You are on page 1of 8

General and Comparative Endocrinology 172 (2011) 323–330

Contents lists available at ScienceDirect

General and Comparative Endocrinology


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

Review

Regulation of crustacean molting: A review and our perspectives


Ernest S. Chang a,⇑, Donald L. Mykles b
a
Bodega Marine Laboratory, University of California-Davis, Bodega Bay, CA 94923, USA
b
Department of Biology, Colorado State University, Fort Collins, CO 80523, USA

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

Article history: Molting is a highly complex process that requires precise coordination to be successful. We describe the
Received 2 February 2011 early classical endocrinological experiments that elucidated the hormones and glands responsible for this
Revised 30 March 2011 process. We then describe the more recent experiments that have provided information on the cellular
Accepted 4 April 2011
and molecular aspects of molting. In addition to providing a review of the scientific literature, we have
Available online 8 April 2011
also included our perspectives.
Ó 2011 Elsevier Inc. All rights reserved.
Keywords:
20-Hydroxyecdysone
Crustacean
Decapod
Ecdysone
Ecdysteroid
Molting

1. Introduction [13]. Several years later, the structure of ecdysone was determined
and its source traced to the prothoracic gland [53,55]. From many
To achieve growth, arthropods must initially loosen the connec- subsequent studies, 20-hydroxyecdysone (20E)1 has been charac-
tives between their living tissues and their extracellular cuticles, terized as the physiologically active form of the ecdysteroids (a
escape from the confines of these cuticles relatively rapidly, take class of steroid molting hormones related in structure to 20E) in
up water or air to expand the new, flexible exoskeletons, and then most species. Parallel studies on the ecdysteroids of crustaceans
quickly harden them for defense and locomotion. Each of the major led to observations that the primary ecdysteroid of crustaceans
arthropodan taxa, the crustaceans and insects, has provided valu- was also 20E [46,52].
able models for the study of the endocrine regulation of molting. Classical morphological observations and endocrinological
In this review, we focus on the Crustacea. It is not comprehensive experiments involving organ ablation [36,40] indicated that the
and contains some of our personal accounts of the journeys that molting gland in the crab Carcinus maenas was the thoracic Y-organ
led to our current state of knowledge of the regulation of crusta- (YO). In brachyurans, the YOs are spheroidal glands located ven-
cean molting. trally and anteriorly in the gill chambers. The macruan YO is more
difficult to locate since it is a monolayer sheet of cells. While in J.
2. Classical endocrinological experiments Dennis O’Connor’s laboratory, Chang (EC) began organ culture
experiments using YOs from the crabs Cancer antennarius and
The molting process is a cyclical event, the culmination of Pachygrapsus crassipes. These experiments involved the character-
which is the actual ecdysis, or shedding of the exoskeleton ization of ecdysone as the primary secretory product of the YO
(Fig. 1). The prohormone of the steroid molting hormone, ecdy- [18]. Those results indicated that ecdysone was the prohormone
sone, was isolated from hundreds of kilograms of moth pupae for 20E. Experimental animals were collected by hand at night un-
der rocks at Palos Verdes, California. It was due partially to those
⇑ Corresponding author. Fax: +1 707 875 2091. collection trips that convinced EC that future scientific endeavors
E-mail address: eschang@ucdavis.edu (E.S. Chang). should be more laboratory-intensive as opposed to field-based.
1
Abbreviations used: 20E, 20-hydroxyecdysone; CaM, calmodulin; CHH, crustacean Those YO experiments involved the determination of appropri-
hyperglycemic hormone; GC, guanylyl cyclase; GC-I, nitric oxide-sensitive guanylyl ate organ culture experiments. Defined media, such as M199
cyclase; ILP, insulin-like peptide; LAFan, Limb Autotomy Factor–Anecdysis; LAFpro, provided prolonged secretory activity of the YO compared to a
Limb Autotomy Factor–Proecdysis; LB, limb bud; LBA, limb bud autotomy; mTOR,
simple saline solution. However, since identification of the secre-
metazoan target of rapamycin; MIH, molt-inhibiting hormone; NO, nitric oxide; NOS,
nitric oxide synthase; PDE, phosphodiesterase; TGFb, transforming growth factor-b; tory product required collection and concentration of hundreds
YO, Y-organ. of milliliters of culture media, the saline solution provided a less

0016-6480/$ - see front matter Ó 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.ygcen.2011.04.003
324 E.S. Chang, D.L. Mykles / General and Comparative Endocrinology 172 (2011) 323–330

Fig. 1. Molting (or ecdysis) of a juvenile lobster (Homarus americanus). The entire sequence took about 30 min from start to finish. Molting is the culmination of a cyclic
process mediated by the steroid hormone 20-hydroxyecdysone. (A) The lobster has resorbed much of its old exoskeleton’s mineralization. The exoskeleton splits at the
junction of the thorax and abdomen. (B) The anterior of the animal retracts from its old exoskeleton by pulling posteriorly. (C) The posterior of the lobster pulls anteriorly. (D)
The lobster is free of its old exoskeleton and begins to take up water to expand its new, flexible, larger exoskeleton. (E) The postmolt lobster is above its shed exoskeleton. It
will take several days for the epidermis to deposit layers of chitin, protein, and calcium carbonate into the exoskeleton. The molt cycle will continue for several days to
months, depending upon the size and age of the animal. Photos by staff of the Bodega Marine Laboratory. Reproduced with permission [16].

contaminated secretory product for eventual mass spectrophoto- hemal organ in the eyestalk of several decapod crustaceans [9,92].
metric analyses compared to the product secreted into M199. This neurohemal organ is called the sinus gland and serves as a
Similar results were obtained from the crayfish Orconectes limo- storage site for neurosecretory products. It consists of the enlarged
sus [57] and the blue crab Callinectes sapidus [99]. More recent data endings of a group of neurosecretory neurons collectively called
have revealed that there are other ecdysteroids secreted by the YOs the X-organ [47]. The shortened molt interval observed in eye-
in some species. These ecdysteroids include 3-dehydroecdysone stalk-ablated decapods is likely due to a rapid elevation in the con-
from C. antennarius [114] and 25-deoxyecdysone from C. maenas centration of circulating ecdysteroids, which is a result of X-organ/
[64] (see [83,121], for reviews). A productive area of future re- sinus gland removal [15,22]. Conversely, the demonstration of de-
search would be the determination of the types and ratios of the creased ecdysteroid titers in eyestalk-ablated crabs that have been
different ecdysteroids secreted by the YOs from various other crus- injected with eyestalk extracts [50,56] lends additional support to
tacean species. The functional significance of these differences is the paradigm of the MIH–YO molt-controlling axis. Further evi-
unclear. dence of this hypothesis was provided by Gersch et al. [41], who
Hemolymph ecdysteroid concentration fluctuates dramatically demonstrated that if crayfish (O. limosus) were initially injected
during the molt cycle (e.g., from <10 pg/ll in postmolt lobster to with sinus gland extracts, the subsequent culture of the YOs re-
>350 pg/ll in premolt lobster) [17] and these changes mediate sulted in a decreased production of ecdysone compared to vehi-
the various biochemical and physiological processes that occur cle-injected animals. In vitro secretion of ecdysteroid by crab YOs
during the cycle. The rate of synthesis and/or secretion of ecdy- could be inhibited when cultured with either conditioned medium
sone by the YO vary during the molt cycle and partially explain that had previously been incubated with explanted sinus glands or
these hemolymph fluctuations in ecdysteroid titer. Just prior to by the addition of eyestalk extracts [74,99,111,122]. In Homarus
the substage of premolt in which the highest concentration of americanus, EC’s laboratory demonstrated that injection of sinus
ecdysteroids was observed in the hemolymph, explanted YOs gland extracts decreased circulating titers of ecdysteroids and in-
were found to secrete the greatest amount of ecdysone [19]. creased the molt interval [12].
Low hemolymph concentrations were correlated with low secre- MIH from C. maenas was among the first of the MIHs to be char-
tory rates. This led to the question – what controls the secretory acterized [123]. EC’s laboratory worked on MIH from H. americanus
rate of the YO? [21]. At that time, Prof. Rainer Keller was visiting EC’s laboratory.
Over a century ago, removal of both stalked eyes of the crab Uca Keller had remembered much of the C. maenas MIH sequence
pugilator [130] resulted in a dramatic shortening of the molt inter- and was very excited to see the similarities in amino acids when
val (length of the molt cycle). This was later confirmed in crayfish he was shown the lobster sequence.
[107] and many other species. These observations led to the postu- MIH is a member of a novel neuropeptide family. Representa-
lation of an endocrine factor present in the eyestalks that normally tives of this family have only been found in arthropods so far
inhibits molting – a molt-inhibiting hormone (MIH). Detailed [37]. This neuropeptide family regulates such diverse functions
microscopical examinations resulted in the description of a neuro- as molting, reproduction, and metabolism [10,15,34,120,125].
E.S. Chang, D.L. Mykles / General and Comparative Endocrinology 172 (2011) 323–330 325

MIH binds to hormone receptors on the membranes of YO cells laboratory at Oak Ridge National Laboratory in 1979 to study a
[124] and likely mediates its action via cyclic nucleotide second molt-induced claw muscle atrophy that Skinner reported in 1966
messengers [97](see [31], for review) or through alterations in [80,103]. DM continued studies on G. lateralis when he moved to
calcium ion fluxes and activity of protein kinase C that Colorado State University in 1985. While in residence at the
phosphorylates enzymes involved in steroidogenesis [113]. The Bodega Marine Laboratory 1998–1999, DM began a collaboration
mode of action of MIH is discussed below. with EC to study the endocrine regulation of molting in G. lateralis,
EC’s group has also worked on the hypothesis that ecdysteroids resulting in the characterization of LAFpro [128]. Subsequent work
may feed back upon the X-organ, the YO, and/or other target tis- has been directed toward neuropeptide signaling pathways in the
sues to mediate normal ecdysis. Chang and O’Connor [19] showed YO and ecdysteroid control of muscle atrophy [29,30,33,58–60,66–
that the activity of various tissues to hydroxylate ecdysone to 20E 70,76,129]. Thus, G. lateralis has been an important experimental
was inhibited with the addition of exogenous 20E. Cheng and animal for crustacean molting physiology for more than 60 years.
Chang [23] demonstrated that normal ecdysis in lobsters could
be delayed by the injection of exogenous 20E. Those results indi- 3.2. Regulation of the Y-organ by eyestalk neuropeptides
cated that the normal completion of processes, such as ecdysis, re-
quired not only increases in hormone concentrations, but also Both MIH and a related peptide, crustacean hyperglycemic hor-
required their subsequent decline. The mechanism of this ecdy- mone (CHH), inhibit YO ecdysteroidogenesis through separate sig-
steroid inhibition is unknown. El Haj et al. [38] published findings naling pathways that converge at cGMP [24,39]. YO membranes
that ecdysteroid receptors are located in eyestalk neural tissue. have distinct receptors for CHH and MIH [2,24,27,124]. The CHH
Those data hinted at a possible positive or negative feedback regu- receptor appears to be a membrane guanylyl cyclase (GC)
lation upon MIH secretion. We recently obtained preliminary data [26,43], whereas the identity of the MIH receptor is not firmly
that a non-steroidal ecdysteroid analog (tebufenozide) was able to established. Thus, binding of CHH leads directly to an increase in
inhibit ecdysone secretion from crab YOs in vitro [90]. This effect cGMP [26]. By contrast, MIH signaling usually involves a transient
was observed at 50 and 100 pg/ll of culture medium, but not at increase in cAMP, followed by a larger, sustained increase in cGMP
lower concentrations. This ecdysteroid inhibition of ecdysone [4,11,31,87,89,97,100,119]. The delayed increase in cGMP suggests
secretion may account for the observation that hormone titers in that MIH activates a soluble GC, as activation of a membrane GC
eyestalk-ablated lobsters continue to cycle up and down, even would result in an immediate increase in cGMP. Both cAMP and
though the eyestalk tissue does not regenerate [14]. cGMP inhibit YO ecdysteroidogenesis in vitro (see [31], for review).
Our recent work indicates that nitric oxide (NO)-sensitive GC (GC-
I) is involved. GC-I agonists (NO donors and YC-1) can inhibit YO
3. Molecular regulation of molting ecdysteroidogenesis in vitro [29,84]. Moreover, YOs express a
Ca2+/CaM-dependent NO synthase (NOS) and the catalytic (b) sub-
3.1. Limb factors unit of GC-I [58,68,70,76].
Much of the data support the organization of the MIH signaling
A variety of signals affect YO activity. External cues, such as pathway into a cAMP/Ca2+-dependent ‘‘triggering’’ phase and a NO/
stress, temperature extremes, crowding, and short photoperiod cGMP-dependent ‘‘summation’’ phase linked by calmodulin (CaM)
can inhibit molting [1,8,48,78,93]. In the blackback land crab, [32,70,84]. A transient increase in cAMP triggers the influx of Ca2+,
Gecarcinus lateralis, loss of five or more walking legs, termed multi- which activates CaM (Fig. 2). A sustained activation of NOS results
ple leg autotomy or MLA, induces molting as growth of limb buds from the combined effects of direct binding of Ca2+/CaM to NOS
(LBs) is restricted to premolt. Upon ecdysis the LBs are extended and the dephosphorylation of NOS by calcineurin, which is a
and become functional appendages [8,104,106]. However, limb Ca2+/CaM-dependent protein phosphatase. Phosphorylation of
bud autotomy (LBA) during early premolt suspends molting 2– NOS reduces its activity; dephosphorylation by calcineurin en-
3 weeks, allowing time for a secondary (2°) LB to grow, so that ani- hances NOS activity [63]. NOS is phosphorylated in the activated
mals molt with a full complement of legs [49,82]. These studies led YO, which is consistent with inactivation of NOS by protein kinases
Dorothy Skinner to propose that LBs produce stimulatory and in the absence of MIH [70]. This is similar to the signaling mecha-
inhibitory factors that she designated Limb Autotomy Factor– nism that stimulates fluid secretion in Drosophila Malpighian tu-
Anecdysis or LAFan and Limb Autotomy Factor–Proecdysis or LAF- bules by the decapeptide cardioacceleratory peptide 2b (see [5],
pro, respectively [104]. LAFan is produced by primary (1°) LBs and for review). MIH is released in pulses, with each pulse having a
stimulates molting in response to MLA. The identity of LAFan half-life of 5–10 min; this results in low MIH titers (<5 fmol/ml)
remains unknown. LAFpro is a MIH-like factor produced by 2° LBs in the hemolymph of most intermolt animals [25]. The sustained
that inhibits 1° LB growth by lowering hemolymph ecdysteroid activation of GC-I in the model provides a mechanism for the inhi-
titers [128]. By mid premolt the animal becomes committed to bition of the YO between pulses, thus explaining how the YO is
molt and, consequently, interventions such as LBA and MIH injec- suppressed for extended periods when average MIH titers are
tions fail to suspend premolt and animals molt without delay low in intermolt animals.
[49,128].
Gecarcinus lateralis is an excellent experimental organism for 3.3. Changes in the Y-organ over the molt cycle
the study of the endocrine control of molting. These air-breathing
animals occur throughout the Caribbean, living in burrows they dig The YO is a dynamic organ that transitions through four physi-
in dune areas above coral sand beaches [6,7]. They are easy to ological states during the molt cycle (Fig. 3). During postmolt
maintain in the laboratory and molting is easily manipulated. (stages A, B, and C1–3) and intermolt (stage C4), the YO is in the ba-
Dorothy Bliss pioneered the use of the G. lateralis in the 1950s sal state, resulting in low (<20 pg/ll) ecdysteroid titers in the
[9] (see [73] for a complete bibliography). In 1962, Skinner pub- hemolymph. A reduction in MIH triggers the transition from the
lished her first paper on G. lateralis, reporting the first microscopic basal state to the activated state; the increasing hemolymph ecdy-
analysis of the changes in integumentary structure during the molt steroid titers initiate premolt processes in the integument (e.g.,
cycle [102], and it became her primary experimental animal for separation of the epithelium from the exoskeleton or apolysis),
nearly four decades (see [86], for a complete bibliography). Mykles gastric epithelium (e.g., gastrolith formation in certain species),
(DM) was introduced to G. lateralis when he joined the Skinner skeletal muscle (e.g., claw muscle atrophy), and limb regenerates
326 E.S. Chang, D.L. Mykles / General and Comparative Endocrinology 172 (2011) 323–330

MIH Ca2+ channel


MIH-R Cell membrane

G AC
PKA

Triggering phase
ATP
cAMP

Ca2+
PDE1

PKA
AMP
CaM

NOS
Pi
CaN

Arginine NOS
dephosphorylation

Summation phase
Pi

Citrulline
NO

GC-I

PDE5
GTP cGMP GMP

PKG

Ecdysteroid synthesis
(Transcriptional/Translational Regulation)

Fig. 2. Proposed MIH signaling pathway regulating ecdysteroidogenesis in decapod crustacean molting gland. The ‘‘triggering’’ phase is initiated by binding of MIH to a G
protein-coupled receptor (MIH-R) and activation of adenylyl cyclase (AC); cAMP increases intracellular Ca2+ via cAMP-dependent protein kinase (PKA) phosphorylation of
Ca2+ channels. Sensitivity to MIH is determined by phosphodiesterase 1 (PDE1) activity, which varies during the molting cycle. The ‘‘summation’’ phase is mediated by NO and
cGMP. Calmodulin (CaM) links the two phases by activating NO synthase (NOS) directly and indirectly via calcineurin (CaN). Dephosphorylation of NOS by CaN can potentially
prolong the response to MIH. CaM can also activate PDE1 to inhibit the triggering phase (PDE1 can also hydrolyze cGMP, thus inhibiting the summation phase). cGMP-
dependent protein kinase (PKG) inhibits ecdysteroidogenesis. Chronic activation of PKA may directly inhibit ecdysteroidogenesis. Chronic elevated intracellular cAMP can
inhibit ecdysteroidogenesis directly, perhaps by inhibiting protein synthesis. Other abbreviations: G, G protein; GC-I, NO-sensitive guanylyl cyclase; PDE5, cGMP PDE. From
[32].

(e.g., LB growth) in early premolt (stage D0) (see [51,80– YO activation at stage D0 is triggered by a decrease in the re-
82,96,105,126], for reviews). At mid premolt (stage D1), the YO lease of MIH from the sinus gland. This de-represses the YO, pre-
transitions from the activated to committed state; the YO increases sumably through inactivation of MIH signaling components,
ecdysteroid biosynthesis and hemolymph ecdysteroid titers reach resulting in hypertrophy and increased ecdysteroid biosynthetic
a peak (>300 pg/ll) at stage D2. LB growth, claw muscle atrophy, capacity (Fig. 3). NOS is phosphorylated in YOs from 1 day eye-
and gastrolith formation are completed by stage D2 [105]. The high stalk-ablated animals, suggesting the NOS is inactivated in the ab-
ecdysteroids trigger the transition of the YO from the committed sence of MIH [70]. The downstream targets are largely unknown,
state to the repressed state in late premolt (D3–4). At ecdysis, the but activation probably involves transcriptional, translational,
YO transitions back to the basal state in postmolt when ecdysteroid and posttranslational regulation. YO activation results in signifi-
titers are low. cant (>3-fold) changes in the levels of hundreds of proteins [70].
E.S. Chang, D.L. Mykles / General and Comparative Endocrinology 172 (2011) 323–330 327

Early Mid Late


Intermolt Premolt Premolt Premolt E Postmolt
(C4) (D 0) (D 1,2) (D3,4) (A, B, C1-3)

Hemolymph
ecdysteroid
synthetic capacity
YO ecdysteroid

YO State: Basal Activated Committed Repressed Basal


Sensitivity to MIH: High High Low Low High
Limb regeneration (R Index): 0 - 10 11 - 15 16 - 23 23

Fig. 3. Hormonal regulation of molting in the blackback land crab, Gecarcinus lateralis. Diagram shows the relationship between molt stage, YO state, YO sensitivity to MIH,
limb regeneration (R index), YO ecdysteroid synthetic capacity, and hemolymph ecdysteroid titer. During postmolt (A, B, C1–3), intermolt (C4), early premolt (D0), and mid
premolt (D1,2), hemolymph ecdysteroid titers are correlated with YO synthetic capacity; during late premolt (D3,4), high ecdysteroid represses YO ecdysteroidogenesis and
ecdysteroid titer falls. The YO transitions through four physiological states during the molt cycle: basal (B), activated (A), committed (C), and repressed (R). The B to A
transition is triggered by a reduction in MIH; the YOs hypertrophy, but remain sensitive to MIH, as premolt is suspended by MIH injection or by limb bud autotomy (LBA). At
the A to C transition, the animal becomes committed to molt, as the YO is less sensitive to MIH and premolt is not suspended by LBA; this transition may be triggered by an
increase in MIH or an unidentified tropic factor. At the C to R transition, YO ecdysteroid synthetic capacity remains high, but high hemolymph ecdysteroid titer inhibits
ecdysteroid secretion. Molting, or ecdysis (E), marks the R to B transition, during which the YO atrophies and becomes sensitive to MIH.

At the transcriptional level, Phantom (Phm), a key enzyme in the [24,88,89]. As neither MIH nor CHH binding to high-affinity recep-
ecdysteroid biosynthetic pathway, is up-regulated during premolt; tors is reduced during premolt [124], the reduction in sensitivity
it encodes a cytochrome P-450 mono-oxygenase that hydroxylates likely results from a down-regulation of signaling components
ecdysteroid precursors at the #25 carbon (see [83], for review). In ‘‘downstream’’ from the receptors. Increased phosphodiesterase
shrimp, Marsupenaeus japonicus, Phm expression in the YO in- (PDE) activity contributes to the reduced response to MIH by keep-
creases as much as 7-fold during premolt and is decreased about ing intracellular cyclic nucleotides low [89]. However, the magni-
2.5-fold by sinus gland extract and recombinant MIH [3]. tude of the increase in PDE activity (2.5-fold) is less than that
Translational regulation is also involved in YO activation. In the of the decrease in MIH sensitivity (10-fold), suggesting that other
C. antennarius YO, the translation inhibitor cycloheximide inhibits components of the MIH signaling pathway, such as NOS and GC-I,
both protein synthesis and ecdysteroidogenesis [75]. By contrast, are down-regulated. In G. lateralis and C. maenas YOs, NOS and GC-
the transcriptional inhibitor actinomycin D inhibits RNA synthesis Ib mRNA levels are increased by eyestalk ablation, indicating that
without having any effect on ecdysteroidogenesis [75]. A potential the expression of these genes is responsive to an acute withdrawal
target of MIH signaling is metazoan target of rapamycin (mTOR), of MIH and other neuropeptides [68,76].
which functions as a nutrient sensor for cellular growth in animals The regulation of the transition from the activated to the com-
(see [94], for review). In insects, mTOR stimulates ecdysteroido- mitted state is largely unknown. It may be triggered by an increase
genesis of the prothoracic gland. Nutrients and insulin-like pep- in MIH (see [32], for review). However, other autocrine/paracrine
tides activate mTOR, which phosphorylates components of the factors may be involved. Members of the transforming growth fac-
protein synthetic machinery, such as p70-S6 kinase and eIF4E- tor-b (TGFb) superfamily of cytokines are potential candidates.
binding protein, to increase translation of mRNA (see [116], for re- They are grouped into the TGFb/Activin/Nodal and BMP/GDF/MIS
view). Ecdysteroidogenesis in the prothoracic gland of Bombyx mori families (see [71], for review). TGFb activates Smad transcription
is stimulated by insulin [45]. In Drosophila melanogaster, the mTOR factors, which regulate gene expression through transcriptional
pathway controls prothoracic gland size and ecdysteroidogenic activation or repression (see [79,118], for reviews). The only
capacity [28,65,77]. mTOR activity may be required for full ecdys- TGFb/Activin/Nodal members that have been characterized in
teroidogenesis in the crustacean YO, as rapamycin, an mTOR inhib- decapods are myostatin (Mstn)-like factors; cDNAs have been
itor, is a potent inhibitor of ecdysteroid secretion by G. lateralis and cloned from G. lateralis, C. maenas, and other decapods
C. maenas YOs in vitro [95]. [33,61,62,72] and may play a role in muscle protein turnover
A hallmark feature of the ‘‘committed’’ YO is that it becomes [30]. Sequences encoding a TGFb factor distinct from Mstn are re-
refractory to MIH and CHH. In C. maenas and crayfish (Procambarus ported for lobster and shrimp in the GenBank database (Accession
clarkii), the sensitivity to MIH declines during premolt and is least Nos. CN854252 and CAA72411). We hypothesize that a factor like
sensitive to MIH by the end of mid premolt and late premolt TGFb is synthesized as an autocrine factor during early premolt and
328 E.S. Chang, D.L. Mykles / General and Comparative Endocrinology 172 (2011) 323–330

alters gene expression in the YO by down-regulating MIH signaling Acknowledgments


components and up-regulating mTOR signaling components and
ecdysteroid synthetic enzymes, such as Phm. This transitions the The authors are grateful to Howard Bern for his mentorship,
YO to the committed state. The animal is now committed to molt, beginning when EC was an undergraduate student and DM was a
as interventions (e.g., LBA or MIH injections) fail to delay molting graduate student at UC Berkeley. DM acknowledges the unfailing
[44,49,88,91]. guidance and generous support of Dorothy Skinner until her death
Repression of the YO in late premolt involves a negative feed- in 2005. Her discoveries in crustacean molting biology are a lasting
back inhibition by ecdysteroids. At the end of premolt there is a legacy. The authors are grateful to the many students, technicians,
sudden drop in hemolymph ecdysteroids within a few days of postdoctoral fellows, and visiting scientists who contributed to this
ecdysis [105]. This drop appears to determine the timing of ecdysis, work over the last 25 years. The authors thank the Editors for the
as artificially elevated ecdysteroid during late premolt delays ecdy- invitation to participate in this project and Sharon Chang for edito-
sis [23,42]. It results from an increase in ecdysteroid excretion and rial assistance. Finally, the authors acknowledge the support of
a decrease in YO ecdysteroid production [20,83,108–110,112]. 20E grants from the California and Connecticut Sea Grant College Pro-
inhibits YO ecdysteroidogenesis when injected into crayfish [35]. grams, the National Institutes of Health, and the National Science
RH-5849 (1,2-dibenzoyl-1-tert-butylhydrazine), a non-steroidal Foundation.
ecdysteroid agonist that mimics the effects of 20E in insects
[127], inhibits ecdysteroid secretion by crayfish YOs in vitro [35].
Both treatments produce significant reductions in ecdysteroido-
References
genesis within 1 h [35], suggesting a non-genomic response
mediated by G protein-coupled and/or membrane-associated [1] D.E. Aiken, Photoperiod endocrinology and crustacean molt cycle, Science 164
ecdysteroid receptors [54,98,115,117]. This inhibition lasts at least (1969) 149–155.
24 h after a single 20E injection [35], which suggests that ecdyster- [2] H. Asazuma, S. Nagata, H. Katayama, T. Ohira, H. Nagasawa, Characterization
of a molt-inhibiting hormone (MIH) receptor in the Y-organ of the kuruma
oid may also affect gene expression. prawn, Marsupenaeus japonicus, Ann. NY Acad. Sci. 1040 (2005) 215–218.
[3] H. Asazuma, S. Nagata, H. Nagasawa, Inhibitory effect of molt-inhibiting
hormone on Phantom expression in the Y-organ of the kuruma prawn,
Marsupenaeus japonicus, Arch. Insect Biochem. Physiol. 72 (2009) 220–233.
[4] D. Baghdassarian, N. de Besse, B. Saïdi, G. Sommé, F. Lachaise, Neuropeptide-
induced inhibition of steroidogenesis in crab molting glands: involvement of
4. Future research cGMP-dependent protein kinase, Gen. Comp. Endocrinol. 104 (1996) 41–51.
[5] K.W. Beyenbach, H. Skaer, J.A.T. Dow, The developmental, molecular, and
transport biology of Malpighian tubules, Ann. Rev. Entomol. 55 (2010) 351–
For many years, crustacean endocrinologists were guided by a 374.
very simple model, in which molting is regulated solely by the le- [6] D.E. Bliss, Transition from water to land in decapod crustaceans, Am. Zool. 8
vel of MIH circulating in the hemolymph. If this were so, then MIH (1968) 355–398.
[7] D.E. Bliss, From sea to tree – Saga of a land crab, Am. Zool. 19 (1979) 385–410.
levels should be high in intermolt and postmolt and low in pre- [8] D.E. Bliss, J.R. Boyer, Environmental regulation of growth in the decapod
molt. However, measurements of MIH in the hemolymph during crustacean Gecarcinus lateralis, Gen. Comp. Endocrinol. 4 (1964) 15–41.
the molt cycle do not show this pattern. During intermolt MIH le- [9] D.E. Bliss, J.H. Welsh, The neurosecretory system of brachyuran Crustacea,
Biol. Bull. 103 (1952) 157–169.
vel is not always high, and during mid- to late-premolt MIH levels [10] D. Böcking, H. Dircksen, R. Keller, The crustacean neuropeptides of the CHH/
can be higher than that during intermolt [25,88]. This indicates MIH/GIH family: Structures and biological activities, in: K. Weise (Ed.), The
that the hormonal control of molting is more complex. Any model Crustacean Nervous System, Springer-Verlag, Berlin, 2002, pp. 84–97.
[11] D. Böcking, D. Sedlmeier, Protein phosphorylation in the molting gland of the
must accommodate the dynamic changes in the YO during the crayfish, Orconectes limosus – role of cyclic nucleotides, calcium, and molt
molt cycle. The release of MIH from the X-organ/sinus gland com- inhibiting hormone (MIH), Invert. Reprod. Dev. 26 (1994) 237–245.
plex is episodic rather than continuous, resulting in relatively low [12] M.J. Bruce, E.S. Chang, Demonstration of a molt-inhibiting hormone from the
sinus gland of the lobster, Homarus americanus, Comp. Biochem. Physiol. A 79
average MIH concentrations in the hemolymph [25]. As brief expo- (1984) 421–424.
sures of YOs to MIH have lasting effects on ecdysteroidogenesis [13] A. Butenandt, P. Karlson, Über die Isolierung eines Metamorphose-Hormons
[101,111], it is thought that periodic releases of MIH are sufficient der Insekten in Kristallisierter Form, Z. Naturforsch. B 9 (1954) 389–391.
[14] E.S. Chang, Hormonal control of molting in decapod Crustacea, Am. Zool. 25
to keep the YO inactive during intermolt [25]. YOs from late pre-
(1985) 179–185.
molt (stage D2–D3) animals are refractory to MIH [24,88,89], which [15] E.S. Chang, Comparative endocrinology of molting and reproduction: insects
may result, at least in part, from increased degradation of cyclic and crustaceans, Annu. Rev. Entomol. 38 (1993) 161–180.
nucleotides by PDEs [89]. Ecdysteroids inhibit YO ecdysteroid [16] E.S. Chang, Invertebrate hormones and behavior, in: M.D. Breed, J. Moore
(Eds.), Encyclopedia of Animal Behavior, Academic Press, Oxford, 2010, pp.
secretion, thus contributing to the rapid decrease in hemolymph 209–215.
ecdysteroids at the end of premolt [35]. [17] E.S. Chang, M.J. Bruce, Ecdysteroid titers of juvenile lobsters following molt
Future research should be directed toward understanding the induction, J. Exp. Zool. 214 (1980) 157–160.
[18] E.S. Chang, J.D. O’Connor, Secretion of a-ecdysone by crab Y-organs in vitro,
signaling mechanisms underlying the transitions between physio- Proc. Natl. Acad. Sci. USA 74 (1977) 615–618.
logical states. A biosystems approach is needed to fully understand [19] E.S. Chang, J.D. O’Connor, In vitro secretion and hydroxylation of a-ecdysone
the regulation of the YO at transcriptional, translational, and post- as a function of the crustacean molt cycle, Gen. Comp. Endocrinol. 36 (1978)
151–160.
translational levels, and how these interact with environmental [20] E.S. Chang, J.D. O’Connor, Crustacea: molting, in: H. Laufer, R.G.H. Downer
signals [70,85]. High throughput sequencing and mass spectrome- (Eds.), Endocrinology of Selected Invertebrate Types, Alan R. Liss, Inc., 1988,
try technologies now make it possible to obtain complete tran- pp. 259–278.
[21] E.S. Chang, G.D. Prestwich, M.J. Bruce, Amino acid sequence of a peptide with
scriptome and proteomic databases of the YO and to quantify both molt-inhibiting and hyperglycemic activities in the lobster, Homarus
mRNA levels and protein levels and posttranslational modifications americanus, Biochem. Biophys. Res. Commun. 171 (1990) 818–826.
at critical transitions during the molt cycle (Fig. 3). A complete [22] E.S. Chang, B.A. Sage, J.D. O’Connor, The qualitative and quantitative
determinations of ecdysones in tissues of the crab, Pachygrapsus crassipes,
transcriptome will facilitate identification of proteins and phos-
following molt induction, Gen. Comp. Endocrinol. 30 (1976) 21–33.
phorylation sites by mass spectrometry. We expect that this anal- [23] J.H. Cheng, E.S. Chang, Ecdysteroid treatment delays ecdysis in the lobster,
ysis will identify genes in the MIH, mTOR, and TGFb signaling Homarus americanus, Biol. Bull. 181 (1991) 169–174.
pathways, as well as identify other genes important in YO regula- [24] J.S. Chung, S.G. Webster, Moult cycle-related changes in biological activity of
moult-inhibiting hormone (MIH) and crustacean hyperglycaemic hormone
tion and function (e.g., Phm and other Halloween genes [83]). It (CHH) in the crab, Carcinus maenas. From target to transcript, Eur. J. Biochem.
may also identify candidate genes encoding the MIH receptor. 270 (2003) 3280–3288.
E.S. Chang, D.L. Mykles / General and Comparative Endocrinology 172 (2011) 323–330 329

[25] J.S. Chung, S.G. Webster, Dynamics of in vivo release of molt-inhibiting [53] R. Huber, W. Hoppe, Zur Chemie des Ecdysons. VII: Die Kristall- und
hormone and crustacean hyperglycemic hormone in the shore crab, Carcinus Molekülstruktur Analyse des Insektenverpuppungshormons Ecdyson mit
maenas, Endocrinology 146 (2005) 5545–5551. der automatisierten Faltmolekulmethode, Chem. Ber. 98 (1965) 2403–2424.
[26] J.S. Chung, S.G. Webster, Binding sites of crustacean hyperglycemic hormone [54] M. Iga, M. Iwami, S. Sakurai, Nongenomic action of an insect steroid hormone
and its second messengers on gills and hindgut of the green shore crab, in steroid-induced programmed cell death, Mol. Cell. Endocrinol. 263 (2007)
Carcinus maenas: a possible osmoregulatory role, Gen. Comp. Endocrinol. 147 18–28.
(2006) 206–213. [55] P. Karlson, H. Hoffmeister, H. Hummel, P. Hocks, G. Spitelle, Zur Chemie des
[27] J.S. Chung, N. Zmora, H. Katayama, N. Tsutsui, Crustacean hyperglycemic Ecdysons. VI: Reaktionen des Ecdysonmolekuls, Chem. Ber. 98 (1965)
hormone (CHH) neuropeptides family: functions, titer, and binding to target 2394–2402.
tissues, Gen. Comp. Endocrinol. 166 (2010) 447–454. [56] R. Keller, J.D. O’Connor, Neuroendocrine regulation of ecdysteroid production
[28] J. Colombani, L. Bianchini, S. Layalle, E. Pondeville, C. Dauphin-Villemant, C. in the crab Pachygrapsus crassipes, Gen. Comp. Endocrinol. 46 (1982) 384.
Antoniewski, C. Carre, S. Noselli, P. Leopold, Antagonistic actions of ecdysone [57] R. Keller, E. Schmid, In vitro secretion of ecdysteroids by Y-organs and lack of
and insulins determine final size in Drosophila, Science 310 (2005) secretion by mandibular organs of the crayfish following molt induction, J.
667–670. Comp. Physiol. 130 (1979) 347–353.
[29] J. Covi, A. Gomez, S. Chang, K. Lee, E. Chang, D. Mykles, Repression of Y-organ [58] H.W. Kim, L.A. Batista, J.L. Hoppes, K.J. Lee, D.L. Mykles, A crustacean nitric
ecdysteroidogenesis by cyclic nucleotides and agonists of NO-sensitive oxide synthase expressed in nerve ganglia, Y-organ, gill and gonad of the
guanylyl cyclase, in: S. Morris, A. Vosloo (Eds.), 4th Meeting of Comparative tropical land crab, Gecarcinus lateralis, J. Exp. Biol. 207 (2004) 2845–2857.
Physiologists & Biochemists in Africa – Mara 2008 – ‘‘Molecules to Migration: [59] H.W. Kim, E.S. Chang, D.L. Mykles, Three calpains and ecdysone receptor in
The Pressures of Life’’, Monduzzi Editore International, Bologna, Italy, 2008, the land crab, Gecarcinus lateralis: sequences, expression, and effects of
pp. 37–46. elevated ecdysteroid induced by eyestalk ablation, J. Exp. Biol. 208 (2005)
[30] J.A. Covi, B.D. Bader, E.S. Chang, D.L. Mykles, Molt cycle regulation of protein 3177–3197.
synthesis in skeletal muscle of the blackback land crab, Gecarcinus lateralis, [60] H.W. Kim, S.G. Lee, D.L. Mykles, Ecdysteroid-responsive genes, RXR and E75,
and the differential expression of a myostatin-like factor during atrophy in the tropical land crab, Gecarcinus lateralis: differential tissue expression of
induced by molting or unweighting, J. Exp. Biol. 213 (2010) 172–183. multiple RXR isoforms generated at three alternative splicing sites in the
[31] J.A. Covi, E.S. Chang, D.L. Mykles, Conserved role of cyclic nucleotides in the hinge and ligand-binding domains, Mol. Cell. Endocrinol. 242 (2005) 80–95.
regulation of ecdysteroidogenesis by the crustacean molting gland, Comp. [61] K.S. Kim, J.M. Jeon, H.-W. Kim, A myostatin-like gene expressed highly in the
Biochem. Physiol. A 152 (2009) 470–477. muscle tissue of Chinese mitten crab, Eriocheir sinensis, Fish. Aqua. Sci. 12
[32] J.A. Covi, E.S. Chang, D.L. Mykles, Neuropeptide signaling mechanisms in (2009) 185–193.
crustacean and insect molting glands, Invert. Reprod. Dev., in press. [62] K.S. Kim, Y.J. Kim, J.M. Jeon, Y.S. Kang, C.W. Oh, H.W. Kim, Molecular
[33] J.A. Covi, H.W. Kim, D.L. Mykles, Expression of alternatively spliced characterization of myostatin-like genes expressed highly in the muscle
transcripts for a myostatin-like protein in the blackback land crab, tissue from Morotoge shrimp, Pandalopsis japonica, Aquacult. Res. 41 (2010)
Gecarcinus lateralis, Comp. Biochem. Physiol. A 150 (2008) 423–430. e862–e871.
[34] D.P.V. De Kleijn, F. Van Herp, Molecular biology of neurohormone precursors [63] B.C. Kone, Molecular biology of natriuretic peptides and nitric oxide
in the eyestalk of Crustacea, Comp. Biochem. Physiol. B 112 (1995) 573– synthases, Cardiovasc. Res. 51 (2001) 429–441.
579. [64] F. Lachaise, G. Carpentier, G. Somme, J. Colardeau, P. Beydon, Ecdysteroid
[35] S. Dell, D. Sedlmeier, D. Böcking, C. Dauphin-Villemant, Ecdysteroid synthesis by crab Y-organs, J. Exp. Zool. 252 (1989) 283–292.
biosynthesis in crayfish Y-organs: feedback regulation by circulating [65] S. Layalle, N. Arquier, P. Leopold, The TOR pathway couples nutrition and
ecdysteroids, Arch. Insect Biochem. Physiol. 41 (1999) 148–155. developmental timing in Drosophila, Dev. Cell 15 (2008) 568–577.
[36] G. Echalier, L’organe Y et le déterminisme de la croissance et de la mue chez [66] K.J. Lee, R.M. Doran, D.L. Mykles, Crustacean hyperglycemic hormone from
Carcinus maenas (L.), Crustacé Décapode, Ann. Sci. Nat. Zool. 12 (1959) 1–59. the tropical land crab, Gecarcinus lateralis: cloning, isoforms, and tissue
[37] P. Edomi, E. Azzoni, R. Mettulio, N. Pandolfelli, E.A. Ferrero, P.G. Giulianini, expression, Gen. Comp. Endocrinol. 154 (2007) 174–183.
Gonad-inhibiting hormone of the Norway lobster (Nephrops norvegicus): [67] K.J. Lee, H.W. Kim, A.M. Gomez, E.S. Chang, J.A. Covi, D.L. Mykles, Molt-
cDNA cloning, expression, recombinant protein production, and inhibiting hormone from the tropical land crab, Gecarcinus lateralis: cloning,
immunolocalization, Gene 284 (2002) 93–102. tissue expression, and expression of biologically active recombinant peptide
[38] A.J. El Haj, P. Harrison, E.S. Chang, Localization of ecdysteroid receptor in yeast, Gen. Comp. Endocrinol. 150 (2007) 505–513.
immunoreactivity in eyestalk and muscle tissue of the American lobster, [68] S.G. Lee, B.D. Bader, E.S. Chang, D.L. Mykles, Effects of elevated ecdysteroid on
Homarus americanus, J. Exp. Zool. 270 (1994) 343–349. tissue expression of three guanylyl cyclases in the tropical land crab
[39] M.L. Fanjul-Moles, Biochemical and functional aspects of crustacean Gecarcinus lateralis: possible roles of neuropeptide signaling in the molting
hyperglycemic hormone in decapod crustaceans: review and update, Comp. gland, J. Exp. Biol. 210 (2007) 3245–3254.
Biochem. Physiol. C 142 (2006) 390–400. [69] S.G. Lee, H.W. Kim, D.L. Mykles, Guanylyl cyclases in the tropical land crab,
[40] M. Gabe, Sur l’existence, chez quelques Crustacés Malacostacés, d’un organe Gecarcinus lateralis: cloning of soluble (NO-sensitive and -insensitive) and
comparable à la glande de la mue des Insectes, CR Hebd. Seances Acad. Sci. membrane receptor forms, Comp. Biochem. Physiol. 2D (2007) 332–344.
237 (1953) 1111–1113. [70] S.G. Lee, D.L. Mykles, Proteomics and signal transduction in the crustacean
[41] M. Gersch, K. Richter, G.-A. Böhm, H. Eibisch, Experimentelle Untersuchungen molting gland, Integr. Comp. Biol. 46 (2006) 965–977.
zur saisonalen Abhangigkeit der neurohormonalen Steuerung der [71] T. Liu, X.H. Feng, Regulation of TGF-beta signalling by protein phosphatases,
Gastrolithenbildung und der Ecdysteroidsynthese des Y-organs von Biochem. J. 430 (2010) 191–198.
Orconectes limosus (Crustacea, Decapoda), Zool. Jahr. Abt. Allgem. Zool. [72] K.S. MacLea, J.A. Covi, H.W. Kim, E. Chao, S. Medler, E.S. Chang, D.L. Mykles,
Physiol. Tiere 84 (1980) 164–180. Myostatin from the American lobster, Homarus americanus: cloning and
[42] M.W. Gilgan, The inhibition of normal molting in the adult male lobster effects of molting on expression in skeletal muscles, Comp. Biochem. Physiol.
(Homarus americanus) by ecdysterone treatment, Comp. Biochem. Physiol. A A 157 (2010) 328–337.
65 (1980) 207–209. [73] L.H. Mantel, Dorothy E. Bliss (1916–1987), J. Crustac. Biol. 8 (1987) 706–709.
[43] M.F. Goy, Activation of membrane guanylate cyclase by an invertebrate [74] M.P. Mattson, E. Spaziani, Characterization of molt-inhibiting hormone (MIH)
peptide hormone, J. Biol. Chem. 265 (1990) 20220–20227. action on crustacean Y-organ segments and dispersed cells in culture and a
[44] P.L. Gu, K.H. Chu, S.M. Chan, Bacterial expression of the shrimp molt- bioassay for MIH activity, J. Exp. Zool. 236 (1985) 93–101.
inhibiting hormone (MIH): antibody production, immunocytochemical study [75] M.P. Mattson, E. Spaziani, Regulation of Y-organ ecdysteroidogenesis by molt-
and biological assay, Cell Tissue Res. 303 (2001) 129–136. inhibiting hormone in crabs: involvement of cyclic AMP-mediated protein
[45] S.H. Gu, J.L. Lin, P.L. Lin, C.H. Chen, Insulin stimulates ecdysteroidogenesis by synthesis, Gen. Comp. Endocrinol. 63 (1986) 414–423.
prothoracic glands in the silkworm, Bombyx mori, Insect Biochem. Mol. Biol. [76] A.A. McDonald, E.S. Chang, D.L. Mykles, Cloning of a nitric oxide synthase
39 (2009) 171–179. from green shore crab, Carcinus maenas: a comparative study of the effects of
[46] F. Hampshire, D.H.S. Horn, Structure of crustecdysone, a crustacean moulting eyestalk ablation on expression in the molting glands (Y-organs) of C. maenas,
hormone, Chem. Commun. 4567 (1966) 37–38. and blackback land crab, Gecarcinus lateralis, Comp. Biochem. Physiol. A 158
[47] B. Hanström, Hormones in Invertebrates, Oxford University Press, London, (2011) 150–162.
1939. [77] C. Mirth, J.W. Truman, L.M. Riddiford, The role of the prothoracic gland in
[48] R.G. Hartnoll, Growth in Crustacea – twenty years on, Hydrobiologia 449 determining critical weight to metamorphosis in Drosophila melanogaster,
(2001) 111–122. Curr. Biol. 15 (2005) 1796–1807.
[49] C.A. Holland, D.M. Skinner, Interactions between molting and regeneration in [78] H. Mohamedeen, R.G. Hartnoll, Larval and postlarval growth of individually
the land crab, Biol. Bull. 150 (1976) 222–240. reared specimens of common shore crab Carcinus maenas (L.), J. Exp. Mar.
[50] P.M. Hopkins, Effects of neurosecretory factors on in vivo levels of Biol. Ecol. 134 (1989) 1–24.
ecdysteroids in hemolymph of the fiddler crab, Uca pugilator, Am. Zool. 22 [79] A. Moustakas, C.H. Heldin, The regulation of TGF beta signal transduction,
(1982) 938. Development 136 (2009) 3699–3714.
[51] P.M. Hopkins, Limb regeneration in the fiddler crab, Uca pugilator: hormonal [80] D.L. Mykles, Getting out of a tight squeeze – enzymatic regulation of claw
and growth factor control, Am. Zool. 41 (2001) 389–398. muscle atrophy in molting, Am. Zool. 32 (1992) 485–494.
[52] D.H.S. Horn, E.J. Middleton, J.A. Wunderlich, F. Hampshire, Identity of the [81] D.L. Mykles, Crustacean muscle plasticity: molecular mechanisms
moulting hormones of insects and crustaceans, Chem. Commun. (1966) determining mass and contractile properties, Comp. Biochem. Physiol. B
339–340. 117 (1997) 367–378.
330 E.S. Chang, D.L. Mykles / General and Comparative Endocrinology 172 (2011) 323–330

[82] D.L. Mykles, Interactions between limb regeneration and molting in decapod [108] M.J. Snyder, E.S. Chang, Ecdysteroids in relation to the molt cycle of the
crustaceans, Am. Zool. 41 (2001) 399–406. American lobster, Homarus americanus. II: Excretion of metabolites, Gen.
[83] D.L. Mykles, Ecdysteroid metabolism in crustaceans, J. Steroid Biochem. Mol. Comp. Endocrinol. 83 (1991) 118–131.
Biol., in press. [109] M.J. Snyder, E.S. Chang, Metabolism and excretion of injected [3H]-ecdysone
[84] D.L. Mykles, M.E. Adams, G. Gade, A.B. Lange, H.G. Marco, I. Orchard, by female lobsters, Homarus americanus, Biol. Bull. 180 (1991) 475–484.
Neuropeptide action in insects and crustaceans, Physiol. Biochem. Zool. 83 [110] M.J. Snyder, E.S. Chang, Role of the midgut gland in metabolism and excretion
(2010) 836–846. of ecdysteroids by lobsters, Homarus americanus, Gen. Comp. Endocrinol. 85
[85] D.L. Mykles, C.K. Ghalambor, J.H. Stillman, L. Tomanek, Grand challenges in (1992) 286–296.
comparative physiology: integration across disciplines and across levels of [111] C. Soumoff, J.D. O’Connor, Repression of Y-organ secretory activity by molt-
biological organization, Integr. Comp. Biol. 50 (2010) 6–16. inhibiting hormone in the crab Pachygrapsus crassipes, Gen. Comp.
[86] D.L. Mykles, L.H. Mantel, Obituary – Dorothy M. Skinner, J. Crustac. Biol. 25 Endocrinol. 48 (1982) 432–439.
(2005) 529–532. [112] E. Spaziani, K. DeSantis, B.D. O’Rourke, W.L. Wang, J.D. Weld, The clearance
[87] T. Nakatsuji, C.Y. Lee, R.D. Watson, Crustacean molt-inhibiting hormone: in vivo and metabolism of ecdysone and 3-dehydroecdysone in tissues of the
structure, function, and cellular mode of action, Comp. Biochem. Physiol. crab Cancer antennarius, J. Exp. Zool. 279 (1997) 609–619.
152A (2009) 139–148. [113] E. Spaziani, T.C. Jegla, W.L. Wang, J.A. Booth, S.M. Connolly, C.C. Conrad, M.J.
[88] T. Nakatsuji, H. Sonobe, Regulation of ecdysteroid secretion from the Y-organ Dewall, C.M. Sarno, D.K. Stone, R. Montgomery, Further studies on signaling
by molt-inhibiting hormone in the American crayfish, Procambarus clarkii, pathways for ecdysteroidogenesis in crustacean Y-organs, Am. Zool. 41
Gen. Comp. Endocrinol. 135 (2004) 358–364. (2001) 418–429.
[89] T. Nakatsuji, H. Sonobe, R.D. Watson, Molt-inhibiting hormone-mediated [114] E. Spaziani, H.H. Rees, W.L. Wang, R.D. Watson, Evidence that Y-organs of the
regulation of ecdysteroid synthesis in Y-organs of the crayfish (Procambarus crab Cancer antennarius secrete 3-dehydroecdysone, Mol. Cell. Endocrinol. 66
clarkii): involvement of cyclic GMP and cycle nucleotide phosphodiesterase, (1989) 17–25.
Mol. Cell. Endocrinol. 253 (2006) 76–82. [115] D.P. Srivastava, E.J. Yu, K. Kennedy, H. Chatwin, V. Reale, M. Hamon, T. Smith,
[90] S. Nimitkul, D.L. Mykles, E.S. Chang, Feedback regulation of ecdysteroid P.D. Evans, Rapid, nongenomic responses to ecdysteroids and catecholamines
analog on Y-organs of the green crab Carcinus maenas, Integr. Comp. Biol. 50 mediated by a novel Drosophila G-protein-coupled receptor, J. Neurosci. 25
(2010) e274. (2005) 6145–6155.
[91] T. Okumura, T. Ohira, H. Katayama, H. Nagasawa, In vivo effects of a [116] A.A. Teleman, Molecular mechanisms of metabolic regulation by insulin in
recombinant molt-inhibiting hormone on molt interval and hemolymph Drosophila, Biochem. J. 425 (2010) 13–26.
ecdysteroid level in the kuruma prawn, Marsupenaeus japonicus, Zool. Sci. 22 [117] K.H. Tomaschko, Nongenomic effects of ecdysteroids, Arch. Insect Biochem.
(2005) 317–320. Physiol. 41 (1999) 89–98.
[92] L.M. Passano, Neurosecretory control of molting in crabs by the X-organ sinus [118] K. Tsuchida, M. Nakatani, K. Hitachi, A. Uezumi, Y. Sunada, H. Ageta, K.
gland complex, Physiol. Comp. Oecol. 3 (1953) 155–189. Inokuchi, Activin signaling as an emerging target for therapeutic
[93] L.M. Passano, Low temperature blockage of molting in Uca pugnax, Biol. Bull. interventions, Cell Commun. Signal. 7 (2009) 23 (Article No. 15).
118 (1960) 129–136. [119] U. Von Gliscynski, D. Sedlmeier, Regulation of ecdysteroid biosynthesis in
[94] C.G. Proud, MTORC1 signalling and mRNA translation, Biochem. Soc. Trans. 37 crayfish Y-organs: II. Role of cyclic nucleotide-dependent protein kinases, J.
(2009) 227–231. Exp. Zool. 265 (1993) 454–458.
[95] K.W. Regelson, J.S. Covi, S.A. Chang, E.S. Chang, D.L. Mykles, Studies on the [120] R.D. Watson, K.J. Lee, S. Qiu, M. Luo, H.R. Umphrey, R.D. Roer, E. Spaziani,
signaling pathways regulating facultative and constitutive Molecular cloning, expression, and tissue distribution of crustacean molt-
ecdysteroidogenesis in the crustacean molting gland, Integr. Comp. Biol. 50 inhibiting hormone, Am. Zool. 41 (2001) 407–417.
(2010) e286. [121] R.D. Watson, E. Spaziani, W.E. Bollenbacher, Regulation of ecdysone
[96] R.D. Roer, R.M. Dillaman, Molt-related change in integumental structure and biosynthesis in insects and crustaceans, in: J. Koolman (Ed.), Ecdysone,
function, in: M.N. Horst, J.A. Freeman (Eds.), The Crustacean Integument from Chemistry to Mode of Action, Georg Thieme Verlag, Stuttgart, 1989, pp.
Morphology and Biochemistry, CRC Press, Boca Raton, 1993, pp. 1–37. 188–203.
[97] B. Saïdi, N. De Bessé, S.G. Webster, D. Sedlmeier, F. Lachaise, Involvement of [122] S.G. Webster, Neurohormonal control of ecdysteroid biosynthesis by Carcinus
cAMP and cGMP in the mode of action of molt-inhibiting hormone (MIH) a maenas Y-organs in vitro and preliminary characterization of the putative
neuropeptide which inhibits steroidogenesis in a crab, Mol. Cell. Endocrinol. molt-inhibiting hormone, Gen. Comp. Endocrinol. 61 (1986) 237–247.
102 (1994) 53–61. [123] S.G. Webster, Amino acid sequence of putative moult-inhibiting hormone
[98] U. Schlattner, X. Vafopoulou, C.G.H. Steel, R.E. Hormann, M. Lezzi, Non- from the crab Carcinus maenas, Proc. R. Soc. Lond. [Biol.] 244 (1991) 247–252.
genomic ecdysone effects and the invertebrate nuclear steroid hormone [124] S.G. Webster, High-affinity binding of putative moult-inhibiting hormone
receptor EcR – new role for an ‘‘old’’ receptor? Mol. Cell. Endocrinol. 247 (MIH) and crustacean hyperglycaemic hormone (CHH) to membrane-bound
(2006) 64–72. receptors on the Y-organ of the shore crab Carcinus maenus, Proc. R. Soc. Lond.
[99] P.J. Schoettker, D.H. Gist, In vitro ecdysteroid production by Y-organs of the [Biol.] 251 (1993) 53–59.
blue crab Callinectes sapidus, J. Crustac. Biol. 10 (1990) 487–491. [125] S.G. Webster, Neuropeptides inhibiting growth and reproduction in
[100] D. Sedlmeier, R. Fenrich, Regulation of ecdysteroid biosynthesis in crayfish Y- crustaceans, in: G.M. Coast, S.G. Webster (Eds.), Recent Advances in
organs. I: Role of cyclic nucleotides, J. Exp. Zool. 265 (1993) 448–453. Arthropod Endocrinology, Cambridge University Press, Cambridge, 1998,
[101] M. Sefiani, J.P. Le Caer, D. Soyez, Characterization of hyperglycemic and molt- pp. 33–52.
inhibiting activity from sinus glands of the penaeid shrimp Penaeus vannamei, [126] M.G. Wheatly, Calcium homeostasis in Crustacea: the evolving role of
Gen. Comp. Endocrinol. 103 (1996) 41–53. branchial, renal, digestive and hypodermal epithelia, J. Exp. Zool. 283 (1999)
[102] D.M. Skinner, The structure and metabolism of a crustacean integumentary 620–640.
tissue during a molt cycle, Biol. Bull. 123 (1962) 635–647. [127] D.R. Williams, J.H. Chen, M.J. Fisher, H.H. Rees, Induction of enzymes involved
[103] D.M. Skinner, Breakdown and reformation of somatic muscle during the molt in molting hormone (ecdysteroid) inactivation by ecdysteroids and an
cycle of the land crab, Gecarcinus lateralis, J. Exp. Zool. 163 (1966) agonist, 1, 2-dibenzoyl-1-tert-butylhydrazine (RH-5849), J. Biol. Chem. 272
115–124. (1997) 8427–8432.
[104] D.M. Skinner, Interacting factors in the control of the crustacean molt cycle, [128] X.L. Yu, E.S. Chang, D.L. Mykles, Characterization of limb autotomy factor-
Am. Zool. 25 (1985) 275–284. proecdysis (LAFpro), isolated from limb regenerates, that suspends molting in
[105] D.M. Skinner, Molting and regeneration, in: D.E. Bliss, L.H. Mantel (Eds.), The the land crab Gecarcinus lateralis, Biol. Bull. 202 (2002) 204–212.
Biology of Crustacea, Academic Press, New York, 1985, pp. 43–146. [129] T.P. Zarubin, E.S. Chang, D.L. Mykles, Expression of recombinant eyestalk
[106] D.M. Skinner, D.E. Graham, Loss of limbs as a stimulus to ecdysis in Brachyura crustacean hyperglycemic hormone from the tropical land crab, Gecarcinus
(true crabs), Biol. Bull. 143 (1972) 222–233. lateralis, that inhibits Y-organ ecdysteroidogenesis in vitro, Mol. Biol. Rep. 36
[107] R.I. Smith, Studies on the effects of eyestalk removal upon young crayfish (2009) 1231–1237.
(Cambarus clarkii Girard), Biol. Bull. 79 (1940) 145–152. [130] C. Zeleny, Compensatory regulation, J. Exp. Zool. 2 (1905) 1–102.

You might also like