2010; Seiliez et al., 2010; Bower et al., 2010; Salmerón et al., 2015). gilthead sea bream (Blasco et al., 2015; Vélez et al., 2016). In these last
ALS participates in the degradation of long-lived proteins and for the studies our group has demonstrated an increase in body weight after
elimination of redundant or damaged cellular structures. During fasting, 5 weeks of swimming.
the PI3K-AKT-TOR signaling decreases leading to activation of FOXO- In order to increase growth rate by means of moderate exercise, the
mediated expression of atrogenes which trigger muscle proteolysis via swimming speed of choice has been suggested to be the optimal (i.e.
the UPS and autophagic-lysosome system (Cohen et al., 2015). Some ev- lowest energy cost swimming, Uopt) (Davison and Herbert, 2013).
idences that these systems are regulated by IGF-I in fish exist. Bower et However, under these conditions, while salmonids increase their food
al. (2010) found that IGF-I down-regulated atrogin1 in muscle cells of intake to offset costs (Davison, 1997; Felip et al., 2012) other species
Atlantic salmon; similarly, IGF-I decreased the expression of atrogin1 as yellowtail (Seriola lalandy) (Palstra et al., 2015) or gilthead sea
and MURF1 in rainbow trout myocytes (Cleveland and Weber, 2010). bream enhance utilization of nutrients without food intake increase
IGF-I stimulation in rainbow trout myotubes, inhibited FoxO1 activity (Ibarz et al., 2011; Felip et al., 2013). Thus, fish seem to adapt their me-
(Seiliez et al., 2010, 2011). The same author (Seiliez et al., 2012a) re- tabolism to nutritional regime and environmental conditions and de-
ported in rainbow trout myocytes that amino acid deprivation is follow- pend on their ability to match fuel supply to energy use to grow
ed by a rapid increase of autophagosome formation and a slower (Magnoni et al., 2013). Sustained swimming can enhance the utilization
induction of the expression of several autophagy-related genes (LC3B, of dietary carbohydrates on a low-protein, high-carbohydrate regime,
gabarapl1, atg4b) via both TOR-dependent and -independent ways. To and spare the use of dietary proteins for muscle growth in both rainbow
investigate the relative contribution of ALS and UPS towards protein trout and gilthead sea bream (Martín-Pérez et al., 2012; Felip et al.,
degradation of myotubes in rainbow trout, Seiliez et al. (2014) found 2012, 2013). This sparing effect, in the case of juvenile sea bream, is
that IGF-I prevented the up-regulation of both systems under serum probably due to a lower entry of amino acids into the tricarboxylic
deprivation. Interestingly, ALS contribution to total protein degradation acid cycle as a reduction in citrate synthase (CS) activity, and an increase
was much higher than UPS. However, combination of both systems only of cytochrome-c-oxidase (COX) activity (Martín-Pérez et al., 2012).
represents 55% of total protein degradation, suggesting the action of However, in fingerlings of this species fed a high-protein diet under
other proteolytic systems. Two potential candidates of these other pro- sustained swimming, amino acid oxidation increased, as reflected by
teolytic systems are the calpains, proteases related with disruption of the high mitochondrial CS activity at the same time of the decrease in
myofibrillar proteins into smaller fragments and accessible for the COX activity. All of this demonstrates the great metabolic plasticity of
UPS, and the caspases, endopeptidases that regulate cell death and in- fish to adjust tissue-specific demands in response to exercise and ener-
flammation; however, they are not very well known in fish muscle gy availability. Interestingly, regardless of oxidative nutrient strategy,
(Macqueen et al., 2010; Preziosa et al., 2013; Salmerón et al., 2013, the response of fish to chronic submaximal swimming is, in general,
2015). qualitatively similar to mammalian aerobic training, and the trend is to-
Recently we have characterized in gilthead sea bream several mem- wards a more aerobic phenotype accelerating muscle growth (Johnston
bers of the calpains (Capn), lysosomal cathepsins (CTS) and UPS fami- and Moon, 1980; McClelland et al., 2006; LeMoine et al., 2010). This
lies, and shown that fasting induces an increase in capns1b, MURF, growth can occur by hypertrophy, hyperplasia or a combination of
atrogin 1 and several CTSs expression but re-feeding reduced CTSs, both depending on the species. We observed increases of capillarization
capn1, capn2, capns1a, capns1b and members of the UPS expression and fiber size of white muscle in gilthead sea bream under moderate
(Salmerón et al., 2013, 2015). Dietary positive effect on texture was swimming indicating vascularization and hypertrophy of this muscle
significantly correlated with decreased capn1 and capns1a expression (Ibarz et al., 2011) as reported previously in other species (Davison,
(Fig. 6). Overall, deeper knowledge of the role that the different 1997; Johnston, 1999; Johnston and Moon, 1980). Moreover, Palstra et
proteogenic and proteolytic systems play in fish will help to improve al. (2010, 2014) have also described that exercise effects in zebrafish

Fig. 6. Correlation between (A) calpain 1 (Capn1) or (B) calpain small subunit 1a (Capns1a) relative gene expression with muscle texture (maximal strength, N) of gilthead sea bream.
(A) r = −0.409, P = 0.043, (B) r = −0.449, P = 0.028. Adapted from Salmerón et al. (2013).
36 E.J. Vélez et al. / Aquaculture 467 (2017) 28–40

are directed by hypertrophy and angiogenesis. However, as Mommsen fingerlings, the isoform increased was IGF-Ic together with the elevated
(2001) suggested in an interesting review, zebrafish is not a good expression of the anabolic GH receptor, GHR-I (Vélez et al., 2016). More-
model of indeterminate growth; and as other authors have demonstrat- over, swimming stimulated the expression of IGFBP-5 in the anterior re-
ed later (Biga and Goetz, 2006), zebrafish have a little capacity of post- gion of the muscle, whereas in caudal muscle enhanced IGF-II mRNA
larval mosaic hyperplasia, being this one of the most important reason levels, suggesting different response to exercise throughout the muscle.
to postulate zebrafish as a species with a determinate growth pattern. Additionally, TOR activation and gene expression augmented in
Despite this data, it is not yet well established if sustained exercise en- the whole muscle mass, suggesting an increase in protein synthesis
hances also hyperplasia processes, but we have recently observed an in- that would sustain the higher growth rate in fish under moderate
crease of small fibers number of white muscle in fingerlings of gilthead swimming.
sea bream under moderate activity (Moya et al., unpublished data), and All these results confirm the positive growth effects of moderate and
it could be that the hyperplasia would be more suitable in species with sustained exercise and support the use of certain molecular markers
indeterminate growth. Global underlying changes in protein expression (such as GH/IGFs, IGFBP-5 or TOR) for monitoring the response of the
of white and red muscle of sea bream demonstrated that exercise affect- fish to growth trials.
ed many aspects of muscle physiology (Martín-Pérez et al., 2012). An-
other interesting observation was the reduction of mesenteric fat
found in gilthead sea bream fingerlings after 5 weeks of moderate and 6. Perspectives on fish growth and aquaculture
sustained exercise (Blasco et al., 2015).
The GH/IGF axis is tightly coupled to the energy status of the fish and The future of aquaculture requires an improvement of the growth
linked to feed intake via the expression of GH, IGF-I and their receptors and quality of the product combined with the sustainability of the activ-
(Raven et al., 2008). However, the literature about the axis response to ity. The endocrine control of growth includes several molecules and
moderate exercise in fish is scarce or partial. Although a close associa- pathways that we have summarized in Fig. 7. The GH/IGFs axis is the
tion between exercise induced growth and circulating levels of GH predominant system and a very valuable tool to monitor the perfor-
and IGF-I was not found for masu salmon (Oncorhynchus masou mance of a determined cultured fish. IGF-I levels appear to be good
masou) (Azuma et al., 2002), increased plasma GH for coho salmon, markers of adequate growth. However, very little information is avail-
and for rainbow trout under sustained swimming was observed able on IGF-II plasma levels and their function. Also better knowledge
(Barrett and McKeown, 1988a, 1988b). We have found a significant in- on circulating IGFBPs is necessary in many fish species to be utilized as
crease of circulating IGF-I concentration in gilthead sea bream juveniles growth markers.
under moderate swimming (Sánchez-Gurmaches et al., 2013). In fin- Regulation of fish myogenesis is still poorly understood and more
gerlings of this species, we have also observed an increased IGF-I/GH studies on the control of MRFs during muscle development and growth
ratio as a consequence of decreased GH and increased IGF-I plasma are required. Myostatin studies in fish are scarce but this could be a very
concentration (Blasco et al., 2015). Furthermore, in the same study, useful molecule for muscle growth improvement. The action of IGFs on
moderate swimming determined an increase of hepatic IGF-I gene muscle cells and the role of local IGFBPs and IGF-I receptors can provide
expression (mainly the IGF-Ia splice variant) in parallel to the suitable information to be applied in fish culture. All these molecules ac-
enhanced IGF-I levels in plasma. In muscle of gilthead sea bream quire special relevance in the continuous growth exhibited by aquacul-
ture fish species, and gives additional interest to try to explain this
characteristic of fish muscle development, not only for aquaculture
but also for vertebrate muscle regeneration.
The search for quality muscle markers is another important area that
seems to be still poorly developed in aquaculture in comparison with
farm animals. TOR complex has become an essential regulator of muscle
growth and needs more investigation in fish to serve as an indicator of
the good balance between nutrition and growth during fish culture.
The data on proteolytic systems has lately increased but more studies
on their role in vivo in fish growth are required. Some of these mole-
cules can be chosen for genetic investigation to be utilized in the pro-
grams of strain and broodstock selection.
In recent years, the discovery of noncoding RNAs, such as microRNAs
(miRNAs), has revealed a new perspective on the regulation of gene ex-
pression. miRNAs are small RNA molecules (19–22 nt) that regulate
gene expression by inhibiting protein translation and inducing mRNA
degradation (Silahtaroglu and Stenvang, 2010). Indeed, it has been ex-
tensively demonstrated that miRNAs are critically relevant in situations
that require adaptation to new conditions and processes that involve a
high degree of adaptability such as, the regulation of metabolism during
exercise (Párrizas et al., 2015). A set of miRNA, called myomiRs, have re-
cently been implicated in muscle growth and development (Güller and
Russell, 2010), indicating that it may be possible to use miRNAs as a new
and valuable tool for understanding such processes. Although this field
is extensively studied in mammals, there is still little information avail-
able about the role of muscle-related miRNAs in fish and their implica-
Fig. 7. Proposed model for muscle growth regulation in fish. The GH/IGFs system regulates tion on muscle growth.
the muscle expression of IGFs and IGFBPs, which can act in both autocrine and paracrine Finally, exercise is indicating a very useful way to increase fish
manner to control IGFs actions. In collaboration with myogenic regulatory factors growth and flesh quality that with an adequate diet can provide a sus-
(MRFs), cell proliferation and differentiation will be controlled. IGFs modulate cell
metabolism (e.g. nutrients uptake) and the TOR signaling pathway, which controls
tainable strategy to improve aquaculture that deserves further research.
protein turnover with also the involvement of proteolytic systems. All these regulatory In this review fundamental parts of skeletal growth regulation have
factors will participate in the control of hyperplasic and hypertrophic growth of muscle. not been covered, but it is also necessary to acquire a better knowledge
E.J. Vélez et al. / Aquaculture 467 (2017) 28–40 37

on the regulation of bone and cartilage development to be able to im- Aquaculture 246, 437–445.
Blasco, J., Moya, A., Milán-Cubillo, A., Vélez, E.J., Capilla, E., Pérez-Sánchez, J., Gutiérrez, J.,
prove final body weight, size and flesh quality. Fernández-Borràs, J., 2015. Growth-promoting effects of sustained swimming in fin-
In conclusion, in the last years there have been very significant ad- gerlings of gilthead sea bream (Sparus aurata L.). J. Comp. Physiol. B. 185, 859–868.
vances in the basic knowledge of the GH/IGF system and its role in the Bower, N.I., Johnston, I.A., 2010a. Paralogs of Atlantic salmon myoblast determinations
factors genes are distinctly regulated in proliferating and differentiating myogenic
regulation of metabolism, myogenesis and growth in fish. However, it cells. Am. J. Phys. Regul. Integr. Comp. Phys. 298, R1615–R1626.
remains still necessary to complete some of the aspects highlighted in Bower, N.I., Johnston, I.A., 2010b. Transcriptional regulation of the IGF signaling pathway
this review, and to transfer this information to fish culture, thus bring- by amino acids and insulin-like growth factors during myogenesis in Atlantic salmon.
PLoS One 5, e11100.
ing aquaculture to the level of quality and sustainability that the society Bower, N.I., Li, X., Taylor, R., Johnston, I.A., 2008. Switching to fast growth: the insulin-like
needs. growth factor (IGF) system in skeletal muscle of Atlantic salmon. J. Exp. Biol. 211,
3859–3870.
Bower, N.I., García de la serrana, D., Johnston, I.A., 2010. Characterization and differential
Acknowledgements
regulation of MAFbx/atrogin-1 α and β transcripts in skeletal muscle of Atlantic
salmon (Salmo salar). Biochem. Biophys. Res. Commun. 396, 265–271.
E.J.V. and E.L. are supported by a predoctoral fellowship from the Calduch-Giner, J.A., Duval, H., Chesnel, F., Boeuf, G., Pérez-Sánchez, J., Boujard, D., 2001.
Fish growth hormone receptor: molecular characterization of two membrane-an-
“Ministerio de Ciencia e Innovación” (MICINN). This study was support-
chored forms. Endocrinology 142, 3269–3273.
ed by the projects from the MICINN AGL2012-39768 and AGL2015- Campinho, M.A., Silva, N., Roman-Padilla, J., Ponce, M., Manchado, M., Power, D.M., 2015.
70679-R to J.G., AGL2014-57974-R to I.N., the SGR2009-00402 and Flatfish metamorphosis: a hypothalamic independent process? Mol. Cell. Endocrinol.
2014SGR-01371 from the “Generalitat de Catalunya” and the “Xarxa 404, 16–25.
Castillo, J., Le Bail, P.-Y., Paboeuf, G., Navarro, I., Weil, C., Fauconneau, B., Gutiérrez, J.,
de Refèrencia d'R + D + I en Aqüicultura”. The authors would like to 2002. IGF-I binding in primary culture of muscle cells of rainbow trout: changes
thank Carlos Mazorra from Tinamenor S.L., Ramon Fontanillas from during in vitro development. Am. J. Phys. Regul. Integr. Comp. Phys. 283,
Skretting and the personnel from the facilities at the School of Biology R647–R652.
Castillo, J., Codina, M., Martínez, M.L., Navarro, I., Gutiérrez, J., 2004. Metabolic and mito-
for the maintenance of the fish. genic effects of IGF-I and insulin on muscle cells of rainbow trout. Am. J. Phys. Regul.
Integr. Comp. Phys. 286, R935–R941.
References Castillo, J., Ammendrup-Johnsen, I., Codina, M., Navarro, I., Gutiérrez, J., 2006. IGF-I and in-
sulin receptor signal transduction in trout muscle cells. Am. J. Phys. Regul. Integr.
Abernathy, J., Panserat, S., Welker, T., Plagnes-Juan, E., Sakhrani, D., Higgs, D.A., Audouin, Comp. Phys. 290, R1683–R1690.
F., Devlin, R.H., Overturf, K., 2015. Food shortage causes differential effects on body Chen, Z., Devlin, R.H., Farrell, A.P., 2015. Upper thermal tolerance of wild-type, domesti-
composition and tissue-specific gene expression in salmon modified for increased cated and growth hormone-transgenic coho salmon Oncorhynchus kisutch. J. Fish
growth hormone production. Mar Biotechnol. (NY) 17 (6), 753–767. http://dx.doi. Biol. 87 (3), 763–773.
org/10.1007/s10126-015-9654-8. Cleveland, B.M., Weber, G.M., 2010. Effects of insulin-like growth factor-I, insulin, and leu-
Abo, T., Iida, R.-h., Kaneko, S., Suga, T., Yamada, H., Hamada, Y., Yamane, A., 2012. IGF and cine on protein turnover and ubiquitin ligase expression in rainbow trout primary
myostatin pathways are respectively induced during the earlier and the later stages myocytes. Am. J. Phys. Regul. Integr. Comp. Phys. 298 (2), R341–R350. http://dx.doi.
of skeletal muscle hypertrophy induced by clenbuterol, a b2-adrenergic agonist. org/10.1152/ajpregu.00516.2009.
Cell Biochem. Funct. 30 (8), 671–676. Cleveland, B.M., Weber, G.M., 2011. Effects of sex steroids on indices of protein turnover
Amaral, I.P.G., Johnston, I.A., 2011. Insulin-like growth factor (IGF) signalling and genome- in rainbow trout (Oncorhynchus mykiss) white muscle. Gen. Comp. Endocrinol. 174
wide transcriptional regulation in fast muscle of zebrafish following a single-satiating (2), 132–142. http://dx.doi.org/10.1016/j.ygcen.2011.08.011.
meal. J. Exp. Biol. 214, 2125–2139. Cleveland, B.M., Weber, G.M., 2015. Effects of sex steroids on expression of genes regulat-
Azizi, Sh, Nematollahi, M.A., Mojazi, A.B., Vélez, E.J., Salmerón, C., Chan, S.J., Navarro, I., ing growth-related mechanisms in rainbow trout (Oncorhynchus mykiss). Gen. Comp.
Capilla, E., Gutiérrez, J., 2016. IGF-I and IGF-II effects on local IGF system and signaling Endocrinol. 216, 103–115. http://dx.doi.org/10.1016/j.ygcen.2014.11.018.
pathways in gilthead sea bream (Sparus aurata) cultured myocytes. Gen. Comp. Codina, M., García de la serrana, D., Sánchez-Gurmaches, J., Montserrat, N., Chistyakova,
Endocrinol. 232, 7–16. O., Navarro, I., Gutiérrez, J., 2008. Metabolic and mitogenic effects of IGF-II in rainbow
Azuma, T., Noda, S., Yada, T., Ototake, M., Nagoya, H., Moriyama, S., Yamada, H., Nakanishi, trout (Oncorhynchus mykiss) myocytes in culture and the role of IGF-II in the PI3K/Akt
T., Iwata, M., 2002. Profiles in growth, smoltification, immune function and swim- and MAPK signalling pathways. Gen. Comp. Endocrinol. 157, 116–124.
ming performance of 1-year old masu salmon, Oncorhynchus masou masou reared Cohen, S., Nathan, J.A., Goldberg, A.L., 2015. Muscle wasting in disease: molecular mech-
in water flow. Fish. Sci. 68, 1282–1294. anisms and promising therapies. Nat. Rev. Drug Discov. 14 (1), 58–74. http://dx.doi.
Baños, N., Moon, T.W., Castejón, C., Gutiérrez, J., Navarro, I., 1997. Insulin and insulin-like org/10.1038/nrd4467.
growth factor-I (IGF-I) binding in fish red muscle: regulation by high insulin levels. Company, R., Astola, A., Pendón, C., Valdivia, M.M., Pérez-Sánchez, J., 2001. Somatotropic
Regul. Pept. 68, 181–187. regulation of fish growth and adiposity: growth hormone (GH) and somatolactin
Barrett, B.A., McKeown, B.A., 1988a. Sustained exercise increases plasma growth hormone (SL) relationship. Comp. Biochem. Physiol. C 130, 435–445.
concentrations in two anadromous salmonids. Can. J. Fish. Aquat. Sci. 45, 747–749. Davison, W., 1997. The effects of exercise training on teleost fish, a review of recent liter-
Barrett, B.A., McKeown, B.A., 1988b. Plasma growth hormone levels in Salmo gairderi: ature. Comp. Biochem. Physiol. A Physiol. 117, 67–75.
studies on temperature and exercise intensity/duration relationship. Comp. Biochem. Davison, W., Herbert, N., 2013. Swimming-enhanced growth. In: Palstra, A.J., Planas, J.V.
Physiol. A 94, 791–794. (Eds.), Swimming Physiology of FISH 2013. Springer, pp. 177–202.
Beckman, B.R., 2011. Perspectives on concordant and discordant relations between insu- de Boeck, G., Alsop, D., Wood, C., 2001. Cortisol effect on aerobic and anaerobic metabo-
lin-like growth factor 1 (IGF1) and growth in fishes. Gen. Comp. Endocrinol. 170, lism, nitrogen excretion and whole body composition in juvenile rainbow trout.
233–252. Physiol. Biochem. Zool. 74 (4), 858–868.
Beckman, B.R., Larsen, D.A., Lee-Pawlak, B., Moriyama, S., Dickhoff, W.W., 1998. Insulin- García de la serrana, D., Codina, M., Capilla, E., Jiménez-Amilburu, V., Navarro, I., Du,
like growth factor-I and environmental modulation of growth during smoltification S.H., Johnston, I.A., Gutiérrez, J., 2014. Characterisation and expression of regula-
of spring chinook salmon (Oncorhynchus tshawytscha). Gen. Comp. Endocrinol. 109, tory factors during in vitro myoblast development and in vivo fasting in the
325–335. gilthead sea bream (Sparus aurata). Comp. Biochem. Physiol. A Mol. Integr. Phys-
Benedet, S., Johansson, V., Sweeney, G., Galay-Burgos, M., Björnsson, B.T., 2005. Cloning of iol. 167, R90–R99.
two Atlantic salmon growth hormone receptor isoforms and in vitro ligand-binding García de la serrana, D., Devlin, R.H., Johnston, I.A., 2015. RNAseq analysis of fast skeletal
response. Fish Physiol. Biochem. 31, 315–329. muscle in restriction-fed transgenic coho salmon (Oncorhynchus kisutch): an experi-
Biga, P.R., Goetz, F.W., 2006. Zebrafish and giant danio as models for muscle growth: de- mental model uncoupling the growth hormone and nutritional signals regulating
terminate vs. indeterminate growth as determined by morphometric analysis. Am. growth. BMC Genomics 16, 564. http://dx.doi.org/10.1186/s12864-015-1782-z.
J. Phys. Regul. Integr. Comp. Phys. 291, R1327–R1337. de Mello, F., Streit Jr., D.P., Sabin, N., Gabillard, J.-C., 2015. Dynamic expression of tgf-β2,
Biga, P.R., Mayer, J., 2009. Growth hormone differentially regulates growth and growth- tgf-β3 and inhibin βA during muscle growth resumption and satellite cell differentia-
related gene expression in closely related fish species. Comp. Biochem. Physiol. A tion in rainbow trout (Oncorhynchus mykiss). Gen. Comp. Endocrinol. 210, 23–29.
154, 465–473. http://dx.doi.org/10.1016/j.ygcen.2014.10.011.
Biga, P.R., Cain, K.D., Hardy, R.W., Schelling, G.T., Overturf, K., Roberts, S.B., Goetz, F.W., Ott, Devlin, R.H., Yesaki, T.Y., Biagi, C.A., Donaldson, E.M., Swanson, P., Chan, W.-K., 1994. Ex-
T.L., 2004a. Growth hormone differentially regulates muscle myostatin1 and -2 and traordinary salmon growth. Nature 371, 209–210.
increases circulating cortisol in rainbow trout (Oncorhynchus mykiss). Gen. Comp. Devlin, R.H., Biagi, C.A., Yesaki, T.Y., Smailus, D.E., Byatt, J.C., 2001. Growth of domesticated
Endocrinol. 138, 32–41. transgenic fish. Nature 409, 781–782. http://dx.doi.org/10.1038/35057314.
Biga, P.R., Schelling, G.T., Hardy, R.W., Cain, K.D., Overturf, K., Ott, T.L., 2004b. The effects of Díaz, M., Vraskou, Y., Gutiérrez, J., Planas, J., 2009. Expression of rainbow trout glu-
recombinant bovine somatotropin (rbST) on tissue IGF-I, IGF-I receptor, and GH cose transporters GLUT1 and GLUT4 during in vitro muscle cell differentiation
mRNA levels in rainbow trout, Oncorhynchus mykiss. Gen. Comp. Endocrinol. 135, and regulation by insulin and IGF-I. Am. J. Phys. Regul. Integr. Comp. Phys. 296
324–333. (R794-R780).
Biga, P.R., Peterson, B.C., Schelling, G.T., Hardy, R.W., Cain, K.D., Overturf, K., Ott, T.L., 2005. Donaldson, E.M., Fagerlund, U.H.M., Higgs, D., McBride, J.R., 1979. Hormonal enhancement
Bovine growth hormone treatment increased IGF-I in circulation and induced the of growth. In: Hoar, W.S., Randall, D.J., Brett, J.R. (Eds.), Fish Physiology: Bioenergetics
production of a specific immune response in rainbow trout (Oncorhynchus mykiss). and Growth 3. Academic Press, New York, pp. 456–578.
38 E.J. Vélez et al. / Aquaculture 467 (2017) 28–40

Duan, C., Xu, Q., 2005. Roles of insulin-like growth factor (IGF) binding proteins in regu- Ibarz, A., Felip, O., Fernández-Borràs, J., Martín-Pérez, M., Blasco, J., Torrella, J.R., 2011.
lating IGF actions. Gen. Comp. Endocrinol. 142, 44–52. http://dx.doi.org/10.1016/j. Sustained swimming improves muscle growth and cellularity in gilthead sea
ygcen.2004.12.022. bream. J. Comp. Physiol. B. 181, 209–217.
Duan, C., Ren, H., Gao, S., 2000. Insulin-like growth factors (IGFs), IGF receptors, and IGF- Inui, Y., Ishioka, H., 1983. Effects of insulin and glucagon on amino acid transport into the
binding proteins: roles in skeletal muscle growth and differentiation. Gen. Comp. liver and opercular muscle of the eel in vitro. Gen. Comp. Endocrinol. 51, 213–218.
Endocrinol. 167, 344–351. http://dx.doi.org/10.1016/j.ygcen.2010.04.009. Jiao, B., Huang, X., Chan, C.B., Zhang, L., Wang, D., Cheng, C.H.K., 2006. The co-existence of
Duarte-Guterman, P., Navarro-Martín, L., Trudeau, V.L., 2014. Mechanisms of crosstalk be- two growth hormone receptors in teleost fish and their differential signal transduc-
tween endocrine systems: regulation of sex steroid hormone synthesis and action by tion, tissue distribution and hormonal regulation of expression in seabream. J. Mol.
thyroid hormones. Gen. Comp. Endocrinol. 203, 69–85. http://dx.doi.org/10.1016/j. Endocrinol. 36, 23–40.
ygcen.2014.03.015. Jiménez-Amilburu, V., Salmerón, C., Codina, M., Navarro, I., Capilla, E., Gutiérrez, J., 2013.
Dyer, A.R., Barlow, C.G., Bransden, M.P., Carter, C.G., Glencross, B.D., Richardson, N., Insulin-like growth factors effects on the expression of myogenic regulatory factors
Thomas, P.M., Williams, K.C., Carragher, J.F., 2004. Correlation of plasma IGF-I concen- in gilthead sea bream muscle cells. Gen. Comp. Endocrinol. 188, 151–158.
trations and growth rate in aquacultured finfish: a tool for assessing the potential of Johnston, I.A., 1999. Muscle development and growth: potential implications for flesh
new diets. Aquaculture 236, 583–592. http://dx.doi.org/10.1016/j.aquaculture.2003. quality in fish. Aquaculture 177, 99–115.
12.025. Johnston, I.A., Moon, T.W., 1980. Exercise training in skeletal muscle of brook trout
Ellis, T., Yildiz, H.Y., López-Olmeda, J., Spedicato, M.T., Tort, L., Øverli, Ø., Martins, C.I.M., (Salvelinus fontinalis). J. Exp. Biol. 87, 177–194.
2012. Cortisol and finfish welfare. Fish Physiol. Biochem. 38, 163–188. Johnston, I.A., Bower, N.I., Macqueen, D.J., 2011. Growth and the regulation of myotomal
Fauconneau, B., Paboeuf, G., 2000. Effect of fasting and refeeding on in vitro muscle cell muscle mass in teleost fish. J. Exp. Biol. 214 (Pt 10), 1617–1628.
proliferation in rainbow trout (Oncorhynchus mykiss). Cell Tissue Res. 301 (3), Kajimura, S., Hirano, T., Visitacion, N., Moriyama, S., Aida, K., Grau, E.G., 2003. Dual mode
459–463. of cortisol action on GH/IGF-I/IGF binding proteins in the tilapia, Oreochromis
Fauconneau, B., Mady, M.P., Le bail, P.-Y., 1996. Effect of growth hormone on muscle pro- mossambicus. J. Endocrinol. 178, 91–99.
tein synthesis in rainbow trout (Oncorhynchus mykiss) and Atlantic salmon (Salmo Kawaguchi, K., Kaneko, N., Fukuda, M., Nakano, Y., Kimura, S., Hara, A., Shimizu, M.,
salar). Fish Physiol. Biochem. 15 (1), 49–56. http://dx.doi.org/10.1007/BF01874837. 2013. Responses of insulin-like growth factor (IGF)-I and two IGF-binding pro-
Felip, O., Ibarz, A., Fernández-Borràs, J., Beltrán, M., Martín-Pérez, M., Planas, J.V., Blasco, J., tein-1 subtypes to fasting and re-feeding, and their relationships with individual
2012. Tracing metabolic routes of dietary carbohydrate and protein in rainbow trout growth rates in yearling masu salmon (Oncorhynchus masou). Comp. Biochem.
(Oncorhynchus mykiss) using stable isotopes ([13C] starch and [15N] protein): Effects Physiol. A Mol. Integr. Physiol. 165 (2), 191–198. http://dx.doi.org/10.1016/j.
of gelatinisation of starches and sustained swimming. Br. J. Nutr. 107, 834–844. cbpa.2013.02.029.
Felip, O., Blasco, J., Ibarz, A., Martín-Pérez, M., Fernández-Borràs, J., 2013. Beneficial effects Kim, J.-H., Leggatt, R.A., Chan, M., Volkoff, H., Devlin, R.H., 2015a. Effects of chronic
of sustained activity on the use of dietary protein and carbohydrate traced with stable growth hormone overexpression on appetite regulating brain gene expression
isotopes 15N and 13C in gilthead sea bream (Sparus aurata). J. Comp. Physiol. B. 183, in coho salmon. Mol. Cell. Endocrinol. 413, 178–188. http://dx.doi.org/10.1016/
223–234. j.mce.2015.06.024.
Figueiredo, M.A., Lanes, C.F.C., Almeida, D.V., Marins, L.F.F., 2007. Improving the produc- Kim, J.-H., White, S.L., Devlin, R.H., 2015b. Interaction of growth hormone overexpression
tion of transgenic fish germlines: in vivo evaluation of mosaicism in zebrafish and nutritional status on pituitary gland clock gene expression in coho salmon, Onco-
(Danio rerio) using a green fluorescent protein (GFP) and growth hormone cDNA rhynchus kisutch. Chronobiol. Int. 32 (1), 113–127. http://dx.doi.org/10.3109/
transgene co-injection strategy. Genet. Mol. Biol. 30, 31–36. 07420528.2014.958160.
Froehlich, J.M., Galt, N.J., Charging, M.J., Meyer, B.M., Biga, P.R., 2013a. In vitro indetermi- Kittilson, J.D., Jones, E., Sheridan, M.A., 2011. ERK, Akt, and STAT5 are differentially activat-
nate teleost myogenesis appears to be dependent on Pax3. In Vitro Cell Dev. Biol. ed by the two growth hormone receptor subtypes of a teleost fish (Oncorhynchus
Anim. 49 (5), 371–385. mykiss). Front. Endocrinol. 2, 30.
Froehlich, J.M., Fowler, Z.G., Galt, N.J., Smith Jr., D.L., Biga, P.R., 2013b. Sarcopenia and pi- Kling, P., Jönsson, E., Nilsen, T.O., Einarsdottir, I.E., Rønnestad, I., Stefansson, S.O.,
scines: the case for indeterminate-growing fish as unique genetic model organisms Björnsson, B.T., 2012. The role of growth hormone in growth, lipid homeostasis, ener-
in aging and longevity research. Front. Genet. 4, 159. gy utilization and partitioning in rainbow trout: Interactions with leptin, ghrelin and
Fuentes, E.N., Björnsson, B.T., Valdés, J.A., Einarsdottir, I.E., Lorca, B., Alvarez, M., Molina, A., insulin-like growth factor I. Gen. Comp. Endocrinol. 175, 153–162. http://dx.doi.org/
2011. IGF-I/PI3K/Akt and IGF-I/MAPK/ERK pathways in vivo in skeletal muscle are 10.1016/j.ygcen.2011.10.014.
regulated by nutrition and contribute to somatic growth in the fine flounder. Am. Laudet, V., 2011. The origins and evolution of vertebrate metamorphosis. Curr. Biol. 21,
J. Phys. Regul. Integr. Comp. Phys. 300 (6), R1532–R1542. http://dx.doi.org/10.1152/ R726–R737. http://dx.doi.org/10.1016/j.cub.2011.07.030.
ajpregu.00535.2010. LeMoine, C.M.R., Craig, P.M., Dhekney, K., Kim, J.J., McClelland, G.B., 2010. Temporal and
Fuentes, E.N., Einarsdottir, I.E., Valdes, J.A., Alvarez, M., Molina, A., Björnsson, B.T., 2012. In- spatial patterns of gene expression in skeletal muscles in response to swim training
herent growth hormone resistance in the skeletal muscle of the fine flounder is mod- in adult zebrafish (Danio rerio). J. Comp. Physiol. B. 180, 151–160.
ulated by nutritional status and is characterized by high contents of truncated GHR, Leung, L.Y., Kwong, A.K.Y., Man, A.K.Y., Woo, N.Y.S., 2008. Direct actions of cortisol, thy-
impairment in the JAK2/STAT5 signaling pathway, and low IGF-I expression. Endocri- roxine and growth hormone on IGF-I mRNA expression in sea bream hepatocytes.
nology 153, 283–294. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 151, 705–710.
Fuentes, E.N., Valdés, J.A., Molina, A., Björnsson, B.T., 2013. Regulation of skeletal muscle Levesque, H.M., Shears, M.A., Fletcher, G.L., Moon, T.W., 2008. Myogenesis and muscle
growth in fish by the growth hormone – insulin-like growth factor system. Gen. metabolism in juvenile Atlantic salmon (Salmo salar) made transgenic for growth
Comp. Endocrinol. 192, 136–148. hormone. J. Exp. Biol. 211, 128–137.
Fuentes, E.N., Einarsdottir, I.E., Paredes, R., Hidalgo, C., Valdes, J.A., Björnsson, B.T., Molina, Little, A.G., Seebacher, F., 2013. Thyroid hormone regulates muscle function during cold
A., 2015. The TORC1/P70S6K and TORC1/4EBP1 signaling pathways have a stronger acclimation in zebrafish (Danio rerio). J. Exp. Biol. 216, 3514–3521. http://dx.doi.
contribution on skeletal muscle growth than MAPK/ERK in an early vertebrate: Dif- org/10.1242/jeb.089136.
ferential involvement of the IGF system and atrogenes. Gen. Comp. Endocrinol. 210, Macqueen, D.J., Delbridge, M.L., Manthri, S., Johnston, I.A., 2010. A newly classified
96–106. vertebrate calpain protease, directly ancestral to CAPN1 and 2, episodically evolved
Gabillard, J.-C., Kamangar, B.B., Montserrat, N., 2006. Coordinated regulation of the GH/IGF a restricted physiological function in placental mammals. Mol. Biol. Evol. 27,
system genes during refeeding in rainbow trout (Oncorhynchus mykiss). J. Endocrinol. 1886–1902.
191, 15–24. Madison, B.N., Tavakoli, S., Kramer, S., Bernier, N.J., 2015. Chronic cortisol and the regula-
Gabillard, J.-C., Sabin, N., Paboeuf, G., 2010. In vitro characterization of proliferation and tion of food intake and the endocrine growth axis in rainbow trout. J. Endocrinol. 226
differentiation of trout satellite cells. Cell Tissue Res. 342, 471–477. (2), 103–119. http://dx.doi.org/10.1530/JOE-15-0186.
Gabillard, J.-C., Biga, P.R., Rescan, P.-Y., Seiliez, I., 2013. Revisiting the paradigm of Magnoni, L.J., Crespo, D., Ibarz, A., Blasco, J., Fernández-Borràs, J., Planas, J.V., 2013. Effects
myostatin in vertebrates: insights from fishes. Gen. Comp. Endocrinol. 194, 45–54. of sustained swimming on the red and white muscle transcriptome of rainbow trout
http://dx.doi.org/10.1016/j.ygcen.2013.08.012. (Oncorhynchus mykiss) fed a carbohydrate-rich diet. Comp. Biochem. Physiol. A Mol.
Gahr, S.A., Vallejo, R.L., Weber, G.M., Shepherd, B.S., Silverstein, J.T., Rexroad, C.E., 2008. Ef- Integr. Physiol. 166, 510–521.
fects of short-term growth hormone treatment on liver and muscle transcriptomes in Martín-Pérez, M., Fernández-Borràs, J., Ibarz, A., Millan-Cubillo, A., Felip, O., de Oliveira, E.,
rainbow trout (Oncorhynchus mykiss). Physiol. Genomics 32, 380–392. http://dx.doi. Blasco, J., 2012. New insights into fish swimming: a proteomic and isotopic approach
org/10.1152/physiolgenomics.00142.2007. in gilthead sea bream. J. Proteome Res. 11, 3533–3547.
Garikipati, D.K., Rodgers, B.D., 2012. Myostatin inhibits myosatellite cell proliferation and Matty, T.W., Chaudhury, M.A., Lone, K.P., 1982. Effect of thyroid hormones on the protein
consequently activates differentiation: evidence for endocrine-regulated transcript metabolism of Tilapia mossambica. Gen. Comp. Endocrinol. 46, 408–413.
processing. J. Endocrinol. 215, 177–187. McClelland, G.B., Craig, P.M., Dhekney, K., Dipardo, S., 2006. Temperature- and exercise-
Gentil, V., Martin, P., Smal, J., Le Bail, P.-Y., 1996. Production of recombinant insulin-like induced gene expression and metabolic enzyme changes in skeletal muscle of adult
growth factor-II in the development of a radioimmunoassay in rainbow trout (Onco- zebrafish (Danio rerio). J. Physiol. 577, 739–751.
rhynchus mykiss). Gen. Comp. Endocrinol. 104 (2), 156–167. McCormick, S.D., Kelley, K.M., Young, G., Nishioka, R.S., Bern, H.A., 1992. Stimulation of
Gomes, A.S., Alves, R.N., Rønnestad, I., Power, D.M., 2015. Orchestrating change: the thy- coho salmon growth by insulin-like growth factor I. Gen. Comp. Endocrinol. 86,
roid hormones and GI-tract development in flatfish metamorphosis. Gen. Comp. 398–406.
Endocrinol. 220, 2–12. McCormick, S.D., O'Dea, M.F., Moeckel, A.M., Lerner, D.T., Bjornsson, B.J., 2005. Endocrine
Güller, I., Russell, A.P., 2010. MicroRNAs in skeletal muscle: their role and regulation in de- disruption of parr-smolt transformation and seawater tolerance of Atlantic salmon
velopment, disease and function. J. Physiol. 588 (Pt 21), 4075–4087. by 4-nonylphenol and 17b-estradiol. Gen. Comp. Endocrinol. 142, 280–288.
Hanson, A.M., Kittilson, J.D., McCormick, S.D., Sheridan, M.A., 2012. Effects of 17bestradiol, McDonald, D.G., Milligan, C.L., McFarlane, W.J., Croke, S., Currie, S., Hooke, B., Angus, R.B.,
4-nonylphenol, and b-sitosterol on the growth hormone-insulin-like growth factor Tufts, B.L., Davidson, K., 1998. Condition and performance of juvenile Atlantic salmon
system and seawater adaptation of rainbow trout (Oncorhynchus mykiss). Aquacul- (Salmo salar): effects of rearing practices on hatchery fish and comparison with wild
ture 362–363, 241–247. fish. Can. J. Fish. Aquat. Sci. 55, 1208–1219.
E.J. Vélez et al. / Aquaculture 467 (2017) 28–40 39

McMenamin, S.K., Minchin, J.E.N., Gordon, T.N., Rawls, J.F., Parichy, D.M., 2013. Dwarfism Rasmussen, R.S., Morrissey, M.T., 2007. Marine biotechnology for production of food in-
and increased adiposity in the gh1 mutant zebrafish vizzini. Endocrinology 154 (4), gredients. Adv. Food Nutr. Res. 52, 237–292.
1476–1487. http://dx.doi.org/10.1210/en.2012-1734. Raven, P.A., Uh, M., Sakhrani, D., Beckman, B.R., Cooper, K., Pinter, J., Leder, E.H.,
Medeiros, E.F., Phelps, M.P., Fuentes, F.D., Bradley, T.M., 2009. Overexpression of follistatin Silverstein, J., Devlin, R.H., 2008. Endocrine effects of growth hormone overexpression
in trout stimulates increased muscling. Am. J. Phys. Regul. Integr. Comp. Phys. 297, in transgenic coho salmon. Gen. Comp. Endocrinol. 159, 26–37.
R235–R242. Raven, P.A., Sakhrani, D., Beckman, B., Neregård, L., Sundström, L.F., Björnsson, B.T., Devlin,
Méndez, E., Planas, J.V., Castillo, J., Navarro, I., Gutiérrez, J., 2001. Identification of a type II R.H., 2012. Growth and endocrine effects of recombinant bovine growth hormone
insulin-like growth factor receptor in fish embryos. Endocrinology 142 (3), treatment in non-transgenic and growth hormone transgenic coho salmon. Gen.
1090–1097. Comp. Endocrinol. 177, 143–152. http://dx.doi.org/10.1016/j.ygcen.2012.03.002.
Milligan, C.L., 1997. The role of cortisol in ammonia mobilization and metabolism follow- Reindl, K.M., Sheridan, M.A., 2012. Peripheral regulation of the growth hormone-insulin-
ing exhaustive exercise in rainbow trout (Oncorhynchus mykiss Walbaum). Fish Phys- like growth factor system in fish and other vertebrates. Comp. Biochem. Physiol. A.
iol. Biochem. 16, 119–128. 163, 231–245. http://dx.doi.org/10.1016/j.cbpa.2012.08.003.
Mingarro, M., Vega-Rubín de Celis, S., Astola, A., Pendón, C., Martínez Valdivia, M., Pérez- Reinecke, M., 2010. Influences of the environment on the endocrine and paracrine fish
Sánchez, J., 2002. Endocrine mediators of seasonal growth in gilthead sea bream growth hormone-insulin-like growth factor-I system. J. Fish Biol. 76 (6),
(Sparus aurata): the growth hormone and somatolactin paradigm. Gen. Comp. 1233–1254. http://dx.doi.org/10.1111/j.1095-8649.2010.02605.x.
Endocrinol. 128, 102–111. Reinecke, M., Björnsson, B.T., Dickhoff, W.W., McCormick, S.D., Navarro, I., Power, D.M.,
Mommsen, T.P., 2001. Paradigms of growth in fish. Comp. Biochem. Physiol. B 129, Gutiérrez, J., 2005. Growth hormone and insulin-like growth factors in fish: where
207–219. we are and where to go. Gen. Comp. Endocrinol. 142, 20–24. http://dx.doi.org/10.
Montserrat, N., Gabillard, J.-C., Capilla, E., Navarro, I., Gutiérrez, J., 2007a. Role of insulin, 1016/j.ygcen.2005.01.016.
insulin-like growth factors, and muscle regulatory factors in the compensatory Rius-Francino, M., Acerete, L., Jiménez-Amilburu, V., Capilla, E., Navarro, I., Gutiérrez, J.,
growth of the trout (Oncorhynchus mykiss). Gen. Comp. Endocrinol. 150, 462–472. 2011. Differential effects on proliferation of GH and IGFs in sea bream (Sparus aurata)
http://dx.doi.org/10.1016/j.ygcen.2006.11.009. cultured myocytes. Gen. Comp. Endocrinol. 172, 44–49.
Montserrat, N., Sánchez-Gurmaches, J., García de la serrana, D., Navarro, I., Gutiérrez, J., Saera-Vila, A., Calduch-Giner, J.A., Pérez-Sánchez, J., 2005. Duplication of growth hormone
2007b. IGF-I binding and receptor signal transduction in primary cell culture of mus- receptor (GHR) in fish genome: gene organization and transcriptional regulation of
cle cells of gilthead sea bream: changes throughout in vitro development. Cell Tissue GHR type I and II in gilthead sea bream (Sparus aurata). Gen. Comp. Endocrinol.
Res. 330, 503–513. 142, 193–203.
Montserrat, N., Capilla, E., Navarro, I., Gutiérrez, J., 2012. Metabolic effects of insulin Safian, D., Fuentes, E.N., Valdés, J.A., Molina, A., 2012. Dynamic transcriptional regulation
and IGFs on gilthead sea bream (Sparus aurata) muscle cells. Front. Endocrinol. of autocrine/paracrine igfbp1, 2, 3, 4, 5, and 6 in the skeletal muscle of the fine floun-
39, 55. der during different nutritional statuses. J. Endocrinol. 214, 95–108.
Moomsen, T.P., Moon, T.W., 2001. Hormonal regulation of muscle growth. In: Johnston, Salem, M., Levesque, H., Moon, T.W., Rexroad, C.E., Yao, J., 2006. Anabolic effects of feeding
I.A. (Ed.), Fish Physiology: Muscle development and growth 18. Academic Press, beta2-adrenergic agonists on rainbow trout muscle proteases and proteins. Comp.
San Diego, pp. 251–308. Biochem. Physiol. A Mol. Integr. Physiol. 144 (2), 145–154.
Moon, T.W., Castejón, C., Baños, N., Maestro, M.A., Plisetskaya, E.M., Gutiérrez, J., Navarro, Salmerón, C., García de la serrana, D., Jiménez-Amilburu, V., Fontanillas, R., Navarro, I.,
I., 1996. Insulin and IGF-I binding in isolated trout cardiomyocytes. Gen. Comp. Johnston, I.A., Gutiérrez, J., Capilla, E., 2013. Characterisation and expression of calpain
Endocrinol. 103, 264–272. family members in relation to nutritional status, diet composition and flesh texture in
Mustin, W.T., Lovell, R.T., 1993. Feeding the repartitioning agent, ractopamine, to channel gilthead sea bream (Sparus aurata). PLoS One 8 (9), e75349.
catfish (Ictalurus punctatus) increases weight gain and reduces fat deposition. Aqua- Salmerón, C., Navarro, I., Johnston, I.A., Gutiérrez, J., Capilla, E., 2015. Characterisation and
culture 109, 145–152. expression analysis of cathepsins and ubiquitin-proteasome genes in gilthead sea
Nakao, N., Higashimoto, Y., Ohkubo, T., Yoshizato, H., Nakai, N., Nakashima, K., Tanaka, M., bream (Sparus aurata) skeletal muscle. BMC Res. Notes 8, 149.
2004. Characterization of structure and expression of the growth hormone Sánchez-Gurmaches, J., Cruz-García, L., Gutiérrez, J., Navarro, I., 2010. Endocrine control of
receptor gene of the Japanese flounder (Paralichtys olivaceus). J. Endocrinol. 182 oleic acid and glucose metabolism in rainbow trout (Oncorhynchus mykiss) muscle
(1), 157–164. cells in culture. Am. J. Phys. Regul. Integr. Comp. Phys. 299, R562–R572.
Ocampo Daza, D., Sundström, G., Bergqvist, C.A., Duan, C., Larhammar, D., 2011. Evolution Sánchez-Gurmaches, J., Cruz-García, L., Ibarz, A., Fernández-Borràs, J., Blasco, J., Gutiérrez,
of the insulin-like growth gactor binding protein (IGFBP) family. Endocrinology 152 J., Navarro, I., 2013. Insulin, IGF-I, and muscle MAPK pathway responses after
(6), 2278–2289. http://dx.doi.org/10.1210/en.2011-0047. sustained exercise and their contribution to growth and lipid metabolism regulation
Palstra, A.P., Tudorache, C., Rovira, M., Brittijn, S.A., Burgerhout, E., van den Thillart, in gilthead sea bream. Domest. Anim. Endocrinol. 45, 145–153.
G.E.E.J.M., Spaink, H.P., Planas, J.V., 2010. Establishing zebrafish as a novel exercise Schjolden, J., Basic, D., Winberg, S., 2009. Aggression in rainbow trout is inhibited by both
model: swimming economy, swimming-enhanced growth and muscle growth mark- MR and GR antagonists. Physiol. Behav. 98 (5), 625–630. http://dx.doi.org/10.1016/j.
er gene expression. PLoS One 5, e14483. physbeh.2009.09.018.
Palstra, A.P., Rovira, M., Rizo-Roca, D., Torrella, J.R., Spaink, H.P., Planas, J.V., 2014. Swim- Seiliez, I., Gabillard, J.-C., Panserat, S., Skiba-Cassy, S., García de la serrana, D., Gutiérrez, J.,
ming-induced exercise promotes hypertrophy and vascularization of fast skeletal Kaushik, S., Tesseraud, S., 2008. An in vivo and in vitro assessment of TOR signaling
muscle fibres and activation of myogenic and angiogenic transcriptional programs cascade in rainbow trout (Oncorhynchus mykiss). Am. J. Phys. Regul. Integr. Comp.
in adult zebrafish. BMC Genomics 15, 1136. http://dx.doi.org/10.1186/1471-2164- Phys. 295, R329–R335.
15-1136. Seiliez, I., Gutiérrez, J., Salmerón, C., Skiba-Cassy, S., Chauvin, C., Dias, K., Kaushik, S.,
Palstra, A.P., Mes, D., Kusters, K., Roques, J.A.C., Flik, G., Kloet, K., Blonk, R.J.W., 2015. Forced Tesseraud, S., Panserat, S., 2010. An in vivo and in vitro assessment of autophagy-re-
sustained swimming exercise at optimal speed enhances growth of juvenile yellow- lated gene expression in muscle of rainbow trout (Oncorhynchus mykiss). Comp.
tail kingfish (Seriola lalandi). Front. Physiol. 5, 506. Biochem. Physiol. B 157, 258–266.
Párrizas, M., Baños, N., Baró, J., Planas, J., Gutiérrez, J., 1994a. Up-regulation of insulin bind- Seiliez, I., Sabin, N., Gabillard, J.-C., 2011. FoxO1 is not a key transcription factor in the reg-
ing in fish skeletal muscle by high insulin levels. Regul. Pept. 53 (3), 211–222. ulation of myostatin (mstn-1a and mstn-1b) gene expression in trout myotubes. Am.
Párrizas, M., Planas, J., Plisetskaya, E.M., Gutiérrez, J., 1994b. Insulin binding and receptor J. Phys. Regul. Integr. Comp. Phys. 301, R97–R104.
tyrosine kinase activity in skeletal muscle of carnivorous and omnivorous fish. Am. Seiliez, I., Gabillard, J.-C., Riflade, M., Sadoul, B., Dias, K., Avérou, J., Tesseraud, S., Skiba-
J. Phys. 266 (6 Pt 2), R1944–R1950. Cassy, S., Panserat, S., 2012a. Amino acids downregulate the expression of several au-
Párrizas, M., Plisetskaya, E.M., Planas, J., Gutiérrez, J., 1995. Abundant insulin-like growth tophagy-related genes in rainbow trout myoblasts. Autophagy 8 (3), 364–375.
factor-1 (IGF-1) receptor binding in fish skeletal muscle. Gen. Comp. Endocrinol. 98, Seiliez, I., Sabin, N., Gabillard, J.-C., 2012b. Myostatin inhibits proliferation but not differ-
16–25. entiation of trout myoblasts. Mol. Cell. Endocrinol. 351, 220–226.
Párrizas, M., Brugnara, L., Esteban, Y., González-Franquesa, A., Canivell, S., Murillo, S., Seiliez, I., Taty, G.C.T., Bugeon, J., Dias, K., Sabin, N., Gabillard, J.-C., 2013. Myostatin induces
Gordillo-Bastidas, E., Cussó, R., Cadefau, J.A., García-Roves, P.M., Servitja, J.M., atrophy of trout myotubes through inhibiting the TORC1 signaling and promoting
Novials, A., 2015. Circulating miR-192 and miR-193b are markers of prediabetes ubiquitin–proteasome and autophagy-lysosome degradative pathways. Gen. Comp.
and are modulated by an exercise intervention. J. Clin. Endocrinol. Metab. 100 (3), Endocrinol. 186, 9–15.
E407–E415. Seiliez, I., Dias, K., Cleveland, B.M., 2014. Contribution of the autophagy-lysosomal and
Peterson, B.C., Small, B.C., Bosworth, B.G., 2004. Effects of bovine growth hormone ubiquitin-proteasomal proteolytic systems to total proteolysis in rainbow trout (On-
(Posilac®) on growth performance, body composition, and IGFBPs in two strains of corhynchus mykiss) myotubes. Am. J. Phys. Regul. Integr. Comp. Phys. 307 (11),
channel catfish. Aquaculture 232, 651–663. R1330–R1337. http://dx.doi.org/10.1152/ajpregu.00370.2014.
Pierce, A.L., Breves, J.P., Moriyama, S., Hirano, T., Grau, E.G., 2011. Differential regulation of Silahtaroglu, A., Stenvang, J., 2010. MicroRNAs, epigenetics and disease. Essays Biochem.
Igf1 and Igf2 mRNA levels in tilapia hepatocytes: effects of insulin and cortisol on GH 48 (1), 165–185.
sensitivity. J. Endocrinol. 211, 201–210. Silva, A.C., Almeida, D.V., Nornberg, B.F., Figueiredo, M.A., Romano, L.A., Marins, L.F., 2015.
Pottinger, T.G., Carrick, T.R., Hughes, S.E., Balm, P.H.M., 1996. Testosterone, 11- Effects of double transgenesis of somatotrophic axis (GH/GHR) on skeletal muscle
ketotestosterone, and estradiol-17β modify baseline and stress-induced interrenal growth of zebrafish (Danio rerio). Zebrafish 12 (6), 408–413. http://dx.doi.org/10.
and corticotropic activity in trout. Gen. Comp. Endocrinol. 104, 284–295. 1089/zeb.2015.29001.sil.
Power, D.M., Llewellyn, L., Faustino, M., Nowell, M.A., Björnsson, B.T., Einarsdottir, I.E., Snijders, T., Nederveen, J.P., McKay, B.R., Joanisse, S., Verdijk, L.B., van Loon, L.J.C., Parise, G.,
Canario, A.V., Sweeney, G.E., 2001. Thyroid hormones in growth and development 2015. Satellite cells in human skeletal muscle plasticity. F. Physiol. 6, 283. http://dx.
of fish. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 130 (4), 447–459. doi.org/10.3389/fphys.2015.00283.
Preziosa, E., Liu, S., Terova, G., Gao, X., Liu, H., Kucuktas, H., Terhune, J., Liu, Z., 2013. Effect Stickland, N.C., 1983. Growth and development of muscle fibers in the rainbow trout
of nutrient restriction and re-feeding on calpain family genes in skeletal muscle of (Salmo gairdneri). J. Anat. 137 (2), 323–333.
channel catfish (Ictalurus punctatus). PLoS One 8 (3), e59404. http://dx.doi.org/10. Tashima, L., Cahill Jr., G.F., 1968. Effects of insulin in the toadfishOpsanus tau. Gen. Comp.
1371/journal.pone.0059404. Endocrinol. 11 (2), 262–271.
40 E.J. Vélez et al. / Aquaculture 467 (2017) 28–40

Tiago, D.M., Laizé, V., Cancela, M.L., 2008. Alternatively spliced transcripts of Sparus aurata Vong, Q.P., Chan, K.M., Cheng, C.H., 2003. Quantification of common carp (Cyprinus carpio)
insulin-like growth factor 1 are differentially expressed in adult tissues and during IGF-I and IGF-II mRNA by real-time PCR: differential regulation of expression by GH.
early development. Gen. Comp. Endocrinol. 157 (2), 107–115. J. Endocrinol. 178 (3), 513–521.
Tse, D.L.Y., Tse, M.C.L., Chan, C.B., Deng, L., Zhang, G.M., Lin, H.R., Cheng, C.H.K., 2003. Walock, C.N., Kittilson, J.D., Sheridan, M.A., 2014. Characterization of a novel growth hor-
Seabream growth hormone receptor: molecular cloning and functional studies of mone receptor-encoding cDNA in rainbow trout and regulation of its expression by
the full-length cDNA, and tissue expression of two alternatively spliced forms. nutritional state. Gene 533, 286–294. http://dx.doi.org/10.1016/j.gene.2013.09.046.
Biochim. Biophys. Acta 1625, 64–76. Wang, X., Lu, L., Li, Y., Li, M., Chen, C., Feng, Q., Zhang, C., Duan, C., 2009. Molecular and
Upton, Z., Yandell, C.A., Degger, B.G., Chan, S.J., Moriyama, S., Francis, G.L., Ballard, F.J., functional characterization of two distinct IGF binding protein-6 genes in zebrafish.
1998. Evolution of insulin-like growth factor-I (IGF-I) action: in vitro characterization Am. J. Phys. Regul. Integr. Comp. Phys. 296, R1348–R1357.
of vertebrate IGF-I proteins. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 121 (1), Wilkinson, R.J., Elliott, P., Hohmann, A., Francis, G., Carragher, J., 2004. Development and
35–41. characterization of a competitive polyclonal antibody enzyme-immunoassay for
Vélez, E.J., Lutfi, E., Jiménez-Amilburu, V., Riera-Codina, M., Capilla, E., Navarro, I., salmon insulin-like growth factor-II. Comp. Biochem. Physiol. B Biochem. Mol. Biol.
Gutiérrez, J., 2014. IGF-I and amino acids effects through TOR signaling on prolifera- 139 (2), 193–201.
tion and differentiation of gilthead sea bream cultured myocytes. Gen. Comp. Wilkinson, R.J., Porter, M., Woolcott, H., Longland, R., Carragher, J.F., 2006. Effects of aqua-
Endocrinol. 205, 296–304. culture related stressors and nutritional restriction on circulating growth factors (GH,
Vélez, E.J., Lutfi, E., Azizi, Sh., Montserrat, N., Riera-Codina, M., Capilla, E., Navarro, I., IGF-I and IGF-II) in Atlantic salmon and rainbow trout. Comp. Biochem. Physiol. A
Gutiérrez, J., 2015. Contribution of in vitro myocytes studies to understanding fish Mol. Integr. Physiol. 145 (2), 214–224.
muscle physiology. Comp. Biochem. Physiol. B http://dx.doi.org/10.1016/j.cbpb. Won, E.T., Borski, R.J., 2013. Endocrine regulation of compensatory growth in fish. Front.
2015.12.003. Endocrinol. (Lausanne) 4, 74. http://dx.doi.org/10.3389/fendo.2013.00074.
Vélez, E.J., Azizi, Sh., Millán-Cubillo, A., Fernández-Borrás, J., Blasco, J., Chan, S.J., Calduch- Yogata, H., Oku, H., 2000. The effects of swimming exercise on growth and whole-body
Giner, J.A., Pérez-Sánchez, J., Navarro, I., Capilla, E., Gutiérrez, J., 2016. Effects of protein and fat contents of fed and unfed fingerling yellowtail. Fish. Sci. 66,
sustained exercise on GH-IGFs axis in gilthead sea bream (Sparus aurata). Am. 1100–1105.
J. Phys. Regul. Integr. Comp. Phys. 310, R313–R322.

You might also like